Network


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

Hotspot


Dive into the research topics where Benjamin M. Ford is active.

Publication


Featured researches published by Benjamin M. Ford.


Journal of Medicinal Chemistry | 2013

Design, Synthesis and Biological Evaluation of Aminoalkylindole Derivatives as Cannabinoid Receptor Ligands with Potential for Treatment of Alcohol Abuse

Tamara Vasiljevik; Lirit N. Franks; Benjamin M. Ford; Justin T. Douglas; Paul L. Prather; William E. Fantegrossi; Thomas E. Prisinzano

Attenuation of increased endocannabinoid signaling with a CB1R neutral antagonist might offer a new therapeutic direction for treatment of alcohol abuse. We have recently reported that a monohydroxylated metabolite of the synthetic aminoalkylindole cannabinoid JHW-073 (3) exhibits neutral antagonist activity at CB1Rs and thus may serve as a promising lead for the development of novel alcohol abuse therapies. In the current study, we show that systematic modification of an aminoalkylindole scaffold identified two new compounds with dual CB1R antagonist/CB2R agonist activity. Similar to the CB1R antagonist/inverse agonist rimonabant, analogues 27 and 30 decrease oral alcohol self-administration without affecting total fluid intake and block the development of alcohol-conditioned place preference. Collectively, these initial findings suggest that design and systematic modification of aminoalkylindoles such as 3 may lead to development of novel cannabinoid ligands with dual CB1R antagonist/CB2R agonist activity with potential for use as treatments of alcohol abuse.


Bioorganic & Medicinal Chemistry Letters | 2013

Evaluation of (Z)-2-((1-benzyl-1H-indol-3-yl)methylene)-quinuclidin-3-one analogues as novel, high affinity ligands for CB1 and CB2 cannabinoid receptors.

Nikhil Reddy Madadi; Narsimha Reddy Penthala; Lisa K. Brents; Benjamin M. Ford; Paul L. Prather; Peter A. Crooks

A small library of N-benzyl indolequinuclidinone (IQD) analogs has been identified as a novel class of cannabinoid ligands. The affinity and selectivity of these IQDs for the two established cannabinoid receptor subtypes, CB1 and CB2, was evaluated. Compounds 8 (R=R(2)=H, R(1)=F) and 13 (R=COOCH3, R(1)=R(2)=H) exhibited high affinity for CB2 receptors with Ki values of 1.33 and 2.50 nM, respectively, and had lower affinities for the CB1 receptor (Ki values of 9.23 and 85.7 nM, respectively). Compound 13 had the highest selectivity of all the compounds examined, and represents a potent cannabinoid ligand with 34-times greater selectivity for CB2R over CB1R. These findings are significant for future drug development, given recent reports demonstrating beneficial use of cannabinoid ligands in a wide variety of human disease states including drug abuse, depression, schizophrenia, inflammation, chronic pain, obesity, osteoporosis and cancer.


Biochemical and Biophysical Research Communications | 2013

CB1 and CB2 receptors are novel molecular targets for Tamoxifen and 4OH-Tamoxifen.

Paul L. Prather; FeAna FrancisDevaraj; Centdrika R. Dates; Aleksandra K. Greer; Stacie M. Bratton; Benjamin M. Ford; Lirit N. Franks; Anna Radominska-Pandya

Tamoxifen (Tam) is classified as a selective estrogen receptor modulator (SERM) and is used for treatment of patients with ER-positive breast cancer. However, it has been shown that Tam and its cytochrome P450-generated metabolite 4-hydroxy-Tam (4OH-Tam) also exhibit cytotoxic effects in ER-negative breast cancer cells. These observations suggest that Tam and 4OH-Tam can produce cytotoxicity via estrogen receptor (ER)-independent mechanism(s) of action. The molecular targets responsible for the ER-independent effects of Tam and its derivatives are poorly understood. Interestingly, similar to Tam and 4OH-Tam, cannabinoids have also been shown to exhibit anti-proliferative and apoptotic effects in ER-negative breast cancer cells, and estrogen can regulate expression levels of cannabinoid receptors (CBRs). Therefore, this study investigated whether CBRs might serve as novel molecular targets for Tam and 4OH-Tam. We report that both compounds bind to CB1 and CB2Rs with moderate affinity (0.9-3 μM). Furthermore, Tam and 4OH-Tam exhibit inverse activity at CB1 and CB2Rs in membrane preparations, reducing basal G-protein activity. Tam and 4OH-Tam also act as CB1/CB2R-inverse agonists to regulate the downstream intracellular effector adenylyl cyclase in intact cells, producing concentration-dependent increases in intracellular cAMP. These results suggest that CBRs are molecular targets for Tam and 4OH-Tam and may contribute to the ER-independent cytotoxic effects reported for these drugs. Importantly, these findings also indicate that Tam and 4OH-Tam might be used as structural scaffolds for development of novel, efficacious, non-toxic cancer drugs acting via CB1 and/or CB2Rs.


European Journal of Pharmacology | 2014

Characterization of the intrinsic activity for a novel class of cannabinoid receptor ligands: Indole quinuclidine analogs

Lirit N. Franks; Benjamin M. Ford; Nikhil Reddy Madadi; Narsimha Reddy Penthala; Peter A. Crooks; Paul L. Prather

Our laboratory recently reported that a group of novel indole quinuclidine analogs bind with nanomolar affinity to cannabinoid type-1 and type-2 receptors. This study characterized the intrinsic activity of these compounds by determining whether they exhibit agonist, antagonist, or inverse agonist activity at cannabinoid type-1 and/or type-2 receptors. Cannabinoid receptors activate Gi/Go-proteins that then proceed to inhibit activity of the downstream intracellular effector adenylyl cyclase. Therefore, intrinsic activity was quantified by measuring the ability of compounds to modulate levels of intracellular cAMP in intact cells. Concerning cannabinoid type-1 receptors endogenously expressed in Neuro2A cells, a single analog exhibited agonist activity, while eight acted as neutral antagonists and two possessed inverse agonist activity. For cannabinoid type-2 receptors stably expressed in CHO cells, all but two analogs acted as agonists; these two exceptions exhibited inverse agonist activity. Confirming specificity at cannabinoid type-1 receptors, modulation of adenylyl cyclase activity by all proposed agonists and inverse agonists was blocked by co-incubation with the neutral cannabinoid type-1 antagonist O-2050. All proposed cannabinoid type-1 receptor antagonists attenuated adenylyl cyclase modulation by cannabinoid agonist CP-55,940. Specificity at cannabinoid type-2 receptors was confirmed by failure of all compounds to modulate adenylyl cyclase activity in CHO cells devoid of cannabinoid type-2 receptors. Further characterization of select analogs demonstrated concentration-dependent modulation of adenylyl cyclase activity with potencies similar to their respective affinities for cannabinoid receptors. Therefore, indole quinuclidines are a novel structural class of compounds exhibiting high affinity and a range of intrinsic activity at cannabinoid type-1 and type-2 receptors.


Pharmacological Research | 2016

Antinociceptive effects of the 6-O-sulfate ester of morphine in normal and diabetic rats: Comparative role of mu- and delta-opioid receptors.

Jai Shankar K. Yadlapalli; Benjamin M. Ford; Amit Ketkar; Anqi Wan; Narasimha R. Penthala; Robert L. Eoff; Paul L. Prather; Maxim Dobretsov; Peter A. Crooks

This study determined the antinociceptive effects of morphine and morphine-6-O-sulfate (M6S) in both normal and diabetic rats, and evaluated the comparative role of mu-opioid receptors (mu-ORs) and delta-opioid receptors (delta-ORs) in the antinociceptive action of these opioids. In vitro characterization of mu-OR and delta-OR-mediated signaling by M6S and morphine in stably transfected Chinese hamster ovary (CHO-K1) cells showed that M6S exhibited a 6-fold higher affinity for delta-ORs and modulated G-protein and adenylyl cyclase activity via delta-ORs more potently than morphine. Interestingly, while morphine acted as a full agonist at delta-ORs in both functional assays examined, M6S exhibited either partial or full agonist activity for modulation of G-protein or adenylyl cyclase activity, respectively. Molecular docking studies indicated that M6S but not morphine binds equally well at the ligand binding site of both mu- and delta-ORs. In vivo analgesic effects of M6S and morphine in both normal and streptozotocin-induced diabetic Sprague-Dawley rats utilizing the hot water tail flick latency test showed that M6S produced more potent antinociception than morphine in both normal rats and diabetic rats. This difference in potency was abrogated following antagonism of delta- but not mu- or kappa (kappa-ORs) opioid receptors. During 9days of chronic treatment, tolerance developed to morphine-treated but not to M6S-treated rats. Rats that developed tolerance to morphine still remained responsive to M6S. Collectively, this study demonstrates that M6S is a potent and efficacious mu/delta opioid analgesic with a delayed tolerance profile when compared to morphine in both normal and diabetic rats. PERSPECTIVE This study demonstrates that M6S acts at both mu- and delta-ORs, and adds to the growing evidence that the use of mixed mu/delta opioid agonists in pain treatment may have clinical benefit.


Pharmacological Research | 2017

Characterization of structurally novel G protein biased CB1 agonists: Implications for drug development

Benjamin M. Ford; Lirit N. Franks; Sherrica Tai; William E. Fantegrossi; Edward L. Stahl; Michael D. Berquist; Christian V. Cabanlong; Catheryn D. Wilson; Narsimha Reddy Penthala; Peter A. Crooks; Paul L. Prather

&NA; The human cannabinoid subtype 1 receptor (hCB1R) is highly expressed in the CNS and serves as a therapeutic target for endogenous ligands as well as plant‐derived and synthetic cannabinoids. Unfortunately, acute use of hCB1R agonists produces unwanted psychotropic effects and chronic administration results in development of tolerance and dependence, limiting the potential clinical use of these ligands. Studies in &bgr;‐arrestin knockout mice suggest that interaction of certain GPCRs, including &mgr;‐, &dgr;‐, &kgr;‐opioid and hCB1Rs, with &bgr;‐arrestins might be responsible for several adverse effects produced by agonists acting at these receptors. Indeed, agonists that bias opioid receptor activation toward G‐protein, relative to &bgr;‐arrestin signaling, produce less severe adverse effects. These observations indicate that therapeutic utility of agonists acting at hCB1Rs might be improved by development of G‐protein biased hCB1R agonists. Our laboratory recently reported a novel class of indole quinulidinone (IQD) compounds that bind cannabinoid receptors with relatively high affinity and act with varying efficacy. The purpose of this study was to determine whether agonists in this novel cannabinoid class exhibit ligand bias at hCB1 receptors. Our studies found that a novel IQD‐derived hCB1 receptor agonist PNR‐4‐20 elicits robust G protein‐dependent signaling, with transduction ratios similar to the non‐biased hCB1R agonist CP‐55,940. In marked contrast to CP‐55,940, PNR‐4‐20 produces little to no &bgr;‐arrestin 2 recruitment. Quantitative calculation of bias factors indicates that PNR‐4‐20 exhibits from 5.4‐fold to 29.5‐fold bias for G protein, relative to &bgr;‐arrestin 2 signaling (when compared to G protein activation or inhibition of forskolin‐stimulated cAMP accumulation, respectively). Importantly, as expected due to reduced &bgr;‐arrestin 2 recruitment, chronic exposure of cells to PNR‐4‐20 results in significantly less desensitization and down‐regulation of hCB1Rs compared to similar treatment with CP‐55,940. PNR‐4‐20 (i.p.) is active in the cannabinoid tetrad in mice and chronic treatment results in development of less persistent tolerance and no significant withdrawal signs when compared to animals repeatedly exposed to the non‐biased full agoinst JWH‐018 or &Dgr;9‐THC. Finally, studies of a structurally similar analog PNR‐ 4‐02 show that it is also a G protein biased hCB1R agonist. It is predicted that cannabinoid agonists that bias hCB1R activation toward G protein, relative to &bgr;‐arrestin 2 signaling, will produce fewer and less severe adverse effects both acutely and chronically. Graphical abstract Figure. No caption available.


PLOS ONE | 2016

Tamoxifen Isomers and Metabolites Exhibit Distinct Affinity and Activity at Cannabinoid Receptors: Potential Scaffold for Drug Development.

Benjamin M. Ford; Lirit N. Franks; Anna Radominska-Pandya; Paul L. Prather; James W. Porter

Tamoxifen (Tam) is a selective estrogen receptor (ER) modulator (SERM) that is an essential drug to treat ER-positive breast cancer. Aside from known actions at ERs, recent studies have suggested that some SERMs like Tam also exhibit novel activity at cannabinoid subtype 1 and 2 receptors (CB1R and CB2Rs). Interestingly, cis- (E-Tam) and trans- (Z-Tam) isomers of Tam exhibit over a 100-fold difference in affinity for ERs. Therefore, the current study assessed individual isomers of Tam and subsequent cytochrome P450 metabolic products, 4-hydroxytamoxifen (4OHT) and 4-hydroxy-N-desmethyl tamoxifen (End) for affinity and activity at CBRs. Results showed that Z-4OHT, but not Z-Tam or Z-End, exhibits higher affinity for both CB1 and CB2Rs relative to the E-isomer. Furthermore, Z- and E-isomers of Tam and 4OHT show slightly higher affinity for CB2Rs, while both End isomers are relatively CB1R-selective. When functional activity was assessed by G-protein activation and regulation of the downstream effector adenylyl cyclase, all isomers examined act as full CB1 and CB2R inverse agonists. Interestingly, Z-Tam appears to be more efficacious than the full inverse agonist AM630 at CB2Rs, while both Z-Tam and Z-End exhibit characteristics of insurmountable antagonism at CB1 and CB2Rs, respectively. Collectively, these results suggest that the SERMs Tam, 4OHT and End elicit ER-independent actions via CBRs in an isomer-specific manner. As such, this novel structural scaffold might be used to develop therapeutically useful drugs for treatment of a variety of diseases mediated via CBRs.


Toxicology and Applied Pharmacology | 2018

The tamoxifen derivative ridaifen-B is a high affinity selective CB 2 receptor inverse agonist exhibiting anti-inflammatory and anti-osteoclastogenic effects

Lirit N. Franks; Benjamin M. Ford; Toshifumi Fujiwara; Haibo Zhao; Paul L. Prather

&NA; Selective estrogen receptor modulators (SERMs) target estrogen receptors (ERs) to treat breast cancer and osteoporosis. Several SERMs exhibit anti‐cancer activity not related to ERs. To discover novel anti‐cancer drugs acting via ER‐independent mechanisms, derivatives of the SERM tamoxifen, known as the “ridaifen” compounds, have been developed that exhibit reduced or no ER affinity, while maintaining cytotoxicity. Tamoxifen and other SERMs bind to cannabinoid receptors with moderate affinity. Therefore, ER‐independent effects of SERMs might be mediated via cannabinoid receptors. This study determined whether RID‐B, a first generation ridaifen compound, exhibits affinity and/or activity at CB1 and/or CB2 cannabinoid receptors. RID‐B binds with high affinity (Ki = 43.7 nM) and 17‐fold selectivity to CB2 over CB1 receptors. RID‐B acts as an inverse agonist at CB2 receptors, modulating G‐protein and adenylyl cyclase activity with potency values predicted by CB2 affinity. Characteristic of an antagonist, RID‐B co‐incubation produces a parallel‐rightward shift in the concentration‐effect curve of CB2 agonist WIN‐55,212‐2 to inhibit adenylyl cyclase activity. CB2 inverse agonists are reported to exhibit anti‐inflammatory and anti‐ostoeclastogenic effects. In LPS‐activated macrophages, RID‐B exhibits anti‐inflammatory effects by reducing levels of nitric oxide (NO), IL‐6 and IL‐1&agr;, but not TNF&agr;. Only reduction of NO concentration by RID‐B is mediated by cannabinoid receptors. RID‐B also exhibits pronounced anti‐osteoclastogenic effects, reducing the number of osteoclasts differentiating from primary bone marrow macrophages in a cannabinoid receptor‐dependent manner. In summary, the tamoxifen derivative RID‐B, developed with reduced affinity for ERs, is a high affinity selective CB2 inverse agonist with anti‐inflammatory and anti‐osteoclastogenic properties. HighlightsSeveral SERMs exhibit anti‐cancer activity not related to estrogen receptors.Several SERMs also bind to cannabinoid receptors with high affinity.Estrogen receptor‐independent effects of SERMs might be mediated via cannabinoid receptors.The tamoxifen derivative RID‐B, with reduced affinity for ERs, is a selective CB2 inverse agonist.RID‐B produces anti‐inflammatory and anti‐osteoclastogenic effects via cannabinoid receptors.


Frontiers in Pharmacology | 2018

Atypical Pharmacodynamic Properties and Metabolic Profile of the Abused Synthetic Cannabinoid AB-PINACA: Potential Contribution to Pronounced Adverse Effects Relative to Δ9-THC

Rachel D. Hutchison; Benjamin M. Ford; Lirit N. Franks; Catheryn D. Wilson; Azure L. Yarbrough; Ryoichi Fujiwara; Mark Su; Denise Fernandez; Laura P. James; Jeffery H. Moran; Amy L. Patton; William E. Fantegrossi; Anna Radominska-Pandya; Paul L. Prather

Recreational use of marijuana is associated with few adverse effects, but abuse of synthetic cannabinoids (SCBs) can result in anxiety, psychosis, chest pain, seizures and death. To potentially explain higher toxicity associated with SCB use, we hypothesized that AB-PINACA, a common second generation SCB, exhibits atypical pharmacodynamic properties at CB1 cannabinoid receptors (CB1Rs) and/or a distinct metabolic profile when compared to Δ9-tetrahydrocannabinol (Δ9-THC), the principal psychoactive cannabinoid present in marijuana. Liquid chromatography tandem mass spectrometry (LC/MS) identified AB-PINACA and monohydroxy metabolite(s) as primary phase I metabolites (4OH-AB-PINACA and/or 5OH-AB-PINACA) in human urine and serum obtained from forensic samples. In vitro experiments demonstrated that when compared to Δ9-THC, AB-PINACA exhibits similar affinity for CB1Rs, but greater efficacy for G-protein activation and higher potency for adenylyl cyclase inhibition. Chronic treatment with AB-PINACA also results in greater desensitization of CB1Rs (e.g., tolerance) than Δ9-THC. Importantly, monohydroxy metabolites of AB-PINACA retain affinity and full agonist activity at CB1Rs. Incubation of 4OH-AB-PINACA and 5OH-AB-PINACA with human liver microsomes (HLMs) results in limited glucuronide formation when compared to that of JWH-018-M2, a major monohydroxylated metabolite of the first generation SCB JWH-018. Finally, AB-PINACA and 4OH-AB-PINACA are active in vivo, producing CB1R-mediated hypothermia in mice. Taken collectively, the atypical pharmacodynamic properties of AB-PINACA at CB1Rs relative to Δ9-THC (e.g., higher potency/efficacy and greater production of desensitization), coupled with an unusual metabolic profile (e.g., production of metabolically stable active phase I metabolites) may contribute to the pronounced adverse effects observed with abuse of this SCB compared to marijuana.


Frontiers in Pharmacology | 2016

Selective Estrogen Receptor Modulators: Cannabinoid Receptor Inverse Agonists with Differential CB1 and CB2 Selectivity

Lirit N. Franks; Benjamin M. Ford; Paul L. Prather

Selective estrogen receptor modulators (SERMs) are used to treat estrogen receptor (ER)-positive breast cancer and osteoporosis. Interestingly, tamoxifen and newer classes of SERMs also exhibit cytotoxic effects in cancers devoid of ERs, indicating a non-estrogenic mechanism of action. Indicative of a potential ER-independent target, reports demonstrate that tamoxifen binds to cannabinoid receptors (CBRs) with affinity in the low μM range and acts as an inverse agonist. To identify cannabinoids with improved pharmacological properties relative to tamoxifen, and further investigate the use of different SERM scaffolds for future cannabinoid drug development, this study characterized the affinity and activity of SERMs in newer structural classes at CBRs. Fourteen SERMs from five structurally distinct classes were screened for binding to human CBRs. Compounds from four of five SERM classes examined bound to CBRs. Subsequent studies fully characterized CBR affinity and activity of one compound from each class. Ospemifine (a triphenylethylene) selectively bound to CB1Rs, while bazedoxifine (an indole) bound to CB2Rs with highest affinity. Nafoxidine (a tetrahydronaphthalene) and raloxifene (RAL; a benzothiaphene) bound to CB1 and CB2Rs non-selectively. All four compounds acted as inverse agonists at CB1 and CB2Rs, reducing basal G-protein activity with IC50 values in the nM to low μM range. Ospemifine, bazedoxifene and RAL also acted as inverse agonists to elevate basal intracellular cAMP levels in intact CHO-hCB2 cells. The four SERMs examined also acted as CB1 and CB2R antagonists in the cAMP assay, producing rightward shifts in the concentration-effect curve of the CBR agonist CP-55,940. In conclusion, newer classes of SERMs exhibit improved pharmacological characteristics (e.g., in CBR affinity and selectivity) relative to initial studies with tamoxifen, and thus suggest that different SERM scaffolds may be useful for development of safe and selective drugs acting via CBRs.

Collaboration


Dive into the Benjamin M. Ford's collaboration.

Top Co-Authors

Avatar

Paul L. Prather

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Lirit N. Franks

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Peter A. Crooks

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

William E. Fantegrossi

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Anna Radominska-Pandya

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Narsimha Reddy Penthala

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Catheryn D. Wilson

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Jai Shankar K. Yadlapalli

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Laura P. James

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Maxim Dobretsov

University of Arkansas for Medical Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge