Network


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

Hotspot


Dive into the research topics where Jonathan B. Rosenberg is active.

Publication


Featured researches published by Jonathan B. Rosenberg.


Molecular Therapy | 2011

Cocaine Analog Coupled to Disrupted Adenovirus: A Vaccine Strategy to Evoke High-titer Immunity Against Addictive Drugs

Martin J. Hicks; Bishnu P. De; Jonathan B. Rosenberg; Jesse T Davidson; Amira Y. Moreno; Kim D. Janda; Sunmee Wee; George F. Koob; Neil R. Hackett; Stephen M. Kaminsky; Stefan Worgall; Miklós Tóth; Jason G. Mezey; Ronald G. Crystal

Based on the concept that anticocaine antibodies could prevent inhaled cocaine from reaching its target receptors in the brain, an effective anticocaine vaccine could help reverse cocaine addiction. Leveraging the knowledge that E1(-)E3(-) adenovirus (Ad) gene transfer vectors are potent immunogens, we have developed a novel vaccine platform for addictive drugs by covalently linking a cocaine analog to the capsid proteins of noninfectious, disrupted Ad vector. The Ad-based anticocaine vaccine evokes high-titer anticocaine antibodies in mice sufficient to completely reverse, on a persistent basis, the hyperlocomotor activity induced by intravenous administration of cocaine.


Neuropsychopharmacology | 2012

Novel Cocaine Vaccine Linked to a Disrupted Adenovirus Gene Transfer Vector Blocks Cocaine Psychostimulant and Reinforcing Effects

Sunmee Wee; Martin J. Hicks; Bishnu P. De; Jonathan B. Rosenberg; Amira Y. Moreno; Stephen M. Kaminsky; Kim D. Janda; Ronald G. Crystal; George F. Koob

Immunotherapy is a promising treatment for drug addiction. However, insufficient immune responses to vaccines in most subjects pose a challenge. In this study, we tested the efficacy of a new cocaine vaccine (dAd5GNE) in antagonizing cocaine addiction-related behaviors in rats. This vaccine used a disrupted serotype 5 adenovirus (Ad) gene transfer vector coupled to a third-generation cocaine hapten, termed GNE (6-(2R,3S)-3-(benzoyloxy)-8-methyl-8-azabicyclo [3.2.1] octane-2-carboxamido-hexanoic acid). Three groups of rats were immunized with dAd5GNE. One group was injected with 3H-cocaine, and radioactivity in the blood and brain was determined. A second group was tested for cocaine-induced locomotor sensitization. A third group was examined for cocaine self-administration, extinction, and reinstatement of responding for cocaine. Antibody titers were determined at various time-points. In each experiment, we added a control group that was immunized with dAd5 without a hapten. The vaccination with dAd5GNE produced long-lasting high titers (>105) of anti-cocaine antibodies in all of the rats. The vaccination inhibited cocaine-induced hyperlocomotor activity and sensitization. Vaccinated rats acquired cocaine self-administration, but they showed less motivation to self-administer cocaine under a progressive-ratio schedule than control rats. When cocaine was not available in a session, control rats exhibited ‘extinction burst’ responding, whereas vaccinated rats did not. Moreover, when primed with cocaine, vaccinated rats did not reinstate responding, suggesting a blockade of cocaine-seeking behavior. These data strongly suggest that our dAd5GNE vector-based vaccine may be effective in treating cocaine abuse and addiction.


Science Translational Medicine | 2012

Aav-directed persistent expression of an anti-nicotine antibody gene for smoking cessation

Ronald G. Crystal; Martin J. Hicks; Jonathan B. Rosenberg; Bishnu P. De; Stephen M. Kaminsky; Robin L. Davisson

Gene therapy with an anti-nicotine monoclonal antibody limits nicotine access to the brain in mice and may be a potential therapy for cigarette addiction. Extinguishing Addiction Those who smoke and try to quit have variously described their feelings as a never-ending hunger, terror, immense panic, rage, and the loss of a loved one—reactions that stem from nicotine addiction and withdrawal. Given cigarette smoke’s destructive effects on health and cost to society, a means of overcoming such addiction would be enormously beneficial. For most smokers, current antismoking therapies fail to work. One newer idea is an anti-nicotine vaccine; in this approach, nicotine (coupled to a larger molecule) is administered, generating an immune response. The resulting anti-nicotine antibodies bind the nicotine from cigarette smoke in the blood, intercepting the drug before it affects reward centers in the brain. In disappointing clinical trials, however, such vaccines result in variable antibody titers and have only limited success in halting smoking, with best results seen in people with the strongest antibody response. Hicks et al. now describe a different tactic to get to the same end; they use gene transfer to attain persistent, high levels of anti-nicotine antibodies in smokers’ blood. The researchers constructed an adeno-associated virus–based vector that expressed a high-affinity anti-nicotine monoclonal antibody. Mice injected with a single dose of this vector made high antibody titers (which were in 40-fold molar excess over the nicotine concentrations seen in people who smoke continuously), which remained high for at least 18 weeks. When these mice were injected with nicotine, the antibodies effectively sequestered this compound in the blood: nicotine concentrations in the brain were only 15% of those in brains of mice that did not express the antibodies. Furthermore, the usual nicotine-induced changes in blood pressure, heart rate, and locomotor activity were abolished or greatly reduced in mice that expressed the anti-nicotine antibodies. Further work will be needed to test this vector in a rodent model trained to self-administer nicotine, because the mice in this study were not addicted to the drug. Successful results from such a test would then support investigating this approach in clinical trials. Current strategies to help tobacco smokers quit have limited success as a result of the addictive properties of the nicotine in cigarette smoke. We hypothesized that a single administration of an adeno-associated virus (AAV) gene transfer vector expressing high levels of an anti-nicotine antibody would persistently prevent nicotine from reaching its receptors in the brain. To test this hypothesis, we constructed an AAVrh.10 vector that expressed a full-length, high-affinity, anti-nicotine antibody derived from the Fab fragment of the anti-nicotine monoclonal antibody NIC9D9 (AAVantiNic). In mice treated with this vector, blood concentrations of the anti-nicotine antibody were dose-dependent, and the antibody showed high specificity and affinity for nicotine. The antibody shielded the brain from systemically administered nicotine, reducing brain nicotine concentrations to 15% of those in naïve mice. The amount of nicotine sequestered in the serum of vector-treated mice was more than seven times greater than that in untreated mice, with 83% of serum nicotine bound to immunoglobulin G. Treatment with the AAVantiNic vector blocked nicotine-mediated alterations in arterial blood pressure, heart rate, and locomotor activity. In summary, a single administration of a gene transfer vector expressing a high-affinity anti-nicotine monoclonal antibody elicited persistent (18 weeks), high titers of an anti-nicotine antibody that obviated the physiologic effects of nicotine. If this degree of efficacy translates to humans, AAVantiNic could be an effective preventative therapy for nicotine addiction.


Human Gene Therapy | 2012

AAVrh.10-mediated expression of an anti-cocaine antibody mediates persistent passive immunization that suppresses cocaine-induced behavior.

Jonathan B. Rosenberg; Martin J. Hicks; Bishnu P. De; Odelya E. Pagovich; Esther Frenk; Kim D. Janda; Sunmee Wee; George F. Koob; Neil R. Hackett; Stephen M. Kaminsky; Stefan Worgall; Nicole Tignor; Jason G. Mezey; Ronald G. Crystal

Cocaine addiction is a major problem affecting all societal and economic classes for which there is no effective therapy. We hypothesized an effective anti-cocaine vaccine could be developed by using an adeno-associated virus (AAV) gene transfer vector as the delivery vehicle to persistently express an anti-cocaine monoclonal antibody in vivo, which would sequester cocaine in the blood, preventing access to cognate receptors in the brain. To accomplish this, we constructed AAVrh.10antiCoc.Mab, an AAVrh.10 gene transfer vector expressing the heavy and light chains of the high affinity anti-cocaine monoclonal antibody GNC92H2. Intravenous administration of AAVrh.10antiCoc.Mab to mice mediated high, persistent serum levels of high-affinity, cocaine-specific antibodies that sequestered intravenously administered cocaine in the blood. With repeated intravenous cocaine challenge, naive mice exhibited hyperactivity, while the AAVrh.10antiCoc.Mab-vaccinated mice were completely resistant to the cocaine. These observations demonstrate a novel strategy for cocaine addiction by requiring only a single administration of an AAV vector mediating persistent, systemic anti-cocaine passive immunity.


Neuropsychopharmacology | 2013

Adenovirus capsid-based anti-cocaine vaccine prevents cocaine from binding to the nonhuman primate CNS dopamine transporter.

Anat Maoz; Martin J. Hicks; Shankar Vallabhjosula; Michael Synan; Paresh J. Kothari; Jonathan P. Dyke; Douglas Ballon; Stephen Kaminsky; Bishnu P. De; Jonathan B. Rosenberg; Diana Martinez; George F. Koob; Kim D. Janda; Ronald G. Crystal

Cocaine addiction is a major problem for which there is no approved pharmacotherapy. We have developed a vaccine to cocaine (dAd5GNE), based on the cocaine analog GNE linked to the capsid proteins of a serotype 5 adenovirus, designed to evoke anti-cocaine antibodies that sequester cocaine in the blood, preventing access to the CNS. To assess the efficacy of dAd5GNE in a large animal model, positron emission tomography (PET) and the radiotracer [11C]PE2I were used to measure cocaine occupancy of the dopamine transporter (DAT) in nonhuman primates. Repeat administration of dAd5GNE induced high anti-cocaine titers. Before vaccination, cocaine displaced PE2I from DAT in the caudate and putamen, resulting in 62±4% cocaine occupancy. In contrast, dAd5GNE-vaccinated animals showed reduced cocaine occupancy such that when anti-cocaine titers were >4 × 105, the cocaine occupancy was reduced to levels of <20%, significantly below the 47% threshold required to evoke the subjective ‘high’ reported in humans.


Human Gene Therapy | 2013

Disrupted Adenovirus-Based Vaccines Against Small Addictive Molecules Circumvent Anti-Adenovirus Immunity

Bishnu P. De; Odelya E. Pagovich; Martin J. Hicks; Jonathan B. Rosenberg; Amira Y. Moreno; Kim D. Janda; George F. Koob; Stefan Worgall; Stephen M. Kaminsky; Dolan Sondhi; Ronald G. Crystal

Adenovirus (Ad) vaccine vectors have been used for many applications due to the capacity of the Ad capsid proteins to evoke potent immune responses, but these vectors are often ineffective in the context of pre-existing anti-Ad immunity. Leveraging the knowledge that E1(-)E3(-) Ad gene transfer vectors are potent immunogens, we have developed a vaccine platform against small molecules by covalently coupling analogs of small molecules to the capsid proteins of disrupted Ad (dAd5). We hypothesized that the dAd5 platform would maintain immunopotency even in the context of anti-Ad neutralizing antibodies. To test this hypothesis, we coupled cocaine and nicotine analogs, GNE and AM1, to dAd5 capsid proteins to generate dAd5GNE and dAd5AM1, respectively. Mice were pre-immunized with Ad5Null, resulting in high titer anti-Ad5 neutralizing antibodies comparable to those observed in the human population. The dAd5GNE and dAd5AM1 vaccines elicited high anti-cocaine and anti-nicotine antibody titers, respectively, in both naive and Ad5-immune mice, and both functioned to prevent cocaine or nicotine from reaching the brain of anti-Ad immune mice. Thus, disrupted Ad5 evokes potent humoral immunity that is effective in the context of pre-existing neutralizing anti-Ad immunity, overcoming a major limitation for current Ad-based vaccines.


Cns & Neurological Disorders-drug Targets | 2011

Anti-Cocaine Vaccine Based on Coupling a Cocaine Analog to a Disrupted Adenovirus

George F. Koob; Martin J. Hicks; Sunmee Wee; Jonathan B. Rosenberg; Bishnu P. De; Stephen M. Kaminksy; Amira Y. Moreno; Kim D. Janda; Ronald G. Crystal

The challenge in developing an anti-cocaine vaccine is that cocaine is a small molecule, invisible to the immune system. Leveraging the knowledge that adenovirus (Ad) capsid proteins are highly immunogenic in humans, we hypothesized that linking a cocaine hapten to Ad capsid proteins would elicit high-affinity, high-titer antibodies against cocaine, sufficient to sequester systemically administered cocaine and prevent access to the brain, thus suppressing cocaine-induced behaviors. Based on these concepts, we developed dAd5GNE, a disrupted E1-E3- serotype 5 Ad with GNE, a stable cocaine analog, covalently linked to the Ad capsid proteins. In pre-clinical studies, dAd5GNE evoked persistent, high titer, high affinity IgG anti-cocaine antibodies, and was highly effective in blocking cocaine-induced hyperactivity and cocaine self-administration behavior in rats. Future studies will be designed to expand the efficacy studies, carry out relevant toxicology studies, and test dAd5GNE in human cocaine addicts.


Human gene therapy. Clinical development | 2014

Comparative Efficacy and Safety of Multiple Routes of Direct CNS Administration of Adeno-Associated Virus Gene Transfer Vector Serotype rh.10 Expressing the Human Arylsulfatase A cDNA to Nonhuman Primates

Jonathan B. Rosenberg; Dolan Sondhi; David G. Rubin; Sebastien Monette; Alvin Chen; Sara Cram; Bishnu P. De; Stephen M. Kaminsky; Caroline Sevin; Patrick Aubourg; Ronald G. Crystal

Metachromatic leukodystrophy (MLD), a fatal disorder caused by deficiency of the lysosomal enzyme arylsulfatase A (ARSA), is associated with an accumulation of sulfatides, causing widespread demyelination in both central and peripheral nervous systems. On the basis of prior studies demonstrating that adeno-associated virus AAVrh.10 can mediate widespread distribution in the CNS of a secreted lysosomal transgene, and as a prelude to human trials, we comparatively assessed the optimal CNS delivery route of an AAVrh.10 vector encoding human ARSA in a large animal model for broadest distribution of ARSA enzyme. Five routes were tested (each total dose, 1.5 × 10(12) genome copies of AAVrh.10hARSA-FLAG): (1) delivery to white matter centrum ovale; (2) deep gray matter delivery (putamen, thalamus, and caudate) plus overlying white matter; (3) convection-enhanced delivery to same deep gray matter locations; (4) lateral cerebral ventricle; and (5) intraarterial delivery with hyperosmotic mannitol to the middle cerebral artery. After 13 weeks, the distribution of ARSA activity subsequent to each of the three direct intraparenchymal administration routes was significantly higher than in phosphate-buffered saline-administered controls, but administration by the intraventricular and intraarterial routes failed to demonstrate measurable levels above controls. Immunohistochemical staining in the cortex, white matter, deep gray matter of the striatum, thalamus, choroid plexus, and spinal cord dorsal root ganglions confirmed these results. Of the five routes studied, administration to the white matter generated the broadest distribution of ARSA, with 80% of the brain displaying more than a therapeutic (10%) increase in ARSA activity above PBS controls. No significant toxicity was observed with any delivery route as measured by safety parameters, although some inflammatory changes were seen by histopathology. We conclude that AAVrh.10-mediated delivery of ARSA via CNS administration into the white matter is likely to be safe and yields the widest distribution of ARSA, making it the most suitable route of vector delivery.


Human gene therapy. Clinical development | 2014

Fate of Systemically Administered Cocaine in Nonhuman Primates Treated with the dAd5GNE Anticocaine Vaccine

Martin J. Hicks; Stephen M. Kaminsky; Bishnu P. De; Jonathan B. Rosenberg; Suzette M. Evans; David M. Andrenyak; David E. Moody; George F. Koob; Kim D. Janda; Rodolfo J. Ricart Arbona; Michelle Lepherd; Ronald G. Crystal

Cocaine use disorders are mediated by the cocaine blockade of the dopamine transporter in the central nervous system (CNS). On the basis of the concept that these effects could be obviated if cocaine were prevented from reaching its cognate receptors in the CNS, we have developed an anticocaine vaccine, dAd5GNE, based on a cocaine analog covalently linked to capsid proteins of an E1(-)E3(-) serotype 5 adenovirus. While the vaccine effectively blocks systemically administered cocaine from reaching the brain by mediating sequestration of the cocaine in the blood, the fact that cocaine also has significant peripheral effects raises concerns that vaccination-mediated redistribution could lead to adverse effects in the visceral organs. The distribution of systemically administered cocaine at a weight-adjusted typical human dose was evaluated along with cocaine metabolites in both dAd5GNE-vaccinated and control nonhuman primates. dAd5GNE sequestration of cocaine to the blood not only prevented cocaine access to the CNS, but also limited access of both the drug and its metabolites to other cocaine-sensitive organs. The levels of cocaine in the blood of vaccinated animals rapidly decreased, suggesting that while the antibody limits access of the drug and its active metabolites to the brain and sensitive organs of the periphery, it does not prolong drug levels in the blood compartment. Gross and histopathology of major organs found no vaccine-mediated untoward effects. These results build on our earlier measures of efficacy and demonstrate that the dAd5GNE vaccine-mediated redistribution of administered cocaine is not likely to impact the vaccine safety profile.


Human Gene Therapy | 2013

Suppression of Nicotine-Induced Pathophysiology by an Adenovirus Hexon-Based Antinicotine Vaccine

Jonathan B. Rosenberg; Bishnu P. De; Martin J. Hicks; Kim D. Janda; Stephen M. Kaminsky; Stefan Worgall; Ronald G. Crystal

Despite antismoking campaigns, cigarette smoking remains a pervasive addiction with significant societal impact, accounting for one of every five deaths. Smoking cessation therapies to help smokers quit are ineffective with a high recidivism rate. With the knowledge that nicotine is the principal addictive compound of cigarettes, we have developed an antismoking vaccine based on the highly immunogenic properties of the hexon protein purified from the serotype 5 adenovirus (Ad) capsid. We hypothesized that an effective antinicotine vaccine could be based on coupling the nicotine hapten AM1 to purified Ad hexon protein. To assess this, AM1 was conjugated to hexon purified from serotype 5 Ad to produce the HexonAM1 vaccine. C57Bl/6 mice were sensitized by 10 daily nicotine administrations (0.5 mg/kg, subcutaneous) to render the mice addicted to nicotine. Control groups were sensitized to phosphate-buffered saline (PBS). The mice were then immunized with HexonAM1 (4 μg, intramuscular) at 0, 3, and 6 weeks. By 6 weeks, the HexonAM1-vaccinated mice had serum antinicotine antibody titers of 1.1×10(6)±7.6×10(4). To demonstrate that these high antinicotine titers were sufficient to suppress the effects of nicotine, HexonAM1-vaccinated mice were evaluated for nicotine-induced hypoactive behavior with nicotine challenges (0.5 mg/kg wt) over 5 weeks. In all challenges, the HexonAM1-vaccinated mice behaved similar to PBS-challenged naive mice. These data demonstrate that a vaccine comprised of a nicotine analog coupled to Ad hexon can evoke a high level of antinicotine antibodies sufficient to inhibit nicotine-induced behavior. The HexonAM1 vaccine represents a platform paradigm for vaccines against small molecules.

Collaboration


Dive into the Jonathan B. Rosenberg's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kim D. Janda

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

George F. Koob

National Institute on Drug Abuse

View shared research outputs
Top Co-Authors

Avatar

Amira Y. Moreno

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Brian R. Davis

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge