throbber
Bioorganic & Medicinal Chemistry Letters 16 (2006) 691–694
`
`Derivatives of tramadol for increased duration of effect
`
`Liming Shao,* Craig Abolin, Michael C. Hewitt, Patrick Koch and Mark Varney
`
`Sepracor Inc., 84 Waterford Drive, Marlborough, MA 01752, USA
`
`Received 23 August 2005; revised 6 October 2005; accepted 7 October 2005
`Available online 27 October 2005
`
`Abstract—Tramadol is a centrally acting opioid analgesic structurally related to codeine and morphine. Analogs of tramadol with
`deuterium-for-hydrogen replacement at metabolically active sites were prepared and evaluated in vitro and in vivo.
`Ó 2005 Elsevier Ltd. All rights reserved.
`
`Tramadol (±)-cis is used for the treatment of moderate
`to moderately severe pain.1 Tramadol has a relatively
`short duration of analgesic effect due to extensive first
`pass metabolism and as such is dosed as frequently as
`100 mg every 4–6 h.2 The analgesic activity of tramadol
`is thought to be the result of a dual mechanism—the
`parent (1) acts as an inhibitor of norepinephrine and
`serotonin reuptake, and the major O-desmethyl metabo-
`lite (M1) is a potent agonist at l-opioid receptors
`(Fig. 1).3
`
`The opioid antagonist naloxone only partially inhibits
`the analgesic activity of tramadol in animal tests. Not
`unlike other opioids, tramadol causes a number of side
`effects including constipation, nausea, dizziness, and
`somnolence. A tramadol analog that had a longer
`half-life (and therefore required less-frequent dosing)
`and was devoid of opioid side effects would have thera-
`peutic benefit. Herein, we wish to disclose our efforts to
`
`develop an improved tramadol analog with a longer
`half-life.
`
`Our approach to tramadol analogs took advantage of
`the primary kinetic isotope effect4 to slow CYP450-med-
`iated metabolism (Fig. 2). Replacing hydrogen with deu-
`terium at metabolically active sites can result in a slower
`metabolism due to the reduced rate of cleavage of a C–D
`bond relative to a C–H bond. This approach has been
`shown to be effective for a number of pharmacological
`agents including amphetamine,5 butethal,6 and mor-
`phine.7 For tramadol, formation of metabolite M1 is
`both the primary route of metabolism and is responsible
`for its opioid-like effects. By slowing down metabolism
`of tramadol, it was anticipated that the formation of
`M1 would be reduced, providing a longer-acting drug
`with potentially fewer opioid-related side effects. To-
`ward that end, the hydrogen atoms on the O-methyl
`and N-methyl groups were replaced by deuterium as
`shown in structures D3, D6, D9, and D6 M1.
`
`OCH3
`
`OH
`
`OH
`
`CH3
`N
`CH3
`
`OH
`
`CH3
`N
`CH3
`
`1
`
`M1
`
`Figure 1. (±)-Tramadol 1 and metabolite (±)-M1.
`
`Keywords: Tramadol; Metabolic stability; Deuterium isotope effect;
`Opioids.
`* Corresponding author. Tel.: +1 5083577467; fax: +1 5083577467;
`e-mail: liming.shao@sepracor.com
`
`0960-894X/$ - see front matter Ó 2005 Elsevier Ltd. All rights reserved.
`doi:10.1016/j.bmcl.2005.10.024
`
`OR1
`
`OH
`
`R2
`
`N
`R2
`
`D3 R1 = CD3, R2 = CH3
`D6 R1 = CH3, R2 = CD3
`D6 M1 R1 = H, R2 = CD3
`D9 R1 = CD3, R2 = CD3
`Dramadol analogs
`
`Figure 2. D-tramadol (dramadol) analogs.
`
`Auspex Exhibit 2011
`Apotex v. Auspex
`IPR2021-01507
`Page 1
`
`

`

`692
`
`L. Shao et al. / Bioorg. Med. Chem. Lett. 16 (2006) 691–694
`
`R1
`
`1. Mg, THF, 4 or 5
`2. HCl, Et2O
`
`OR2
`
`O
`
`N
`R1
`2 R1 = CH3
`3 R1 = CD3
`
`OR2
`
`Br
`4 R2 = CH3
`5 R2 = CD3
`
`1. n-BuLi, PHPh2, THF
`2. HCl, Et2O
`
`HCl
`
`OH
`
`R1
`
`N
`R1
`
`6 (D3) R1 = CH3, R2 = CD3
`7 (D6) R1 = CD3, R2 = CH3
`
`OH
`
`OH
`
`R1
`
`N
`R1
`
`HCl
`
`8 (D6 M1) R1 = CD3
`
`Scheme 1. Synthesis of D3, D6, and D6 M1 metabolites.
`
`CD3
`N
`CD3
`
`1. t-BuLi, Et2O, 5
`2. HCl, Et2O
`OCD3
`
`3
`
`OCD3
`
`OH
`
`HCl
`
`CD
`N
`CD3
`
`Br
`
`9 (D9)
`
`Scheme 2. Synthesis of 9 (D9).
`
`the deuterated tramadol analogs
`The syntheses of
`were relatively straightforward and began with Man-
`nich base 2 or 3 (Scheme 1). Specifically, addition of
`the Grignard reagent 4 or 5 to ketones 2 or 3 provid-
`ed crude 6 (D3) and 7 (D6) as mixtures of diastereo-
`mers (76% cis for D6). The abundance of the cis
`isomer could be improved to 93% after formation of
`the HCl salt and multiple recrystallizations from iso-
`propanol.8 Demethylation of 7 (D6) occurred without
`incident to provide phenol 8 (D6 M1).9 Purification
`was effected via formation of the HCl salt and recrys-
`tallization from isopropanol/ethyl acetate to give the
`phenol salt as an analytically pure white powder.
`The reaction to form D9 was attempted several times
`with starting materials 3 and 5, with little product for-
`
`mation seen by LCMS and HPLC analysis. This led
`to an alternate strategy to synthesize D9 that relied
`on a lithium–halogen exchange reaction that had been
`outlined previously in the literature,10 and was suc-
`cessfully employed using Mannich base 3 and aromat-
`ic bromide 5 (Scheme 2).
`
`Crude 9 (D9) was also converted to its HCl salt and
`recrystallized several times in isopropanol to provide a
`93% pure cis isomer.11
`
`The receptor binding affinity and functional monoamine
`uptake inhibition by tramadol and the deuterated tram-
`adol analogs are shown in Table 1.
`
`As expected, the deuterated analog of M1 retained the
`l-opioid receptor binding affinity present in metabolite
`M1. In general, the deuterated analogs retained in vitro
`activity comparable to their non-deuterated parent mol-
`ecules, with a slightly diminished reuptake inhibition of
`norepinephrine compared to tramadol.
`
`The in vitro metabolic stability results for the deuterated
`tramadol compounds are detailed in Table 2.12,13 Based
`on disappearance of parent compound in incubation
`media, the most stable compound of the six tested in hu-
`man liver microsomes and hepatocytes was clearly the
`D9-tramadol derivative 9. Tramadol and D6-tramadol
`
`Table 1. l-Opioid binding, and 5-HT and NE reuptake inhibition of (±)-cis tramadol and deuterated derivatives
`OR2
`
`Compound
`
`1 Tramadol
`Metabolite M1
`6 (D3)
`7 (D6)
`8 (D6 M1)
`9 (D9)
`
`R1
`
`CH3
`CH3
`CH3
`CD3
`CD3
`CD3
`
`R2
`
`CH3
`H
`CD3
`CH3
`H
`CD3
`
`OR3
`
`R1
`
`N
`R1
`
`R3
`
`H
`H
`H
`H
`H
`H
`
`l
`
`7600
`47
`>10,000
`>10,000
`43
`5300
`
`IC50 (nM)
`
`5-HT
`
`4300
`4600
`1900
`3100
`9900
`1100
`
`NE
`
`790
`>10,000
`3600
`3200
`6700
`2000
`
`Note: IC50 is the concentration required to inhibit 50% binding to l-opioid receptors or reuptake of 5-hydroxytryptamine (5-HT) and norepinephrine
`(NE).
`
`Auspex Exhibit 2011
`Apotex v. Auspex
`IPR2021-01507
`Page 2
`
`

`

`L. Shao et al. / Bioorg. Med. Chem. Lett. 16 (2006) 691–694
`
`693
`
`hepatocytes and microsomes in Figures 3 and 4, respec-
`tively. Consistent with a deuterium isotope effect on the
`metabolically labile O-methyl group, 6 (D3) and 9 (D9)
`slowed the formation of the primary metabolite M1 by
`approximately 5-fold. The formation rate of metabolite
`M1 from 7 (D6) was nearly indistinguishable from that
`of tramadol in human hepatocytes.
`
`The in vivo activities of tramadol (1), 7 (D6), and 9 (D9)
`were tested in the rat tail-flick model. Tramadol (1), 7
`(D6), and 9 (D9) were administered at a dose of
`50 mg/kg intraperitoneally. Intraperitoneal morphine
`(5 mg/kg) and saline were used as controls. Analgesic
`activity (tail-flick latency) was evaluated at 1, 2, 4, and
`8 h after the dose. The results of efficacy (tail-flick laten-
`cy) are shown in Figure 5.
`
`Tramadol and two of its deuterated derivatives (7 (D6)
`and 9 (D9)) significantly lengthened tail-flick latency
`when tested at 1 and 2 h after intraperitoneal adminis-
`tration of 50 mg/kg. Tramadol and 9 (D9) were also
`effective when tested 4 h after dosing. By 8 h the effects
`of the test compounds were no longer statistically signif-
`icant. As expected, morphine increased tail-flick latency
`at the 1 h time point, but at later times the effect was not
`significantly greater than that of vehicle. The results
`indicate that the test compounds effectively reduced
`acute nociception for 2–4 h following a single acute
`intraperitoneal dose.
`
`In conclusion, the substitution of deuterium for hydro-
`gen in the methyl groups of tramadol did not adversely
`affect the in vitro binding affinity. In vitro testing in hu-
`man microsomes confirmed that replacing hydrogen
`with deuterium in the metabolically labile O-methyl po-
`sition of 1 slowed the formation of the primary metabo-
`lite M1 by approximately 5-fold. The deuterated
`derivatives 7 (D6) and 9 (D9) were active analgesics in
`the rat tail-flick model, but were not superior to trama-
`dol in terms of potency or duration of effect. Deuterium
`
`**
`
`**
`
`**
`
`**
`
`****
`
`**
`
`**
`
`*
`
`60
`
`120
`
`240
`
`Minutes
`D6
`Tramadol
`Morphine Control
`Saline Control
`Dunnett's test: *, **/ p<0.05, p<0.01 (n = 9 - 10 per group)
`
`480
`
`D9
`
`25
`
`20
`
`15
`
`10
`
`05
`
`Latency (s)
`
`Table 2. Summary of in vitro metabolic stability of tramadol and
`deuterated derivatives
`
`Compound
`
`Half-lifea (min)/rankb
`HLMc
`
`Hepatocytes
`
`9 (D9), (+/)-cis
`7210/1
`3610/1
`8 (M1 D6), (+/)-cis
`747/5
`1650/2
`Metabolite M1, (+/)-cis
`2820/2
`1560/3
`6 (D3), (+/)-cis
`744/4
`1330/4
`1 Tramadol, (+/)-cis
`741/3
`968/5
`7 (D6), (+/)-cis
`547/6
`852/6
`a Calculated from samples taken at 0, 180, and 360 min for hepatocytes
`and 0, 90, and 180 min for microsomes.
`b Ranked from 1, the most stable to 6, the least stable.
`c HLM = human liver microsomes.
`
`7 were the least stable compounds in human liver micro-
`somes and hepatocytes.
`
`Metabolic stability was also evaluated based on the for-
`mation rate of metabolite M1 from tramadol (1) and
`three tramadol deuterated derivatives capable of pro-
`ducing metabolite M1. The results are summarized for
`
`D6 Tramadol
`
`D3 Tramadol
`
`D9 Tramadol
`
`180
`
`Incubation Time (min)
`
`360
`
`100
`
`80
`
`60
`
`40
`
`20
`
`0
`
`M1 Formation Rate (% Control)
`
`Figure 3. Stability of tramadol analogs in human hepatocytes.
`
`D6 Tramadol
`
`D3 Tramadol
`
`D9 Tramadol
`
`90
`
`Incubation Time (min)
`
`180
`
`100
`
`80
`
`60
`
`40
`
`20
`
`M1 Formation Rate (% Control)
`
`Figure 4. Stability of tramadol analogs in human liver microsomes.
`
`Figure 5. Effect of test compounds on tail-flick latency.
`
`Auspex Exhibit 2011
`Apotex v. Auspex
`IPR2021-01507
`Page 3
`
`

`

`694
`
`L. Shao et al. / Bioorg. Med. Chem. Lett. 16 (2006) 691–694
`
`for hydrogen replacement at metabolically active sites
`had no obvious deleterious effects in vivo but did not
`result
`in a longer duration of effect. In this case,
`deuteration at metabolically active sites produced a
`pharmacological
`agent
`equipotent
`in vivo with
`tramadol.
`
`References and notes
`
`1. (a) Raffa, R. B.; Friderichs, E.; Reimann, W.; Shank, R.
`P.; Good, E. E.; Vaught, J. L. J. Pharmacol. Exp. Ther.
`1992, 260, 275; (b) Gibson, T. P. Am. J. Med. 1996,
`101(Suppl 1A), 47S; (c) Radbruch, L.; Grond, S.; Leh-
`mann, K. Drug Safety 1996, 15, 8; (d) Raffa, R. B. Am. J.
`Med. 1996, 101(Suppl 1A), 41S; (e) Garrido, M.; Valle,
`M.; Campanero, M. A.; Calvo, R.; Troconiz, I. F. J.
`Pharmacol. Exp. Ther. 2000, 295, 352; (f) Subrahmanyam,
`V.; Renwick, A. B.; Walters, D. G.; Young, P. J.; Price, R.
`J.; Tonelli, A. P.; Lake, B. G. Drug Metab. Dispos. 2001,
`29, 1146; (g) Wu, W. N.; McKown, L. A.; Liao, S.
`Xenobiotica 2002, 32, 411; (h) Leppert, W.; Luczak, J.
`Support Care Cancer 2005, 13, 5; (i) Duhmke, R. M.;
`Cornblath, D. D.; Hollingshead, J. R. Cochrane Database
`Syst. Rev. 2004, 2, CD003726; (j) Grond, S.; Sablotzki, A.
`Clin. Pharmacol. Ther. 2004, 43, 879;
`(k) Klotz, U.
`Arzneim.-Forsch. 2003, 53, 681.
`2. PDR Electronic Library, Thomson Scientific, http://
`www.thomsonhc.com/pdrel/librarian,
`reference
`for
`UltramÒ tablets (Ortho-McNeil).
`3. Raffa, R. B.; Nayak, R. K.; Liao, S.; Minn, F. L. Rev.
`Contemp. Pharmacother. 1995, 6, 485.
`4. (a) Wilberg, K. B. Chem. Rev. 1955, 55, 713;
`Westheimer, F. H. Chem. Rev. 1961, 61, 265.
`5. Parli, C. J.; McMahon, R. E. Drug Metab. Dispos. 1973, 1,
`337.
`6. Tanabe, M.; Yasuda, D.; LeValley, S.; Mitoma, C. Life
`Sci. 1969, 8, 1123.
`7. Elison, C.; Rapoport, H.; Laursen, R.; Elliott, H. W.
`Science 1961, 134, 1078.
`8. NMR data for 16 (D3): 1H NMR (400 MHz, CD3OD) 7.36–
`7.31 (m, 1H), 7.16–7.11 (m, 2H), 6.85–6.83 (m, 1H), 3.04–
`2.97 (m, 1H), 2.74–2.64 (m, 7H), 2.34–2.26 (m, 1H), 1.97–
`1.60 (m, 8H); 13C NMR (100 MHz, CD3OD) 161.3, 150.5,
`130.6, 118.2, 113.0, 112.2, 75.8, 61.8, 46.2, 42.8, 42.6, 41.6,
`27.3, 26.0, 22.4; mp: 180–184 °C; TLC (10% MeOH in
`CH2Cl2) Rf = 0.48. NMR data for 17 (D6): 1H NMR
`(400 MHz, CD3OD) 7.30–7.27 (m, 1H), 7.10–7.05 (m, 2H),
`6.83–6.80 (m, 1H), 3.79 (s, 3H), 2.98–2.93 (m, 1H), 2.66–
`2.62 (m, 1H), 2.25–2.20 (m, 1H), 1.94–1.51 (m, 8H); 13C
`NMR (100 MHz, CD3OD) 162.3, 151.4, 131.6, 119.1, 113.8,
`113.2, 76.7, 62.6, 56.5, 43.8, 42.4, 27.9, 27.0, 23.3; mp: 175–
`180 °C; TLC (10% MeOH in CH2Cl2) Rf = 0.43.
`
`(b)
`
`9. NMR data for 18 (D6 M1): 1H NMR (400 MHz, CD3OD)
`7.14–7.11 (m, 1H), 6.90–6.88 (m, 2H), 6.62–6.60 (m, 1H),
`2.91–2.85 (m, 1H), 2.61 (dd, J = 2.57, 13.2 Hz, 1H), 2.13–
`2.07 (m, 1H), 1.94–1.43 (m, 8H); 13C NMR (100 MHz,
`CD3OD) 158.8, 150.5, 130.6, 117.1, 114.7, 113.2, 75.8,
`61.8, 43.1, 41.5, 27.1, 26.2, 22.4; mp: 221–226 °C; TLC
`(20% MeOH in CH2Cl2) Rf = 0.23.
`10. Draper, R. W.; Hou, D.; Iyer, R.; Lee, G. M.; Liang, J. T.;
`Mas, J. L.; Vater, E. J. Org. Process Res. Dev. 1998, 2, 186.
`11. NMR data for 19 (D9): 1H NMR (400 MHz, CD3OD)
`7.27–7.24 (m, 1H), 7.09–7.04 (m, 2H), 6.80 (d, J = 8.07 Hz,
`1H), 2.94 (dd, J = 9.16, 13.2 Hz, 1H), 2.21–2.16 (m, 1H),
`1.95–1.50 (m, 8H); 13C NMR (100 MHz, CD3OD) 161.0,
`150.1, 130.5, 117.9, 112.7, 112.0, 75.6, 61.5, 42.8, 41.3,
`27.0, 25.9, 22.1; mp: 176–182 °C; TLC (10% MeOH in
`CH2Cl2) Rf = 0.45.
`12. General procedure for microsomal stability assay in
`Human liver microsomes: The test compounds at 23 lM
`were incubated separately with human liver microsomes
`(4 mg protein/mL) at 37 ± 1 °C in 3-mL incubation
`mixtures
`containing
`potassium phosphate
`buffer
`(50 mM, pH 7.4), MgCl2 (3 mM), and EDTA (1 mM).
`Reactions were started by the addition of the NADPH-
`generating system. At designated times (0, 90, and
`180 min), a 500-lL aliquot was
`removed from the
`incubation and added to 500 lL acetonitrile to terminate
`the reaction. The amount of unchanged test compound
`was quantified by LC/MS/MS. Where appropriate, the
`amount of O-desmethyl metabolite formed was also
`assessed by LC/MS/MS.
`13. General procedure for in vitro stability assay in human
`hepatocytes. Test compounds (23 lM) were incubated
`separately with a pool (n = 2) of cryopreserved human
`hepatocytes (2 million cells/mL) in WaymouthÕs+ (Way-
`mouthÕs medium [without phenol
`red]
`supplemented
`(5.6 lg/mL),
`with FBS
`(4.5%),
`insulin
`glutamine
`(3.6 mM), sodium pyruvate (4.5 mM), and dexametha-
`sone (0.9 lM)) at
`the final concentrations indicated.
`Each test article was added to incubations in 2.5 lL
`(1%) of methanol. Reactions were started when placed
`in the incubator. At designated times (0, 180, and
`360 min), a 250-lL aliquot was
`removed from the
`incubation and added to 250 lL acetonitrile to terminate
`the reaction. Precipitated protein was
`removed by
`centrifugation (920g for 10 min at 10 °C). Amount of
`unchanged parent compound was quantified as follows;
`(20 lL) of
`an aliquot
`the supernatant
`fraction was
`transferred to 1 mL of
`internal
`standard (50 ng/mL
`dextrorphan in 1:1 methanol/water, 51-fold dilution) and
`analyzed by LC/MS/MS. Additional
`incubations were
`performed with hepatocytes in which the test article was
`replaced with 7-ethoxycoumarin (500 lM, marker sub-
`strate) to determine if the hepatocytes were metaboli-
`cally competent.
`
`Auspex Exhibit 2011
`Apotex v. Auspex
`IPR2021-01507
`Page 4
`
`

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket