Skip to main content
Top
Published in: Arthritis Research & Therapy 1/2018

Open Access 01-12-2018 | Research article

A bivalent compound targeting CCR5 and the mu opioid receptor treats inflammatory arthritis pain in mice without inducing pharmacologic tolerance

Authors: Raini Dutta, Mary M. Lunzer, Jennifer L. Auger, Eyup Akgün, Philip S. Portoghese, Bryce A. Binstadt

Published in: Arthritis Research & Therapy | Issue 1/2018

Login to get access

Abstract

Background

Pain accompanies rheumatoid arthritis and other chronic inflammatory conditions and is difficult to manage. Although opioids provide potent analgesia, chronic opioid use can cause tolerance and addiction. Recent studies have demonstrated functional interactions between chemokine and opioid receptor signaling pathways. Reported heterodimerization of chemokine and opioid receptors led our group to develop bivalent compounds that bind both types of receptors, with the goal of targeting opioids to sites of inflammation. MCC22 is a novel bivalent compound containing a CCR5 antagonist and mu opioid receptor (MOR) agonist pharmacophores linked through a 22-atom spacer. We evaluated the efficacy of MCC22 in the K/B.g7 T-cell receptor transgenic mouse model of spontaneous inflammatory arthritis.

Methods

MCC22 or morphine was administered intraperitoneally at varying doses to arthritic K/B.g7 mice or nonarthritic control mice. Mechanical pain hypersensitivity was measured each day before and after drug administration, using the electronic von Frey test. The potency of MCC22 relative to that of morphine was calculated. Functional readouts of pain included grip strength and nesting behavior. A separate dosing regimen was used to determine whether the drugs induced pharmacologic tolerance.

Results

MCC22 provided ~ 3000-fold more potent analgesia than morphine in this model. Daily treatment with MCC22 also led to a cumulative analgesic effect, reducing the daily baseline pain level. MCC22 produced no observable analgesic effect in nonarthritic control mice. Importantly, repeated administration of MCC22 did not induce pharmacologic tolerance, whereas a similar regimen of morphine did. Both grip strength and nesting behaviors improved among arthritic mice treated with MCC22. Ankle thickness and arthritis scores were not affected by MCC22. The analgesic effect of MCC22 was abolished in K/B.g7 mice genetically lacking CCR5, demonstrating the receptor specificity of the antagonist pharmacophore.

Conclusions

MCC22 is a novel bivalent ligand that targets CCR5 and MOR. Our findings demonstrate that MCC22 provides highly potent analgesia and improved functional outcomes in a model of inflammatory arthritis, without inducing typical opioid tolerance. These findings suggest that MCC22 or similar compounds could be used to treat the pain associated with inflammatory arthritis and related conditions, while minimizing the risks typically associated with chronic opioid use.
Appendix
Available only for authorised users
Literature
1.
go back to reference McWilliams DF, Walsh DA. Pain mechanisms in rheumatoid arthritis. Clin Exp Rheumatol. 2017;35(Suppl 107):94–101.PubMed McWilliams DF, Walsh DA. Pain mechanisms in rheumatoid arthritis. Clin Exp Rheumatol. 2017;35(Suppl 107):94–101.PubMed
2.
go back to reference Volkow ND, McLellan AT. Opioid abuse in chronic pain—misconceptions and mitigation strategies. N Engl J Med. 2016;374:1253–63.CrossRefPubMed Volkow ND, McLellan AT. Opioid abuse in chronic pain—misconceptions and mitigation strategies. N Engl J Med. 2016;374:1253–63.CrossRefPubMed
3.
go back to reference Stein C, Kuchler S. Targeting inflammation and wound healing by opioids. Trends Pharmacol Sci. 2013;34:303–12.CrossRefPubMed Stein C, Kuchler S. Targeting inflammation and wound healing by opioids. Trends Pharmacol Sci. 2013;34:303–12.CrossRefPubMed
4.
go back to reference Melik Parsadaniantz S, Rivat C, Rostene W, Reaux-Le Goazigo A. Opioid and chemokine receptor crosstalk: a promising target for pain therapy? Nat Rev Neurosci. 2015;16:69–78.CrossRefPubMed Melik Parsadaniantz S, Rivat C, Rostene W, Reaux-Le Goazigo A. Opioid and chemokine receptor crosstalk: a promising target for pain therapy? Nat Rev Neurosci. 2015;16:69–78.CrossRefPubMed
5.
go back to reference Koch AE. Chemokines and their receptors in rheumatoid arthritis: future targets? Arthritis Rheum. 2005;52:710–21.CrossRefPubMed Koch AE. Chemokines and their receptors in rheumatoid arthritis: future targets? Arthritis Rheum. 2005;52:710–21.CrossRefPubMed
6.
go back to reference Hinks A, Martin P, Flynn E, Eyre S, Packham J, Barton A, Worthington J, Thomson W. Association of the CCR5 gene with juvenile idiopathic arthritis. Genes Immun. 2010;11:584–9.CrossRefPubMedPubMedCentral Hinks A, Martin P, Flynn E, Eyre S, Packham J, Barton A, Worthington J, Thomson W. Association of the CCR5 gene with juvenile idiopathic arthritis. Genes Immun. 2010;11:584–9.CrossRefPubMedPubMedCentral
7.
go back to reference Fleishaker DL, Garcia Meijide JA, Petrov A, Kohen MD, Wang X, Menon S, Stock TC, Mebus CA, Goodrich JM, Mayer HB, Zeiher BG. Maraviroc, a chemokine receptor-5 antagonist, fails to demonstrate efficacy in the treatment of patients with rheumatoid arthritis in a randomized, double-blind placebo-controlled trial. Arthritis Res Ther. 2012;14:R11.CrossRefPubMedPubMedCentral Fleishaker DL, Garcia Meijide JA, Petrov A, Kohen MD, Wang X, Menon S, Stock TC, Mebus CA, Goodrich JM, Mayer HB, Zeiher BG. Maraviroc, a chemokine receptor-5 antagonist, fails to demonstrate efficacy in the treatment of patients with rheumatoid arthritis in a randomized, double-blind placebo-controlled trial. Arthritis Res Ther. 2012;14:R11.CrossRefPubMedPubMedCentral
8.
go back to reference Gerlag DM, Hollis S, Layton M, Vencovsky J, Szekanecz Z, Braddock M, Tak PP. Preclinical and clinical investigation of a CCR5 antagonist, AZD5672, in patients with rheumatoid arthritis receiving methotrexate. Arthritis Rheum. 2010;62:3154–60.CrossRefPubMed Gerlag DM, Hollis S, Layton M, Vencovsky J, Szekanecz Z, Braddock M, Tak PP. Preclinical and clinical investigation of a CCR5 antagonist, AZD5672, in patients with rheumatoid arthritis receiving methotrexate. Arthritis Rheum. 2010;62:3154–60.CrossRefPubMed
9.
go back to reference van Kuijk AW, Vergunst CE, Gerlag DM, Bresnihan B, Gomez-Reino JJ, Rouzier R, Verschueren PC, van der Leij C, Maas M, Kraan MC, Tak PP. CCR5 blockade in rheumatoid arthritis: a randomised, double-blind, placebo-controlled clinical trial. Ann Rheum Dis. 2010;69:2013–6.CrossRefPubMed van Kuijk AW, Vergunst CE, Gerlag DM, Bresnihan B, Gomez-Reino JJ, Rouzier R, Verschueren PC, van der Leij C, Maas M, Kraan MC, Tak PP. CCR5 blockade in rheumatoid arthritis: a randomised, double-blind, placebo-controlled clinical trial. Ann Rheum Dis. 2010;69:2013–6.CrossRefPubMed
10.
go back to reference Chen C, Li J, Bot G, Szabo I, Rogers TJ, Liu-Chen LY. Heterodimerization and cross-desensitization between the mu-opioid receptor and the chemokine CCR5 receptor. Eur J Pharmacol. 2004;483:175–86.CrossRefPubMed Chen C, Li J, Bot G, Szabo I, Rogers TJ, Liu-Chen LY. Heterodimerization and cross-desensitization between the mu-opioid receptor and the chemokine CCR5 receptor. Eur J Pharmacol. 2004;483:175–86.CrossRefPubMed
11.
go back to reference Akgun E, Javed MI, Lunzer MM, Powers MD, Sham YY, Watanabe Y, Portoghese PS. Inhibition of inflammatory and neuropathic pain by targeting a mu opioid receptor/chemokine receptor 5 heteromer (MOR-CCR5). J Med Chem. 2015;58:8647–57.CrossRefPubMedPubMedCentral Akgun E, Javed MI, Lunzer MM, Powers MD, Sham YY, Watanabe Y, Portoghese PS. Inhibition of inflammatory and neuropathic pain by targeting a mu opioid receptor/chemokine receptor 5 heteromer (MOR-CCR5). J Med Chem. 2015;58:8647–57.CrossRefPubMedPubMedCentral
12.
go back to reference Takashima K, Miyake H, Kanzaki N, Tagawa Y, Wang X, Sugihara Y, Iizawa Y, Baba M. Highly potent inhibition of human immunodeficiency virus type 1 replication by TAK-220, an orally bioavailable small-molecule CCR5 antagonist. Antimicrob Agents Chemother. 2005;49:3474–82.CrossRefPubMedPubMedCentral Takashima K, Miyake H, Kanzaki N, Tagawa Y, Wang X, Sugihara Y, Iizawa Y, Baba M. Highly potent inhibition of human immunodeficiency virus type 1 replication by TAK-220, an orally bioavailable small-molecule CCR5 antagonist. Antimicrob Agents Chemother. 2005;49:3474–82.CrossRefPubMedPubMedCentral
13.
go back to reference Rothman RB, Long JB, Bykov V, Jacobson AE, Rice KC, Holaday JW. Beta-FNA binds irreversibly to the opiate receptor complex: in vivo and in vitro evidence. J Pharmacol Exp Ther. 1988;247:405–16.PubMed Rothman RB, Long JB, Bykov V, Jacobson AE, Rice KC, Holaday JW. Beta-FNA binds irreversibly to the opiate receptor complex: in vivo and in vitro evidence. J Pharmacol Exp Ther. 1988;247:405–16.PubMed
14.
go back to reference Akgun E, Javed MI, Lunzer MM, Smeester BA, Beitz AJ, Portoghese PS. Ligands that interact with putative MOR-mGluR5 heteromer in mice with inflammatory pain produce potent antinociception. Proc Natl Acad Sci U S A. 2013;110:11595–9.CrossRefPubMedPubMedCentral Akgun E, Javed MI, Lunzer MM, Smeester BA, Beitz AJ, Portoghese PS. Ligands that interact with putative MOR-mGluR5 heteromer in mice with inflammatory pain produce potent antinociception. Proc Natl Acad Sci U S A. 2013;110:11595–9.CrossRefPubMedPubMedCentral
15.
go back to reference Monach PA, Mathis D, Benoist C. The K/BxN arthritis model. Curr Protoc Immunol. 2008;Chapter 15(Unit 15):22.PubMed Monach PA, Mathis D, Benoist C. The K/BxN arthritis model. Curr Protoc Immunol. 2008;Chapter 15(Unit 15):22.PubMed
16.
go back to reference Kuziel WA, Dawson TC, Quinones M, Garavito E, Chenaux G, Ahuja SS, Reddick RL, Maeda N. CCR5 deficiency is not protective in the early stages of atherogenesis in apoE knockout mice. Atherosclerosis. 2003;167:25–32.CrossRefPubMed Kuziel WA, Dawson TC, Quinones M, Garavito E, Chenaux G, Ahuja SS, Reddick RL, Maeda N. CCR5 deficiency is not protective in the early stages of atherogenesis in apoE knockout mice. Atherosclerosis. 2003;167:25–32.CrossRefPubMed
17.
go back to reference Auger JL, Haasken SS, Binstadt BA. Autoantibody-mediated arthritis in the absence of C3 and activating Fcgamma receptors: C5 is activated by the coagulation cascade. Arthritis Res Ther. 2012;14:R269.CrossRefPubMedPubMedCentral Auger JL, Haasken SS, Binstadt BA. Autoantibody-mediated arthritis in the absence of C3 and activating Fcgamma receptors: C5 is activated by the coagulation cascade. Arthritis Res Ther. 2012;14:R269.CrossRefPubMedPubMedCentral
18.
go back to reference Haasken S, Auger JL, Binstadt BA. Absence of beta2 integrins impairs regulatory T cells and exacerbates CD4+ T cell-dependent autoimmune carditis. J Immunol. 2011;187:2702–10.CrossRefPubMedPubMedCentral Haasken S, Auger JL, Binstadt BA. Absence of beta2 integrins impairs regulatory T cells and exacerbates CD4+ T cell-dependent autoimmune carditis. J Immunol. 2011;187:2702–10.CrossRefPubMedPubMedCentral
19.
go back to reference Gaskill BN, Karas AZ, Garner JP, Pritchett-Corning KR. Nest building as an indicator of health and welfare in laboratory mice. J Vis Exp. 2013;82:51012. Gaskill BN, Karas AZ, Garner JP, Pritchett-Corning KR. Nest building as an indicator of health and welfare in laboratory mice. J Vis Exp. 2013;82:51012.
20.
go back to reference Caplazi P, Diehl L. Histopathology in mouse models of rheumatoid arthritis. In: Potts SJ, Eberhard DA, Wharton KA, editors. Molecular histopathology and tissue biomarkers in drug and diagnostic development. New York: Springer; 2015. p. 65–78. Caplazi P, Diehl L. Histopathology in mouse models of rheumatoid arthritis. In: Potts SJ, Eberhard DA, Wharton KA, editors. Molecular histopathology and tissue biomarkers in drug and diagnostic development. New York: Springer; 2015. p. 65–78.
21.
go back to reference Rottman JB, Ganley KP, Williams K, Wu L, Mackay CR, Ringler DJ. Cellular localization of the chemokine receptor CCR5. Correlation to cellular targets of HIV-1 infection. Am J Pathol. 1997;151:1341–51.PubMedPubMedCentral Rottman JB, Ganley KP, Williams K, Wu L, Mackay CR, Ringler DJ. Cellular localization of the chemokine receptor CCR5. Correlation to cellular targets of HIV-1 infection. Am J Pathol. 1997;151:1341–51.PubMedPubMedCentral
22.
go back to reference Abbadie C, Bhangoo S, De Koninck Y, Malcangio M, Melik-Parsadaniantz S, White FA. Chemokines and pain mechanisms. Brain Res Rev. 2009;60:125–34.CrossRefPubMed Abbadie C, Bhangoo S, De Koninck Y, Malcangio M, Melik-Parsadaniantz S, White FA. Chemokines and pain mechanisms. Brain Res Rev. 2009;60:125–34.CrossRefPubMed
23.
go back to reference Jacobs JP, Ortiz-Lopez A, Campbell JJ, Gerard CJ, Mathis D, Benoist C. Deficiency of CXCR2, but not other chemokine receptors, attenuates autoantibody-mediated arthritis in a murine model. Arthritis Rheum. 2010;62:1921–32.PubMedPubMedCentral Jacobs JP, Ortiz-Lopez A, Campbell JJ, Gerard CJ, Mathis D, Benoist C. Deficiency of CXCR2, but not other chemokine receptors, attenuates autoantibody-mediated arthritis in a murine model. Arthritis Rheum. 2010;62:1921–32.PubMedPubMedCentral
24.
go back to reference Aceto MD, Harris LS, Negus SS, Banks ML, Hughes LD, Akgun E, Portoghese PS. MDAN-21: a bivalent opioid ligand containing mu-agonist and delta-antagonist pharmacophores and its effects in rhesus monkeys. Int J Med Chem. 2012;2012:327257.PubMedPubMedCentral Aceto MD, Harris LS, Negus SS, Banks ML, Hughes LD, Akgun E, Portoghese PS. MDAN-21: a bivalent opioid ligand containing mu-agonist and delta-antagonist pharmacophores and its effects in rhesus monkeys. Int J Med Chem. 2012;2012:327257.PubMedPubMedCentral
25.
go back to reference Le Naour M, Akgun E, Yekkirala A, Lunzer MM, Powers MD, Kalyuzhny AE, Portoghese PS. Bivalent ligands that target mu opioid (MOP) and cannabinoid1 (CB1) receptors are potent analgesics devoid of tolerance. J Med Chem. 2013;56:5505–13.CrossRefPubMed Le Naour M, Akgun E, Yekkirala A, Lunzer MM, Powers MD, Kalyuzhny AE, Portoghese PS. Bivalent ligands that target mu opioid (MOP) and cannabinoid1 (CB1) receptors are potent analgesics devoid of tolerance. J Med Chem. 2013;56:5505–13.CrossRefPubMed
26.
go back to reference Spahn V, Del Vecchio G, Labuz D, Rodriguez-Gaztelumendi A, Massaly N, Temp J, Durmaz V, Sabri P, Reidelbach M, Machelska H, Weber M, Stein C. A nontoxic pain killer designed by modeling of pathological receptor conformations. Science. 2017;355:966–9.CrossRefPubMed Spahn V, Del Vecchio G, Labuz D, Rodriguez-Gaztelumendi A, Massaly N, Temp J, Durmaz V, Sabri P, Reidelbach M, Machelska H, Weber M, Stein C. A nontoxic pain killer designed by modeling of pathological receptor conformations. Science. 2017;355:966–9.CrossRefPubMed
Metadata
Title
A bivalent compound targeting CCR5 and the mu opioid receptor treats inflammatory arthritis pain in mice without inducing pharmacologic tolerance
Authors
Raini Dutta
Mary M. Lunzer
Jennifer L. Auger
Eyup Akgün
Philip S. Portoghese
Bryce A. Binstadt
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Arthritis Research & Therapy / Issue 1/2018
Electronic ISSN: 1478-6362
DOI
https://doi.org/10.1186/s13075-018-1661-5

Other articles of this Issue 1/2018

Arthritis Research & Therapy 1/2018 Go to the issue