US20020111499A1 - Novel intermediate for the synthesis of prostaglandins - Google Patents

Novel intermediate for the synthesis of prostaglandins Download PDF

Info

Publication number
US20020111499A1
US20020111499A1 US09/898,241 US89824101A US2002111499A1 US 20020111499 A1 US20020111499 A1 US 20020111499A1 US 89824101 A US89824101 A US 89824101A US 2002111499 A1 US2002111499 A1 US 2002111499A1
Authority
US
United States
Prior art keywords
solution
synthesis
added
lactone
minutes
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US09/898,241
Other versions
US6437152B1 (en
Inventor
Philip Jackson
Ian Lennon
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GBGB9908326.3A external-priority patent/GB9908326D0/en
Application filed by Individual filed Critical Individual
Priority to US09/898,241 priority Critical patent/US6437152B1/en
Publication of US20020111499A1 publication Critical patent/US20020111499A1/en
Priority to US10/224,250 priority patent/US20030004368A1/en
Application granted granted Critical
Publication of US6437152B1 publication Critical patent/US6437152B1/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C405/00Compounds containing a five-membered ring having two side-chains in ortho position to each other, and having oxygen atoms directly attached to the ring in ortho position to one of the side-chains, one side-chain containing, not directly attached to the ring, a carbon atom having three bonds to hetero atoms with at the most one bond to halogen, and the other side-chain having oxygen atoms attached in gamma-position to the ring, e.g. prostaglandins ; Analogues or derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F7/00Compounds containing elements of Groups 4 or 14 of the Periodic System
    • C07F7/02Silicon compounds
    • C07F7/08Compounds having one or more C—Si linkages
    • C07F7/18Compounds having one or more C—Si linkages as well as one or more C—O—Si linkages
    • C07F7/1804Compounds having Si-O-C linkages

Abstract

An enantiomerically enriched compound of formula 1,
Figure US20020111499A1-20020815-C00001
wherein Ar is phenyl optionally substituted with one or more groups selected from haloalkyl, alkyl and halide. This compound can be isolated in crystalline form, and used in the preparation of (+)-16-[3-trifluoromethyl)phenoxy]-17,18,19,20-tetranor PGF isopropyl ester.

Description

    FIELD OF THE INVENTION
  • This invention relates to the preparation of a novel crystalline lactone and its use in prostaglandin synthesis. [0001]
  • BACKGROUND OF THE INVENTION
  • The synthetic prostaglandin 16-[3-(trifluoromethyl)phenoxy]-17,18,19,20-tetranor PGF[0002] and its ester derivatives, in particular the isopropyl ester (2), are potent drugs for the treatment of glaucoma and ocular hypertension. Optimum therapeutic benefit is achieved when compound (2) is used in the form of the dextrorotatory single enantiomer (+)-2, as depicted below. For development as a pharmaceutical product, an economically viable route is required for the synthesis of (+)-2 in quantities of at least 1 kg.
    Figure US20020111499A1-20020815-C00002
  • EP-A-0639563 describes biological studies of compound 2 and analogues, and synthetic methods which are applicable to its preparation. The synthetic strategy employed is based on that used by Corey in his pioneering synthesis of prostaglandin F[0003] (Corey and Cheng, The Logic of Chemical Synthesis, Wiley, 1989, p. 250-266 and references therein), wherein the cyclopentane ring embedded in a lactone intermediate of type 3 (PG=protecting group, e.g. Me) has relative stereochemistry correctly defined across four chiral centres. Lactones of type 3 can be prepared in single enantiomer form. Although such a route was successfully utilised to prepare small quantities of (+)-2 for preliminary biological evaluation, for a number of reasons it is unsuitable for industrial manufacture as a high-purity pharmaceutical product for administration to human patients. At least 15 steps (from cyclopentadiene) are required, with loss of yield in individual steps exacerbated by the linear nature of the synthesis. Fractional column chromatography is required after many of these steps to effect purification of intermediates. For example, a late stage stereoselective reduction of a 15-keto function in the (ω-side chain using (−)-B-chlorodiisopinocampheylborane requires the removal of the unwanted 15S isomer, formed as a by-product, by a chromatographic separation.
    Figure US20020111499A1-20020815-C00003
  • An alternative and more convergent approach to prostaglandins involves addition of a cuprate reagent, incorporating the entire ω-side chain, to the tricyclo[3,2,0,0[0004] 2,7]heptanone 4 (Lee et al, J. Chem. Soc., Perkin Trans 1, 1978, 1176). Tricyclic ketone 4 is prepared from a TBDMS-protected bromohydrin which is in turn derived from bicyclo[3,2,0]hept-2-en-6-one (5). In comparison to routes proceeding by a Corey lactone of type 3, significantly fewer steps are required. For example, preparation of prostaglandin F from compound 5 requires only 8 steps (10 steps from cyclopentadiene). Avoidance of awkward late-stage reduction to establish the required configuration of the C—15—OH functionality provides another advantage.
  • EP-A-0074856 describes resolution of racemic bicyclo[3,2,0]hept-2-en-6-one (5) by forming diastereomeric salts of its α-hydroxysulfonic acid derivative with a chiral amine, and separation by crystallisation. [0005]
  • Certain lactones are described in Newton et al, Tetrahedron, 1980, 2163. None is crystalline. [0006]
  • SUMMARY OF THE INVENTION
  • This invention is based on the unexpected discovery of a crystalline lactone 1 [0007]
    Figure US20020111499A1-20020815-C00004
  • This compound can be used in the stereoselective synthesis of 16-[3-(trifluoromethyl)phenoxy]-17,18,19,20-tetranor PGF[0008] and its ester derivatives, for example (+)-2. The crystalline lactone can be obtained in highly pure form. The crystallinity of the lactone is crucial in enabling impurities to be removed at this stage without resort to column chromatography. This provides the basis for an industrially viable synthesis of the prostaglandin 2 for pharmaceutical use.
  • According to the invention, this discovery may also be applied to other substituted 16-phenoxy prostaglandins, where the substituent is haloalkyl, alkyl or halide. The alkyl group may have up to 6 C atoms. Halide is preferably 3-Cl. [0009]
  • DESCRIPTION OF THE INVENTION
  • By way of illustration, the synthesis of the lactone 1 is depicted in Scheme I. All reactants depicted are used in enantiomerically enriched form, typically in >95% ee or higher. [0010]
  • Step (i) is the preparation of the tricyclic ketone 4. This is achieved by treatment of the bromohydrin 6 with base in an appropriate solvent, preferably potassium tert-butoxide in toluene. The unstable tricycle 4 is used without purification in step (iii). It is not necessary to evaporate the tricycle solution to dryness. [0011]
  • Step (ii) is the formation of an alkenylcuprate reagent from the vinyl iodide 7, precursor to the ω-side chain. The preparation of vinyl iodide 7 in enantiomerically enriched form is disclosed in the copending patent application, filed on the same date, entitled Process for the Preparation of Prostaglandin Precursors, and claiming priority from British Patent Application No. 9908327.1. The vinyl iodide is metallated with an alkyllithium reagent, preferably tert-butyllithium, and then treated with a cuprate of the form RCu(CN)Li where R is a non-transferable group which may be 2-thienyl. Step (iii) is reaction of the alkenylcuprate with the tricycle to form the bicyclic ketone 8. [0012]
  • Step (iv) is the Baeyer-Villiger reaction producing the lactone 1. A peracid, preferably peracetic acid, is used, resulting in a 3:1 mixture of regioisomers, isolated as an oil. Further processing is then required to render this material as usable in subsequent steps. Conveniently, the minor and unwanted regioisomer can be selectively hydrolysed by treatment with aqueous alkali, for example, aqueous sodium hydroxide in acetonitrile. Extraction of the unreacted lactone 1 into an organic solvent, followed by evaporation of solvent, yields a solid residue which can be recrystallised at low temperature to give highly pure crystalline material with convenient handling and storage characteristics. These processing operations are pivotal to the success of the overall synthetic route. [0013]
    Figure US20020111499A1-20020815-C00005
  • Scheme II summarises the conversion of lactone 1 to the target prostaglandin (+)-2, using conventional processes (for analogous methods see Lee et al, [0014] J. Chem. Soc., Perkin Trans 1, 1978, 1176; and EP-A-0639563). Typically, reduction to the lactol using diisobutylaluminium hydride is followed by Wittig reaction with the ylide generated from (4-carboxybutyl)triphenylphosphonium bromide and potassium tert-butoxide. Esterification and O-deprotection steps complete the synthesis.
    Figure US20020111499A1-20020815-C00006
  • The following Examples illustrate the invention.[0015]
  • EXAMPLE 1 3-endo-tert-Butyldimethylsilyloxytricyclo[3,2,0,02,7]heptan-6-one
  • Potassium tert-butoxide (114.2 g, 1.02 mol) is suspended in toluene (2 L) and cooled to −15° C. The suspension is stirred under a nitrogen atmosphere and a solution of the bromoketone (250 g, 0.783 mol) in dry toluene (400 ml) is added over 1 hour. The internal temperature is maintained at −10 to −20° C. The mixture is stirred for 1 hour. The mixture is warmed to room temperature, activated carbon (75 g) is added, and the mixture is stirred for 5 minutes. The mixture is filtered through Celite and the cake is washed with toluene (2.5 L). The filtrates are concentrated under reduced pressure, at 20° C., to approximately 700 ml volume. This solution is used directly in the next step. [0016]
  • EXAMPLE 2 7-anti{4-[3-(Trifluoromethyl)phenoxy]-3(R)-tert-butyldimethylsilyloxy-1(E)butenyl}-5-endo-tert-butyldimethylsilyloxybicyclo[2,2,1]hepten-2-one
  • A dry 10 L flange flask, fitted with an overhead stirrer, temperature probe, nitrogen inlet and pressure equalised dropping funnel, is purged with nitrogen and cooled to −70° C. A solution of tert-butyllithium (1.7 M in pentane, 1013 ml, 1.72 mol) is added. The solution is re-cooled to −70° C. and a solution of the vinyl iodide (436 g, 0.923 mol) in diethyl ether (1.3 L) is added over 90 minutes, maintaining the internal temperature below −60° C. [0017]
  • In the mean time, thiophene (75.2 ml, 0.94 mol) is placed in a dry 2L 3-necked flask, under a nitrogen atmosphere. THF (600 ml) is added, and the solution is cooled to −30° C. n-Butyllithium (2.5 M in hexanes, 376 ml, 0.94 mol) is added over 20 minutes. The solution is stirred for 20 minutes at −20° C., then the resulting yellow solution is added to a suspension of copper(I) cyanide (84.15 g, 0.94 mol) in THF (800 ml) at −20° C. over 15 minutes. The resulting dark brown solution is re-cooled to −10° C., and stirred for 20 minutes. [0018]
  • The freshly prepared lithium 2-thienylcyanocuprate solution, at −10° C., is added to the vinyllithium solution at −70° C. over 20-30 minutes. The resulting solution is stirred for 30 minutes at −70° C. The tricycle solution (approximately 187 g in 600 ml toluene, +100 ml THF added) is cooled to −70° C., and added to the cuprate solution, at −70° C. over 20 minutes. The mixture is stirred at −70° C. for one hour, then the cooling bath is removed from the reaction vessel and saturated ammonium chloride (3 L) is added. The mixture is stirred for 20 minutes, the aqueous layer becomes deep blue in colour and a yellow/green precipitate forms. The mixture is filtered through a No 3 filter paper and the filter cake is washed with methyl tert-butyl ether (1 L). The organic layer is separated, and the aqueous layer is extracted with methyl tert-butyl ether (1 L). The combined organic layers are washed with brine (2L), dried (MgSO[0019] 4) and decolourised with activated carbon. After 20 minutes the solution is filtered, the cake is washed with methyl tert-butyl ether (2.5 L), and the filtrate is evaporated under reduced pressure.
  • The residue is taken up in heptane and passed through a plug of silica (1.5 Kg), eluting with 2% EtOAc/heptane to 10% EtOAc/heptane to provide the pure ketone as a yellow solid (293 g, 64%), m.p. 64-72° C.; [α][0020] D 20+35.9° (c=1.05, DCM); δH (400 MHz, CDCl3) 7.38 (1 H, t, J 8), 7.25 (1 H, d, J 8), 7.13 (1 H, s), 7.07 (1 H, d, J 8), 5.86 (1 H, dd, J 16, 8), 5.73 (1 H, dd, J 16, 6), 4.55 (2 H, m), 3.90 (2 H, d, J 7), 2.80 (1 H, m), 2.77 (1 H, d, J 18), 2.57 (2 H, m), 2.45 (1 H, m), 2.05 (1 H, dd, J 18, 4), 1.35 (1 H, m), 0.95 (9 H, s), 0.90 (9 H, s), 0.15 (3 H, s), 0.14 (3 H, s) and 0.05 (6 H, s); δc (100 MHz, CDCl3) 216.0, 158.82, 132.02, 131.54 (q, J 32), 129.98, 127.77, 123.90 (q, J 270), 118.04, 117.55, 111.13, 72.32, 71.34, 69.92, 54.37, 50.23, 46.24, 38.80, 33.39, 25.76, 18.30, 17.97, −4.67, −4.74, −4.86 and −4.92.
  • EXAMPLE 3 8-anti{4-[3-(trifluoromethyl)phenoxy]-3(R)-tert-butyldimethylsilyl-1(E)-butenyl}-6-endo-tert-butyldimethylsilyloxy-2-oxabicyclo[3,2,1]octan-3-one
  • The ketone (362.6 g, 0.62 mol) and sodium acetate (170 g, 2.07 mol) are dissolved in glacial acetic acid (1.7 L). The reaction vessel is placed in a water bath at 20° C. and peracetic acid (40% in dilute acetic acid, 176.7 ml, 0.93 mol) is added over a period of 20 minutes. The solution is stirred at room temperature for 3 h. More peracetic acid (30 ml) is added, and the solution is stirred for a further 2 h. The reaction mixture is poured onto water (2.5 L), and the products are extracted into MTBE (2×750 ml, then 500 ml). The combined organic phases are washed with water (2 L). The aqueous phase is back extracted with MTBE (500 ml). The combined organic extracts are neutralised with saturated sodium carbonate solution (500 ml) and water (2 L) is added to aid phase separation. The organic phase is washed with water (1 L), brine (1 L), dried (MgSO[0021] 4) and evaporated under reduced pressure, to give a yellow oil (363.8 g). The crude product (consisting of a mixture of regioisomers) is dissolved in acetonitrile (1 L) at room temperature. Sodium hydroxide solution (1M, 300 ml) is added and the solution stirred at room temperature for 2 h. Water (1 L) is added and the product is extracted into MTBE (3×500 ml). The combined organic phases are washed with brine (500 ml), dried (MgSO4), filtered and evaporated under reduced pressure. The residue is recrystallised from heptane (1 L) at −70° C. (cold bath temperature), filtered and washed with cold (−70° C., cold bath) heptane (2×200 ml). The solid is dried, to give the lactone as a white solid (141.2 g, 38%). The mother liquors are filtered through silica gel (1 Kg) eluting with 20% DCM/Heptane, to remove baseline. Recrystallisation from cold (−70° C., cold bath) heptane (400 ml) gave a second batch of lactone (34 g, 9%), m.p. 77-78° C.; [α]D 20−10.9° (c=1.05, DCM); δH (400 MHz, CDCl3) 7.40 (1 H, t, J 8), 7.22 (1 H, d, J 8), 7.10 (1 H, s), 7.05 (1 H, d, J 8), 5.70 (2 H, m), 4.52 (3 H, m), 3.87 (2 H, d, J 6), 3.16 (1 H, d, J 18), 3.00 (1 H, d, J 6), 2.56 (1 H, dd, J 18, 6), 2.46 (1 H, m), 2.39 (1 H, m), 1.88 (1 H, dt, J 16, 3), 0.95 (9 H, s), 0.89 (9 H, s), 0.10 (6 H, s) and 0.04 (6 H, s); δc (100 MHz, CDCl3) 170.39, 158.75, 132.43, 131.92 (q, J 32), 130.05, 127.87, 123.91 (q, J 270), 118.06, 117.68, 111.11, 82.20, 72.26, 71.64, 71.10, 48.94, 42.48, 40.40, 33.05, 25.75, 18.30, 18.04, −4.64, −4.76, −4.88 and −5.10.
  • The lactone of example 7 can be converted to (+)-16-[3-(trifluoromethyl)phenoxy]- 17,18,19,20-tetranor PGF[0022] isopropyl ester using conventional processes; see Scheme II and associated publications.

Claims (2)

We claim:
1. A process for preparing a prostaglandin, or an ester derivative thereof, wherein said process comprises the conversion of a compound of formula 1
Figure US20020111499A1-20020815-C00007
into said prostaglandin, or ester derivative thereof.
2. The process according to claim 1, wherein the prostaglandin is (+)-16-[3-trifluoromethyl)phenoxy]-17,18,19,20-tetranor PGF isopropyl ester.
US09/898,241 1999-04-12 2001-07-03 Intermediate for the synthesis of prostaglandins Expired - Lifetime US6437152B1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US09/898,241 US6437152B1 (en) 1999-04-12 2001-07-03 Intermediate for the synthesis of prostaglandins
US10/224,250 US20030004368A1 (en) 1999-04-12 2002-08-19 Novel intermediate for the synthesis of prostaglandins

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
GB9908326 1999-04-12
GB9908326.3 1999-04-12
GBGB9908326.3A GB9908326D0 (en) 1999-04-12 1999-04-12 Novel process intermediate for the synthesis of prostaglandins
US13646499P 1999-05-28 1999-05-28
US09/547,205 US6294679B1 (en) 1999-04-12 2000-04-12 Intermediate for the synthesis of prostaglandins
US09/898,241 US6437152B1 (en) 1999-04-12 2001-07-03 Intermediate for the synthesis of prostaglandins

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/547,205 Division US6294679B1 (en) 1999-04-12 2000-04-12 Intermediate for the synthesis of prostaglandins

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US10/224,250 Continuation US20030004368A1 (en) 1999-04-12 2002-08-19 Novel intermediate for the synthesis of prostaglandins

Publications (2)

Publication Number Publication Date
US20020111499A1 true US20020111499A1 (en) 2002-08-15
US6437152B1 US6437152B1 (en) 2002-08-20

Family

ID=27269692

Family Applications (3)

Application Number Title Priority Date Filing Date
US09/547,205 Expired - Fee Related US6294679B1 (en) 1999-04-12 2000-04-12 Intermediate for the synthesis of prostaglandins
US09/898,241 Expired - Lifetime US6437152B1 (en) 1999-04-12 2001-07-03 Intermediate for the synthesis of prostaglandins
US10/224,250 Abandoned US20030004368A1 (en) 1999-04-12 2002-08-19 Novel intermediate for the synthesis of prostaglandins

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/547,205 Expired - Fee Related US6294679B1 (en) 1999-04-12 2000-04-12 Intermediate for the synthesis of prostaglandins

Family Applications After (1)

Application Number Title Priority Date Filing Date
US10/224,250 Abandoned US20030004368A1 (en) 1999-04-12 2002-08-19 Novel intermediate for the synthesis of prostaglandins

Country Status (1)

Country Link
US (3) US6294679B1 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1810967B1 (en) 2006-01-18 2014-07-30 Chirogate International Inc. Processes and intermediates for the preparations of prostaglandins
US8524939B2 (en) 2011-08-24 2013-09-03 Chirogate International Inc. Intermediates for the synthesis of benzindene prostaglandins and preparations thereof
WO2017062770A1 (en) 2015-10-08 2017-04-13 Silverberg Noah Punctal plug and bioadhesives
JP7451732B2 (en) 2020-02-06 2024-03-18 オキュラ セラピューティクス,インコーポレイテッド Compositions and methods for treating eye diseases
US11884613B2 (en) 2022-05-05 2024-01-30 Chirogate International Inc. Processes and intermediates for the preparation of carbaprostacyclin analogues

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH648286A5 (en) * 1978-07-11 1985-03-15 Glaxo Group Ltd PROSTAGLAND DERIVATIVES, THEIR PRODUCTION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THESE DERIVATIVES.
EP0074856A3 (en) 1981-09-16 1984-02-01 Glaxo Group Limited Resolution of racemic bicycloheptenones
US4824993A (en) * 1984-04-23 1989-04-25 Glaxo Group Limited Cyclopentyl ethers and their preparation and pharmaceutical formulation
US5510383A (en) * 1993-08-03 1996-04-23 Alcon Laboratories, Inc. Use of cloprostenol, fluprostenol and their salts and esters to treat glaucoma and ocular hypertension

Also Published As

Publication number Publication date
US6294679B1 (en) 2001-09-25
US20030004368A1 (en) 2003-01-02
US6437152B1 (en) 2002-08-20

Similar Documents

Publication Publication Date Title
JP5485980B2 (en) Process for producing prostaglandin analogues and intermediates thereof
KR101561171B1 (en) Process for the preparation of f-series prostaglandins
KR20140107541A (en) Process for the preparation of travoprost
US6437152B1 (en) Intermediate for the synthesis of prostaglandins
KR101522218B1 (en) Processes and intermediates for the preparations of prostaglandins
EP1169325B1 (en) Novel intermediate for the synthesis of prostaglandins
WO2014094511A1 (en) Intermediates for synthesizing treprostinil and preparation method thereof as well as the preparation method of treprostinil thereby
US5292946A (en) In-situ preparation of diisopinocamphenyl chloroborane
US6388128B1 (en) Stannane synthesis of prostanoids
JPH0717627B2 (en) Process for producing 2,5-diaryltetrahydrofurans and analogs thereof useful as PAF antagonists
KR19980702884A (en) Process for preparing prostaglandins E1, E2 and analogs thereof using furylcopper reagent
KR100340760B1 (en) Stereoselective process for preparing (R)-(-)-muscone
CN117843599A (en) Preparation method of prostaglandin intermediate containing 1-alkyl-1-alkenyl ethanol structure
JP2023165632A (en) Processes and intermediates for preparation of carbaprostacyclin analogues
TW202248197A (en) Processes and intermediates for the preparations of benzoprostacyclin analogues and benzoprostacyclin analogues prepared therefrom
JP3403761B2 (en) 3,5-Dihydroxy-1-octen-7-ynes and method for producing the same
JPH07233175A (en) Asymmetric production of metal-substituted cyclopropylmethanol derivative.
JPH0651676B2 (en) (5E) -Prostaglandin E (2) Manufacturing method
JPH0470297B2 (en)
JPS6377853A (en) Isocarbacyclines and production thereof
JPS638091B2 (en)

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: PAYER NUMBER DE-ASSIGNED (ORIGINAL EVENT CODE: RMPN); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Free format text: PAYOR NUMBER ASSIGNED (ORIGINAL EVENT CODE: ASPN); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STCF Information on status: patent grant

Free format text: PATENTED CASE

FPAY Fee payment

Year of fee payment: 4

REMI Maintenance fee reminder mailed
FPAY Fee payment

Year of fee payment: 8

SULP Surcharge for late payment

Year of fee payment: 7

FPAY Fee payment

Year of fee payment: 12

SULP Surcharge for late payment

Year of fee payment: 11