1
|
Rodarte J, Baehr C, Hicks D, McGovern M, Zhang Y, Silva-Ortiz P, Hannon B, Duddu S, Pancera M, Pravetoni M. Structure-Based Engineering of Monoclonal Antibodies for Improved Binding to Counteract the Effects of Fentanyl and Carfentanil. ACS OMEGA 2024; 9:42506-42519. [PMID: 39431098 PMCID: PMC11483391 DOI: 10.1021/acsomega.4c06617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/22/2024] [Accepted: 09/23/2024] [Indexed: 10/22/2024]
Abstract
The opioid overdose epidemic is a growing and evolving public health crisis fueled by the widespread presence of fentanyl and fentanyl analogues (F/FAs) in both street mixtures and counterfeit pills. To expand current treatment options, drug-targeting monoclonal antibodies (mAbs) offer a viable therapeutic for both pre- and postexposure clinical scenarios. This study reports the isolation, in vitro characterization, and in vivo efficacy of two murine mAb families targeting fentanyl, carfentanil, or both. Because humanization of the mAbs by CDR grafting negatively impacted affinity for both fentanyl and carfentanil, crystal structures of mAbs in complex with fentanyl or carfentanil were analyzed to identify key residues involved in ligand binding in murine versus humanized structures, and site-directed mutagenesis was used to verify their functional importance. The structural analysis identified a framework residue, Tyr36, present in the murine germline sequence of two mAbs, which was critical for binding to fentanyl and carfentanil. These studies emphasize the importance of structural considerations in mAb engineering to optimize mAbs targeting small molecules including opioids and other drugs of public health interest.
Collapse
Affiliation(s)
- Justas Rodarte
- Vaccine
and Infectious Disease Division, Fred Hutchinson
Cancer Center, Seattle, Washington 98109, United States
| | - Carly Baehr
- Department
of Pharmacology, University of Minnesota
Medical School, Minneapolis, Minnesota 55455, United States
| | - Dustin Hicks
- Department
of Pharmacology, University of Minnesota
Medical School, Minneapolis, Minnesota 55455, United States
| | - Morgan McGovern
- Vaccine
and Infectious Disease Division, Fred Hutchinson
Cancer Center, Seattle, Washington 98109, United States
| | - Yue Zhang
- Department
of Pharmacology, University of Minnesota
Medical School, Minneapolis, Minnesota 55455, United States
- Department
of Psychiatry and Behavioral Sciences, School of Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Pedro Silva-Ortiz
- Department
of Pharmacology, University of Minnesota
Medical School, Minneapolis, Minnesota 55455, United States
| | - Bryan Hannon
- Vaccine
and Infectious Disease Division, Fred Hutchinson
Cancer Center, Seattle, Washington 98109, United States
- Department
of Psychiatry and Behavioral Sciences, School of Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Sowmya Duddu
- Vaccine
and Infectious Disease Division, Fred Hutchinson
Cancer Center, Seattle, Washington 98109, United States
| | - Marie Pancera
- Vaccine
and Infectious Disease Division, Fred Hutchinson
Cancer Center, Seattle, Washington 98109, United States
| | - Marco Pravetoni
- Department
of Psychiatry and Behavioral Sciences, School of Medicine, University of Washington, Seattle, Washington 98195, United States
- Center
for Medication Development for Substance Use Disorders and Overdose, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
2
|
Khaimraj A, Baehr CA, Hicks D, Raleigh MD, Pravetoni M. Monoclonal Antibodies Engineered with Fc Region Mutations to Extend Protection against Fentanyl Toxicity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:663-668. [PMID: 39018496 PMCID: PMC11333160 DOI: 10.4049/jimmunol.2400170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/19/2024] [Indexed: 07/19/2024]
Abstract
Fentanyl and other synthetic opioids are the leading cause of drug-related deaths in the United States. mAbs that selectively target fentanyl and fentanyl analogues offer a promising strategy for treating both opioid-related overdoses and opioid use disorders. To increase the duration of efficacy of a candidate mAb against fentanyl, we selected three sets of mutations in the Fc region of an IgG1 anti-fentanyl mAb (HY6-F9DF215, HY6-F9DHS, HY6-F9YTE) to increase binding to the neonatal Fc receptor (FcRn). The mAb mutants were compared against unmodified (wild-type [WT], HY6-F9WT) anti-fentanyl mAb for fentanyl binding, thermal stability, and FcRn affinity in vitro, and for efficacy against fentanyl and mAb half-life in vivo in mice. Biolayer interferometry showed a >10-fold increase in the affinity for recombinant FcRn of the three mutant mAbs compared with HY6-F9WT. During an acute fentanyl challenge in mice, all FcRn-mutated mAbs provided equal protection against fentanyl-induced effects, and all mAbs reduced brain fentanyl levels compared with the saline group. Serum persistence of the mutant mAbs was tested in Tg276 transgenic mice expressing human FcRn. After administration of 40 mg/kg HY6-F9WT, HY6-F9DF215, HY6-F9DHS, and HY6-F9YTE, the mAbs showed half-lives of 6.3, 26.4, 14.7, and 6.9 d, respectively. These data suggest that modification of mAbs against fentanyl to bind to FcRn with higher affinity can increase their half-life relative to WT mAbs while maintaining efficacy against the toxic effects of fentanyl, further supporting their potential role as a therapeutic treatment option for opioid use disorder and overdose.
Collapse
Affiliation(s)
- Aaron Khaimraj
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Carly A. Baehr
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Dustin Hicks
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Michael D. Raleigh
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Marco Pravetoni
- Department of Psychiatry and Behavioral Sciences, University of Washington, University of Washington Center for Medication Development for Substance Use Disorders, Garvey Institute for Brain Solutions, Seattle, WA
| |
Collapse
|
3
|
Gallant JP, Hicks D, Shi K, Moeller NH, Hoppe B, Lake EW, Baehr C, Pravetoni M, Aihara H, LeBeau AM. Identification and biophysical characterization of a novel domain-swapped camelid antibody specific for fentanyl. J Biol Chem 2024; 300:107502. [PMID: 38945452 PMCID: PMC11321312 DOI: 10.1016/j.jbc.2024.107502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/04/2024] [Accepted: 06/17/2024] [Indexed: 07/02/2024] Open
Abstract
Opioid use disorders (OUD) and overdoses are ever-evolving public health threats that continue to grow in incidence and prevalence in the United States and abroad. Current treatments consist of opioid receptor agonists and antagonists, which are safe and effective but still suffer from some limitations. Murine and humanized monoclonal antibodies (mAb) have emerged as an alternative and complementary strategy to reverse and prevent opioid-induced respiratory depression. To explore antibody applications beyond traditional heavy-light chain mAbs, we identified and biophysically characterized a novel single-domain antibody specific for fentanyl from a camelid variable-heavy-heavy (VHH) domain phage display library. Structural data suggested that VHH binding to fentanyl was facilitated by a unique domain-swapped dimerization mechanism, which accompanied a rearrangement of complementarity-determining region loops leading to the formation of a fentanyl-binding pocket. Structure-guided mutagenesis further identified an amino acid substitution that improved the affinity and relaxed the requirement for dimerization of the VHH in fentanyl binding. Our studies demonstrate VHH engagement of an opioid and inform on how to further engineer a VHH for enhanced stability and efficacy, laying the groundwork for exploring the in vivo applications of VHH-based biologics against OUD and overdose.
Collapse
Affiliation(s)
- Joseph P Gallant
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Dustin Hicks
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Ke Shi
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Nicholas H Moeller
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Brooke Hoppe
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington, USA
| | - Eric W Lake
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Carly Baehr
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Marco Pravetoni
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington, USA; Center for Medication Development for Substance Use Disorders, University of Washington, Seattle, Washington, USA.
| | - Hideki Aihara
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA.
| | - Aaron M LeBeau
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA; Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA; Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA.
| |
Collapse
|
4
|
Shao W, Sorescu DC, Liu Z, Star A. Machine Learning Discrimination and Ultrasensitive Detection of Fentanyl Using Gold Nanoparticle-Decorated Carbon Nanotube-Based Field-Effect Transistor Sensors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2311835. [PMID: 38679787 DOI: 10.1002/smll.202311835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/12/2024] [Indexed: 05/01/2024]
Abstract
The opioid overdose crisis is a global health challenge. Fentanyl, an exceedingly potent synthetic opioid, has emerged as a leading contributor to the surge in opioid-related overdose deaths. The surge in overdose fatalities, particularly due to illicitly manufactured fentanyl and its contamination of street drugs, emphasizes the urgency for drug-testing technologies that can quickly and accurately identify fentanyl from other drugs and quantify trace amounts of fentanyl. In this paper, gold nanoparticle (AuNP)-decorated single-walled carbon nanotube (SWCNT)-based field-effect transistors (FETs) are utilized for machine learning-assisted identification of fentanyl from codeine, hydrocodone, and morphine. The unique sensing performance of fentanyl led to use machine learning approaches for accurate identification of fentanyl. Employing linear discriminant analysis (LDA) with a leave-one-out cross-validation approach, a validation accuracy of 91.2% is achieved. Meanwhile, density functional theory (DFT) calculations reveal the factors that contributed to the enhanced sensitivity of the Au-SWCNT FET sensor toward fentanyl as well as the underlying sensing mechanism. Finally, fentanyl antibodies are introduced to the Au-SWCNT FET sensor as specific receptors, expanding the linear range of the sensor in the lower concentration range, and enabling ultrasensitive detection of fentanyl with a limit of detection at 10.8 fg mL-1.
Collapse
Affiliation(s)
- Wenting Shao
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260, USA
| | - Dan C Sorescu
- United States Department of Energy, National Energy Technology Laboratory, Pittsburgh, Pennsylvania, 15236, USA
- Department of Chemical & Petroleum Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA
| | - Zhengru Liu
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260, USA
| | - Alexander Star
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA
| |
Collapse
|
5
|
Chen XY, Wang L, Ma X, Yang F, Wang X, Xu P, Xu LL, Di B. Development of fentanyl-specific monoclonal antibody (mAb) to antagonize the pharmacological effects of fentanyl. Toxicol Appl Pharmacol 2024; 486:116918. [PMID: 38570042 DOI: 10.1016/j.taap.2024.116918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 03/17/2024] [Accepted: 03/30/2024] [Indexed: 04/05/2024]
Abstract
Fentanyl, a critical component of opioid analgesics, poses a severe threat to public health, exacerbating the drug problem due to its potential fatality. Herein, we present two novel haptens designed with different attachment sites conjugated to keyhole limpet hemocyanin (KLH), aiming to develop an efficacious vaccine against fentanyl. KLH-Fent-1 demonstrated superior performance over KLH-Fent-2 in antibody titer, blood-brain distribution, and antinociceptive tests. Consequently, we immunized mice with KLH-Fent-1 to generate fentanyl-specific monoclonal antibodies (mAbs) using the hybridoma technique to compensate for the defects of active immunization in the treatment of opioid overdose and addiction. The mAb produced by hybridoma 9D5 exhibited the ability to recognize fentanyl and its analogs with a binding affinity of 10-10 M. Subsequently, we developed a human IgG1 chimeric mAb to improve the degree of humanization. Pre-treatment with murine and chimeric mAb significantly reduced the analgesic effect of fentanyl and altered its blood-brain biodistribution in vivo. Furthermore, in a mouse model of fentanyl-induced respiratory depression, the chimeric mAb effectively reversed respiratory depression promptly and maintained a certain level during the week. The development of high-affinity chimeric mAb gives support to combat the challenges of fentanyl misuse and its detrimental consequences. In conclusion, mAb passive immunization represents a viable strategy for addressing fentanyl addiction and overdose.
Collapse
Affiliation(s)
- Xiao-Yi Chen
- Office of China National Narcotics Control Commission, China Pharmaceutical University Joint Laboratory on Key Technologies of Narcotics Control, Nanjing 210009, China; Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
| | - Li Wang
- Office of China National Narcotics Control Commission, China Pharmaceutical University Joint Laboratory on Key Technologies of Narcotics Control, Nanjing 210009, China; Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao Ma
- Office of China National Narcotics Control Commission, China Pharmaceutical University Joint Laboratory on Key Technologies of Narcotics Control, Nanjing 210009, China; Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
| | - Fan Yang
- Office of China National Narcotics Control Commission, China Pharmaceutical University Joint Laboratory on Key Technologies of Narcotics Control, Nanjing 210009, China; Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao Wang
- Office of China National Narcotics Control Commission, China Pharmaceutical University Joint Laboratory on Key Technologies of Narcotics Control, Nanjing 210009, China; Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
| | - Peng Xu
- Office of China National Narcotics Control Commission, China Pharmaceutical University Joint Laboratory on Key Technologies of Narcotics Control, Nanjing 210009, China; Key Laboratory of Drug Monitoring and Control, Drug Intelligence and Forensic Center, Ministry of Public Security, Beijing 100193, China.
| | - Li-Li Xu
- Office of China National Narcotics Control Commission, China Pharmaceutical University Joint Laboratory on Key Technologies of Narcotics Control, Nanjing 210009, China; Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.
| | - Bin Di
- Office of China National Narcotics Control Commission, China Pharmaceutical University Joint Laboratory on Key Technologies of Narcotics Control, Nanjing 210009, China; Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
6
|
Lu B, Wei L, Shi G, Du J. Nanotherapeutics for Alleviating Anesthesia-Associated Complications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308241. [PMID: 38342603 PMCID: PMC11022745 DOI: 10.1002/advs.202308241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/22/2023] [Indexed: 02/13/2024]
Abstract
Current management of anesthesia-associated complications falls short in terms of both efficacy and safety. Nanomaterials with versatile properties and unique nano-bio interactions hold substantial promise as therapeutics for addressing these complications. This review conducts a thorough examination of the existing nanotherapeutics and highlights the strategies for developing prospective nanomedicines to mitigate anesthetics-related toxicity. Initially, general, regional, and local anesthesia along with the commonly used anesthetics and related prevalent side effects are introduced. Furthermore, employing nanotechnology to prevent and alleviate the complications of anesthetics is systematically demonstrated from three aspects, that is, developing 1) safe nano-formulization for anesthetics; 2) nano-antidotes to sequester overdosed anesthetics and alter their pharmacokinetics; 3) nanomedicines with pharmacodynamic activities to treat anesthetics toxicity. Finally, the prospects and challenges facing the clinical translation of nanotherapeutics for anesthesia-related complications are discussed. This work provides a comprehensive roadmap for developing effective nanotherapeutics to prevent and mitigate anesthesia-associated toxicity, which can potentially revolutionize the management of anesthesia complications.
Collapse
Affiliation(s)
- Bin Lu
- Department of AnesthesiologyThird Hospital of Shanxi Medical UniversityShanxi Bethune HospitalShanxi Academy of Medical SciencesTongji Shanxi HospitalTaiyuan030032China
- Key Laboratory of Cellular Physiology at Shanxi Medical UniversityMinistry of EducationTaiyuanShanxi Province030001China
| | - Ling Wei
- Shanxi Bethune Hospital Center Surgery DepartmentShanxi Academy of Medical SciencesTongji Shanxi HospitalThird Hospital of Shanxi Medical UniversityTaiyuan030032China
| | - Gaoxiang Shi
- Department of AnesthesiologyThird Hospital of Shanxi Medical UniversityShanxi Bethune HospitalShanxi Academy of Medical SciencesTongji Shanxi HospitalTaiyuan030032China
| | - Jiangfeng Du
- Key Laboratory of Cellular Physiology at Shanxi Medical UniversityMinistry of EducationTaiyuanShanxi Province030001China
- Department of Medical ImagingShanxi Key Laboratory of Intelligent Imaging and NanomedicineFirst Hospital of Shanxi Medical UniversityTaiyuanShanxi Province030001China
| |
Collapse
|
7
|
Bremer PT, Burke EL, Barrett AC, Desai RI. Investigation of monoclonal antibody CSX-1004 for fentanyl overdose. Nat Commun 2023; 14:7700. [PMID: 38052779 PMCID: PMC10698161 DOI: 10.1038/s41467-023-43126-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/01/2023] [Indexed: 12/07/2023] Open
Abstract
The opioid crisis in the United States is primarily driven by the highly potent synthetic opioid fentanyl leading to >70,000 overdose deaths annually; thus, new therapies for fentanyl overdose are urgently needed. Here, we present the first clinic-ready, fully human monoclonal antibody CSX-1004 with picomolar affinity for fentanyl and related analogs. In mice CSX-1004 reverses fentanyl antinociception and the intractable respiratory depression caused by the ultrapotent opioid carfentanil. Moreover, toxicokinetic evaluation in a repeat-dose rat study and human tissue cross-reactivity study reveals a favorable pharmacokinetic profile of CSX-1004 with no safety-related issues. Using a highly translational non-human primate (NHP) model of respiratory depression, we demonstrate CSX-1004-mediated protection from repeated fentanyl challenges for 3-4 weeks. Furthermore, treatment with CSX-1004 produces up to a 15-fold potency reduction of fentanyl in NHP respiration, antinociception and operant responding assays without affecting non-fentanyl opioids like oxycodone. Taken together, our data establish the feasibility of CSX-1004 as a promising candidate medication for preventing and reversing fentanyl-induced overdose.
Collapse
Affiliation(s)
| | - Emily L Burke
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Behavioral Biology Program, Integrative Neurochemistry Laboratory, McLean Hospital, Belmont, MA, USA
| | | | - Rajeev I Desai
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Behavioral Biology Program, Integrative Neurochemistry Laboratory, McLean Hospital, Belmont, MA, USA
| |
Collapse
|
8
|
Lin M, Eubanks LM, Karadkhelkar NM, Blake S, Janda KD. Catalytic Antibody Blunts Carfentanil-Induced Respiratory Depression. ACS Pharmacol Transl Sci 2023; 6:802-811. [PMID: 37200811 PMCID: PMC10186356 DOI: 10.1021/acsptsci.3c00031] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Indexed: 05/20/2023]
Abstract
Carfentanil, the most potent of the fentanyl analogues, is at the forefront of synthetic opioid-related deaths, second to fentanyl. Moreover, the administration of the opioid receptor antagonist naloxone has proven inadequate for an increasing number of opioid-related conditions, often requiring higher/additional doses to be effective, as such interest in alternative strategies to combat more potent synthetic opioids has intensified. Increasing drug metabolism would be one strategy to detoxify carfentanil; however, carfentanil's major metabolic pathways involve N-dealkylation or monohydroxylation, which do not lend themselves readily to exogenous enzyme addition. Herein, we report, to our knowledge, the first demonstration that carfentanil's methyl ester when hydrolyzed to its acid was found to be 40,000 times less potent than carfentanil in activating the μ-opioid receptor. Physiological consequences of carfentanil and its acid were also examined through plethysmography, and carfentanil's acid was found to be incapable of inducing respiratory depression. Based upon this information, a hapten was chemically synthesized and immunized, allowing the generation of antibodies that were screened for carfentanil ester hydrolysis. From the screening campaign, three antibodies were found to accelerate the hydrolysis of carfentanil's methyl ester. From this series of catalytic antibodies, the most active underwent extensive kinetic analysis, allowing us to postulate its mechanism of hydrolysis against this synthetic opioid. In the context of potential clinical applications, the antibody, when passively administered, was able to reduce respiratory depression induced by carfentanil. The data presented supports further development of antibody catalysis as a biologic strategy to complement carfentanil overdose reversal.
Collapse
Affiliation(s)
- Mingliang Lin
- Departments of Chemistry
and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute
of Research and Medicine (WIRM), The Scripps
Research Institute, La Jolla, California 92037, United States
| | - Lisa M. Eubanks
- Departments of Chemistry
and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute
of Research and Medicine (WIRM), The Scripps
Research Institute, La Jolla, California 92037, United States
| | - Nishant M. Karadkhelkar
- Departments of Chemistry
and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute
of Research and Medicine (WIRM), The Scripps
Research Institute, La Jolla, California 92037, United States
| | - Steven Blake
- Departments of Chemistry
and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute
of Research and Medicine (WIRM), The Scripps
Research Institute, La Jolla, California 92037, United States
| | - Kim D. Janda
- Departments of Chemistry
and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute
of Research and Medicine (WIRM), The Scripps
Research Institute, La Jolla, California 92037, United States
| |
Collapse
|
9
|
Chamoun K, Chevillard L, Hajj A, Callebert J, Mégarbane B. Mechanisms of Neurorespiratory Toxicity Induced by Fentanyl Analogs—Lessons from Animal Studies. Pharmaceuticals (Basel) 2023; 16:ph16030382. [PMID: 36986482 PMCID: PMC10051837 DOI: 10.3390/ph16030382] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 02/26/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
In 2020, fentanyl and its analogs contributed to ~65% of drug-attributed fatalities in the USA, with a threatening increasing trend during the last ten years. These synthetic opioids used as potent analgesics in human and veterinary medicine have been diverted to recreational aims, illegally produced and sold. Like all opioids, central nervous system depression resulting from overdose or misuse of fentanyl analogs is characterized clinically by the onset of consciousness impairment, pinpoint miosis and bradypnea. However, contrasting with what observed with most opioids, thoracic rigidity may occur rapidly with fentanyl analogs, contributing to increasing the risk of death in the absence of immediate life support. Various mechanisms have been proposed to explain this particularity associated with fentanyl analogs, including the activation of noradrenergic and glutamatergic coerulospinal neurons and dopaminergic basal ganglia neurons. Due to the high affinities to the mu-opioid receptor, the need for more elevated naloxone doses than usually required in morphine overdose to reverse the neurorespiratory depression induced by fentanyl analogs has been questioned. This review on the neurorespiratory toxicity of fentanyl and analogs highlights the need for specific research focused on these agents to better understand the involved mechanisms of toxicity and develop dedicated strategies to limit the resulting fatalities.
Collapse
Affiliation(s)
- Karam Chamoun
- Inserm, UMR-S1144, Paris Cité University, 75006 Paris, France
- Faculty of Pharmacy, Saint-Joseph University, Beirut 1100, Lebanon
- Laboratory of Pharmacology, Clinical Pharmacy, and Medicine Quality Control, Saint-Joseph University, Beirut 1100, Lebanon
| | | | - Aline Hajj
- Faculty of Pharmacy, Saint-Joseph University, Beirut 1100, Lebanon
- Laboratory of Pharmacology, Clinical Pharmacy, and Medicine Quality Control, Saint-Joseph University, Beirut 1100, Lebanon
- Research Center, Quebec University Hospital, Laval University, Québec, QC G1V 0A6, Canada
| | - Jacques Callebert
- Inserm, UMR-S1144, Paris Cité University, 75006 Paris, France
- Laboratory of Biochemistry and Molecular Biology, AP-HP, Lariboisière Hospital, 75010 Paris, France
| | - Bruno Mégarbane
- Inserm, UMR-S1144, Paris Cité University, 75006 Paris, France
- Department of Medical and Toxicological Critical Care, Lariboisière Hospital, Federation of Toxicology APHP, 75010 Paris, France
- Correspondence:
| |
Collapse
|
10
|
Martinez S, Harris H, Chao T, Luba R, Pravetoni M, Comer SD, Jones JD. The potential role of opioid vaccines and monoclonal antibodies in the opioid overdose crisis. Expert Opin Investig Drugs 2023; 32:181-185. [PMID: 36863002 PMCID: PMC10065938 DOI: 10.1080/13543784.2023.2187286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/01/2023] [Indexed: 03/04/2023]
Affiliation(s)
- Suky Martinez
- Division on Substance Use Disorders, Columbia University Irving Medical Center & New York State Psychiatric Institute, New York, NY, USA
| | - Hannah Harris
- Division on Substance Use Disorders, Columbia University Irving Medical Center & New York State Psychiatric Institute, New York, NY, USA
| | - Thomas Chao
- Behavioural Reward Affect + Impulsivity Neuroscience Lab, Faculty of Medicine, the University of British Columbia, Vancouver, Bc, Canada
| | - Rachel Luba
- Division on Substance Use Disorders, Columbia University Irving Medical Center & New York State Psychiatric Institute, New York, NY, USA
| | - Marco Pravetoni
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Sandra D Comer
- Division on Substance Use Disorders, Columbia University Irving Medical Center & New York State Psychiatric Institute, New York, NY, USA
| | - Jermaine D Jones
- Division on Substance Use Disorders, Columbia University Irving Medical Center & New York State Psychiatric Institute, New York, NY, USA
| |
Collapse
|
11
|
Triller G, Vlachou EP, Hashemi H, van Straaten M, Zeelen JP, Kelemen Y, Baehr C, Marker CL, Ruf S, Svirina A, Chandra M, Urban K, Gkeka A, Kruse S, Baumann A, Miller AK, Bartel M, Pravetoni M, Stebbins CE, Papavasiliou FN, Verdi JP. A trypanosome-derived immunotherapeutics platform elicits potent high-affinity antibodies, negating the effects of the synthetic opioid fentanyl. Cell Rep 2023; 42:112049. [PMID: 36719797 PMCID: PMC10387133 DOI: 10.1016/j.celrep.2023.112049] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 12/02/2022] [Accepted: 01/13/2023] [Indexed: 01/31/2023] Open
Abstract
Poorly immunogenic small molecules pose challenges for the production of clinically efficacious vaccines and antibodies. To address this, we generate an immunization platform derived from the immunogenic surface coat of the African trypanosome. Through sortase-based conjugation of the target molecules to the variant surface glycoprotein (VSG) of the trypanosome surface coat, we develop VSG-immunogen array by sortase tagging (VAST). VAST elicits antigen-specific memory B cells and antibodies in a murine model after deploying the poorly immunogenic molecule fentanyl as a proof of concept. We also develop a single-cell RNA sequencing (RNA-seq)-based computational method that synergizes with VAST to specifically identify memory B cell-encoded antibodies. All computationally selected antibodies bind to fentanyl with picomolar affinity. Moreover, these antibodies protect mice from fentanyl effects after passive immunization, demonstrating the ability of these two coupled technologies to elicit therapeutic antibodies to challenging immunogens.
Collapse
Affiliation(s)
- Gianna Triller
- Division of Immune Diversity, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Evi P Vlachou
- Division of Immune Diversity, German Cancer Research Center, 69120 Heidelberg, Germany; Panosome GmbH, 69123 Heidelberg, Germany
| | - Hamidreza Hashemi
- Division of Immune Diversity, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Monique van Straaten
- Division of Structural Biology of Infection and Immunity, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Johan P Zeelen
- Division of Structural Biology of Infection and Immunity, German Cancer Research Center, 69120 Heidelberg, Germany
| | | | - Carly Baehr
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Cheryl L Marker
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Iuvo Bioscience, Rush, NY 14543, USA
| | - Sandra Ruf
- Division of Immune Diversity, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Anna Svirina
- Division of Immune Diversity, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Monica Chandra
- Panosome GmbH, 69123 Heidelberg, Germany; Division of Structural Biology of Infection and Immunity, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Katharina Urban
- Division of Immune Diversity, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Anastasia Gkeka
- Division of Immune Diversity, German Cancer Research Center, 69120 Heidelberg, Germany; Panosome GmbH, 69123 Heidelberg, Germany
| | | | - Andreas Baumann
- Cancer Drug Development Group, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Aubry K Miller
- Cancer Drug Development Group, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Marc Bartel
- Forensic Toxicology, Institute of Forensic and Traffic Medicine, Heidelberg University Hospital, 69115 Heidelberg, Germany
| | - Marco Pravetoni
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Department of Psychiatry and Behavioral Sciences, Department of Pharmacology, University of Washington School of Medicine, Center for Medication Development for Substance Use Disorders, Seattle, WA 98195, USA
| | - C Erec Stebbins
- Division of Structural Biology of Infection and Immunity, German Cancer Research Center, 69120 Heidelberg, Germany
| | - F Nina Papavasiliou
- Division of Immune Diversity, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Joseph P Verdi
- Division of Immune Diversity, German Cancer Research Center, 69120 Heidelberg, Germany; Hepione Therapeutics, Inc., New York, NY 10014, USA.
| |
Collapse
|
12
|
Rodarte JV, Baehr C, Hicks D, Liban TL, Weidle C, Rupert PB, Jahan R, Wall A, McGuire AT, Strong RK, Runyon S, Pravetoni M, Pancera M. Structures of drug-specific monoclonal antibodies bound to opioids and nicotine reveal a common mode of binding. Structure 2023; 31:20-32.e5. [PMID: 36513069 DOI: 10.1016/j.str.2022.11.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 08/03/2022] [Accepted: 11/16/2022] [Indexed: 12/15/2022]
Abstract
Opioid-related fatal overdoses have reached epidemic proportions. Because existing treatments for opioid use disorders offer limited long-term protection, accelerating the development of newer approaches is critical. Monoclonal antibodies (mAbs) are an emerging treatment strategy that targets and sequesters selected opioids in the bloodstream, reducing drug distribution across the blood-brain barrier, thus preventing or reversing opioid toxicity. We previously identified a series of murine mAbs with high affinity and selectivity for oxycodone, morphine, fentanyl, and nicotine. To determine their binding mechanism, we used X-ray crystallography to solve the structures of mAbs bound to their respective targets, to 2.2 Å resolution or higher. Structural analysis showed a critical convergent hydrogen bonding mode that is dependent on a glutamic acid residue in the mAbs' heavy chain and a tertiary amine of the ligand. Characterizing drug-mAb complexes represents a significant step toward rational antibody engineering and future manufacturing activities to support clinical evaluation.
Collapse
Affiliation(s)
- Justas V Rodarte
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Carly Baehr
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Dustin Hicks
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Tyler L Liban
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Connor Weidle
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Peter B Rupert
- Basic Science Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Rajwana Jahan
- Research Triangle Institute International, Research Triangle Park, Durham, NC, USA
| | - Abigail Wall
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Andrew T McGuire
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA, USA; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Roland K Strong
- Basic Science Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Scott Runyon
- Research Triangle Institute International, Research Triangle Park, Durham, NC, USA
| | - Marco Pravetoni
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, USA; Department of Psychiatry and Behavioral Sciences, Department of Pharmacology, School of Medicine, University of Washington, Seattle, WA, USA; Center for Medication Development for Substance Use Disorders and Overdose, University of Washington, Seattle, WA, USA.
| | - Marie Pancera
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|