throbber
MSNv. Bausch - IPR2023-00016
`
`orw\t
`WEALTH &‘erat‘of WisconsA
`a
`7305 Linden Nr 50 Wis. 53706
`MAR 1909
`
`V Exhibit 1025 - Page 1 of 27
`
`

`

`Editorial Board
`
`—————ee
`
`EDITOR
`
`BOARD
`
`Pentti Siiteri, Ph.D.,
`University of California San Francisco
`School of Medicine HS W- 1656
`Department of Obstetrics/Gynecology-Reproductive Medicine
`San Francisco, CA 94143
`
`L. E. Braverman, M.D.
`University of Massachusetts Medical School, Worcester, MA
`J. W. Funder, M.D.
`Medical Research Center Prince Henry’s Hospital, Melbourne, Victoria, Australia
`M. M. Grumbach, M.D.
`University of California San Francisco, San Francisco, CA
`E. Herbert, Ph.D.
`
`Oregon Health Sciences University, Portland, OR HEAEfAL TH
`ieEi0ES
`B. S. Katzenellenbogen, Ph.D.
`y
`University of Illinois, Urbana, IL
`1305 Ling "versity of Wj
`J. L. Kostyo, Ph.D.
`a Dr, MV.
`G
`University of Michigan Medical School, Ann Arbor, MI
`MAR
`H. H. Samuels, M.D.
`New York University Medical Center, New York, NY
`_
`R. S. Sherwin, M. D.
`Yale University School of Medicine, New Haven, CT" Ot
`F. Sweet, Ph.D.
`Washington University Medical School, St. Louis, MO
`R. I. Weiner, Ph.D.
`University of California San Francisco, San Francisco, CA
`J.D. Wilson, M.D.
`University of Texas Southwestern Medical School, Dallas, TX
`S. S.C. Yen, M.D.
`University of California at San Diego, La Jolla, CA
`
`oe 2
`
`LIBRaz rm
`
`te,
`
`‘
`
`Contents
`
`Vol. 7, No. 1, February 1986
`
`Editorial Note
`
`Introduction
`
`The Molecular Basis of Gonadotropin-Releasing
`Hormone Action
`
`Hypogonadotropic Disorders in Men and Women:
`Diagnosis and Therapy with Pulsatile
`Gonadotropin-Releasing Hormone
`
`Pentti Siiteri
`
`P. Michael Conn
`
`P. Michael Conn
`
`Nanette Santoro, Marco Filicori,
`and William F. Crowley, Jr.
`
`MSNExhibit 1025 - Page 2 of 27
`MSNv. Bausch - IPR2023-00016
`
`1
`
`1
`
`3
`
`11
`
`

`

`Use of a Potent, Long Acting Agonist of
`Gonadotropin-Releasing Hormonein the Treatment
`of Precocious Puberty
`
`Induction of Ovulation in Primate Models
`
`Gonadotropin-Releasing Hormone Analog Design.
`Structure-Function Studies Toward the
`Developmentof Agonists and Antagonists: Rationale
`and Perspective
`
`Treatment of Prostate Cancer with Gonadotropin-
`Releasing Hormone Agonists
`
`Paul A. Boepple,
`M. Joan Mansfield,
`Margaret E. Wierman,
`Craig R. Rudlin,
`Hans H. Bode,
`John F. Crigler, Jr.,
`John D. Crawford, and
`William F. Crowley, Jr.
`
`Daniel Kenigsberg,
`Burt A. Littman, and
`Gary D. Hodgen
`
`Marvin J. Karten and
`Jean E. Rivier
`
`F. Labrie, A. Dupont,
`A. Bélanger, R. St-Arnaud,
`M. Giguere, Y. Lacourciere,
`J. Emond, and G. Monfette
`
`Gonadal Regulation of Hypothalamic Gonadotropin-
`Releasing Hormone Release in Primates
`
`Tony M.Plant
`
`Andrea Manni, Richard Santen,
`Harold Harvey, Allan Lipton, and
`Devorah Max
`
`David J. Handelsman and
`Ronald 8. Swerdloff
`
`S. Bhasin and R. S. Swerdloff
`
`Brian H. Vickery
`
`Treatment of Breast Cancer with Gonadotropin-
`Releasing Hormone
`
`Pharmacokinetics of Gonadotropin-Releasing
`Hormoneand Its Analogs
`
`Mechanisms of Gonadotropin-Releasing Hormone
`Agonist Action in the Human Male
`
`Comparison of the Potential for Therapeutic
`Utilities with Gonadotropin-Releasing Hormone
`Agonists and Antagonists
`
`Special Issue Index
`
`Endocrine Society Guidelines for Membership
`Application
`
`Endocrine Society Nomination for Membership
`
`The Endocrine Society 68th Annual Meeting
`
`Registration Form
`
`24
`
`34
`
`44
`
`67
`
`75
`
`89
`
`95
`
`106
`
`115
`
`125
`
`126
`
`127
`
`128
`
`129
`
`MSN Exhibit 1025 - Page 3 of 27
`MSNv. Bausch - [PR2023-00016
`
`

`

`AUTHOR AND SUBSCRIPTION INFORMATION
`
`Address manuscripts and correspondenceto:
`Pentti K. Siiteri, Ph.D., Editor
`543 McClay Road
`Novato, California 94947
`(415) 898-1581
`
`All business matters, including correspondence and remittance relating to nonmember subscriptions,
`back volumesand advertising, and orders and remittance for reprints, should be sent to ENDOCRINE
`REVIEWS, 428 E. Preston St., Baltimore, Md. 21202. Copies will not be replaced without charge
`unless we receive a request within 60 days of the mailing in the U.S. or within 90 daysin all other
`countries.
`
`Bound volume number6 will be available in early 1986 to subscribers only. The volumewill be hand
`bound in black buckrum,and the journal name, volume number, and year of issue will be stamped in
`gold on the spine. The price is $45.00 (plus an additional $5.00 for orders outside the U.S.) with a
`$2.00 discount given for advance payment. Orders must be received by the publisher before December
`1, 1985.
`
`Change of Address: Notify both Williams & Wilkins, 428 E. Preston St., Baltimore, Md. 21202,
`and The Endocrine Society, 9650 Rockville Pike, Bethesda, Md. 20814,of all address changes.
`Publication of advertisements in the Journals of The Endocrine Society does not imply endorsement
`by the Society of the reagents or services offered.
`No part may be duplicated or reproduced without permission of the Publisher and the Editor.
`Printed in U.S.A.
`
`THE ENDOCRINE SOCIETY
`
`Bert W. O’Malley, President
`Sidney H.Ingbar, President-Elect
`
`Nicholas P. Christy, Secretary-Treasurer
`
`9650 Rockville Pike, Bethesda, Maryland 20814.
`
`PUBLICATIONS COMMITTEE—HILTon A. SALHANICK, Chairman; BERT W. O’MALLEY,
`Ex-Officio; SIDNEY H. INGBAR, Ex-Officio; NICHOLAS P. CHristy, Ex-Officio; ALFRED E.
`WILHELMI, Ad Hoc; GLENN D. BRAUNSTEIN, MARY F. DALLMAN, HUNTER HEATHIII, PHILIP
`TROEN.
`
`COUNCIL—GERALD D. AURBACH, Louis VY. AviotI, LEwis E. BRAVERMAN, DELBERT A.
`FISHER, LAWRENCE A. FROHMAN, RICHARD HORTON, SELNA L. KAPLAN, JACK L. Kostyo,
`Jack H. OPPENHEIMER, JEAN D. WILSON.
`
`NETTIE C. KARPIN,Executive Director, 9650 Rockville Pike, Bethesda, Maryland 20814.
`
`Correspondencerelating to membership applications and Society business including remission
`of members’ dues and subscriptions should be sent toNicholas P. Christy, Secretary-Treasurer,
`
`ENDOCRINE REVIEWS(ISSN 0163-769X) is published quarterly by The Endocrine Society, 428 E. Preston St., Baltimore,
`MD21202. Memberprice: $20.00 ($27.00 foreign) for journal subscription. Subscription rates: individuals $45.00; foreign $55.00;
`institutions $60.00, foreign $70.00; in-training $30.00 ($40.00 foreign); single copy $10.00 ($12.00 foreign), Subscription prices
`subject to change. Japanese Yen price is available from our sole agent U.S.-Asiatic Co. Ltd., 13-12, Shimbashi 1-Chome, Minato-
`Ku, Tokyo 105, Japan, telephone 03-502-6471. Second class postage paid at Baltimore, MD and at additional mailing offices.
`Postmaster, send address changes (Form 3579) to Williams & Wilkins, 428 E, Preston St., Baltimore, MD 21202. Indexed by
`Current Contents and Index Medicus. Copyright © 1986 by The Endocrine Society.
`
`MSNExhibit 1025 - Page 4 of 27
`MSNv. Bausch - IPR2023-00016
`
`

`

`0163-769X/86/0701-0044$02.00/0
`Endocrine Reviews
`Copyright © 1986 by The Endocrine Society
`
`Vol. 7, No. 1
`Printed in U.S.A.
`
`|
`Gonadotropin-Releasing Hormone Analog Design.
`Structure-Function Studies Toward the Developmentof
`Agonists and Antagonists: Rationale and Perspective
`
`MARVIN J. KARTEN anp JEAN E. RIVIER
`
`Contraceptive Development Branch (M.J.K.), Center for Population Research, National Institute of Child
`Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892; and The
`Clayton Foundation Laboratories for Peptide Biology(J.E.R.), The Salk Institute, La Jolla, California 92037
`
`Introduction
`
`N JUNE 24, 1971, Andrew V. Schally announced
`the determination of the primary structure of por-
`cine GnRH at The Endocrine Society Meeting in San
`Francisco. This announcement was followed by publica-
`tions by Matsuoet al. (1, 2) and Babaet al. (3) on the
`proposed aminoacid sequence for porcine GnRHandits
`synthesis and by Burgus et al.
`(4) who characterized
`ovine GnRH andfound the sequenceto be identical with
`that of porcine GnRH. Thephysiological and therapeutic
`importance attributed to the discovery of the new sub-
`stance was greatly increased by the prospect of the design
`of potent and long acting GnRH agonists and antago-
`nists. Since that time more than 2000 analogs of GnRH
`have been synthesized. The impact of research of GnRH
`and its analogs on clinical medicine recently led Ziporyn
`(5) to note, “There’s almost no subspecialty of medicine
`that will be left untouched by the [research] advances
`associated with LHRHorits analogs.” It is the intent of
`this article to provide a historical review of the major,
`and some minor, aspects of the chemical development of
`GnRHagonists and antagonists up to the present state
`of development (July 1, 1985). The synthetic chemical
`efforts have been devoted largely to increasing the affin-
`ity of the peptides to the GnRH receptor and their
`resistance to degradation or elimination in in vivo sys-
`tems, characteristics which, for the GnRH analogs, are
`generally interrelated.
`An annualcompilation and review of structure-activity
`relationships of GnRH analogsis available in the Spe-
`cialist Periodical Reports which coverthe literature pub-
`lished during 1971-1980 (volumes 4—13) (6-15). At irreg-
`ular intervals, Annual Reports in Medicinal Chemistry
`
`Address requests for reprints to: Dr. Marvin J. Karten, Contracep-
`tive Development Branch, Center for Population Research, National
`Institute of Child Health and Human Development, NationalInstitutes
`of Health, Bethesda, Maryland 20892.
`
`furnish brief updates of the studies of structure-activity
`relationships of GnRH analogs (16-21). These reports,
`in conjunction with two recently published comprehen-
`sive monographs should provide the reader with a bal-
`anced account of the development of GnRH analogs (22-
`29).
`
`Synthesis, Purification, and Characterization of
`GnRH Analogs
`
`It is important to note that the rapid developmentof
`GnRHanalogs was made possible through the extensive
`use of solid phase peptide synthesis (SPPS) introduced
`by Merrifield (30). As one of the codevelopers of the
`method (31), Stewart (32) has pointed out that the use
`of automated equipment for SPPS, benzhydrylamine-
`like resins for peptide amide synthesis (33, 34), and
`adequate methods for the purification of peptides, par-
`ticularly reverse phase HPLC (RP-HPLC) in recent
`years (35), have made the synthesis of mammalian
`GnRH,
`<Glu-His-Trp-Ser-Tyr—Gly—Leu—Arg—Pro-
`Gly-NHp, and its analogs, a relatively simple task. Al-
`thoughclassical (solution) methods have been employed
`for the synthesis of GnRH (see Ref. 36 and references
`therein) and its analogs (37), it is quite clear that the use
`of SPPS and RP-HPLC were essential for the rapid
`exploration of structure-activity relationships as well as
`providing investigators with relatively large amounts of
`these substances for pharmacological, toxicological, and
`clinical studies.
`While the purity of the agonists synthesized either by
`SPPSorclassical (solution) methods was always of con-
`cern in terms of the concomitant biological activity of
`potential racemization products, it was a critical factor
`in the biological evaluation of the antagonists. This was
`particularly true in the early stages of development when
`the GnRHinhibitory activities were very low and could
`be masked by a small amount of racemized material
`
`MSN Exhibit 1025 - Page 5 of 27
`MSNv. Bausch - IPR2023-00016
`
`

`

`February, 1986
`
`GnRH ANALOG DESIGN
`
`45
`
`present as a contaminant and acting as an agonist. For
`example,
`the high GnRH potency of [p-Ala®*]GnRH
`(350-450% of GnRH) would barely be affected by a 10%
`racemization contaminant of
`[L-Ala®*]GnRH (4% of
`GnRH) (38). However, a very weak antagonist, e.g. [D-
`Phe”]GnRH,(39) could haveits activity masked by the
`presence of small amounts of {L-Phe*]GnRH (40) with
`only 2-4% of the potency of GnRHbutwithfull intrinsic
`activity in vitro. The separation of the possible diaster-
`eomers, e.g.
`[D-Ala®]GnRH and [L-Ala®]GnRH, which
`could result from racemization, was eventually made
`feasible through the use of HPLC, thus eliminating one
`element of uncertainty from the interpretation of the
`biological results (35). Similarly, a preparation of [D-
`His’?]GnRHexhibiting 10% GnRH-like potency (39) was
`subsequently shown, when purified by HPLC,
`to be
`inactive either as an agonist or an antagonist
`(41).
`Racemization of histidine during peptide synthesis is
`well documented anddifficult to prevent irrespective of
`whetherclassical methods of synthesis or SPPSare used.
`The widespread use of HPLC resulted from the recog-
`nition that classical methodsof purification were inferior
`to HPLC asa tool for the separation of these diastereo-
`meric peptides. Most GnRH analogs reported in the
`literature have been characterized by amino acid analysis
`only, often without a quantitative determination of the
`unnatural amino acids. Investigators have relied on the
`presumed authenticity of the starting protected amino
`acid and on high coupling efficiency during the assem-
`bling of the peptides on the resins rather than pursuing
`rigorous methods of characterization of the peptides.
`Nuclear magnetic resonance (NMR) and mass spectrom-
`etry were employed in those cases where definite proof
`of structure was required. Optical rotations were gener-
`ally measured, and TLC and HPLC, in several systems,
`were used for proof of homogeniety.
`
`Development of GnRH Agonists
`
`The original incentive for the development of more
`potent GnRH agonists was the expectation that the
`knowledge of the LH-releasing and ovulation-inducing
`effects of GnRHobservedin laboratory animals could be
`applied to the treatment of male and female infertility
`(42), However, the half-life of GnRH is very short (43,
`44) and more potent and longer acting analogs were
`thought
`to be necessary for practical clinical utility,
`Tegardless of any anticipated or unanticipated therapeu-
`tic applications. Potent agonists, referred to as supera-
`f0nists, were rapidly produced and were subsequently
`discovered, along with GnRH,to have, ironically, anti-
`reproductive effects. They were available for reproduc-
`tive pharmacological evaluation (42, 45, 46) within 3 yr
`of the structure elucidation of GnRH;this accounts for
`
`their rapid clinical exploration exemplified by the first
`demonstration (in 1978) of inhibition of reproductive
`function in women by a superagonist (47). Once super-
`agonists had been synthesized and their potential ther-
`apeutic value was recognized, further incentive, after
`1976, to seek structurally novel and longer acting ago-
`nists was provided by promising commercial considera-
`tions.
`The various biological assays and animal models that
`have beenutilized for the testing of GnRH agonists have
`been recently reviewed (42, 48). The most widely used in
`vitro assays have been,initially, the dispersed pituitary
`cell for the measurement of LH and FSHsecretions and,
`morerecently, the receptor binding assay using purified
`pituitary membranefractions for the estimation of the
`potenciesof the analogs (49, 50). In vivo biological assays,
`which have been utilized to determine the potencies of
`GnRHagonists, include induction of ovulation, disrup-
`tion of the estrus cycle, stimulation of uterine growth,
`inhibition of pregnancy, stimulation of LH release in
`ovariectomized rats, and stimulation of LH/FSH release
`in immature rats using an infusion technique (42, 48).
`
`Pro®-ethylamide (NEt) modifications
`
`The first important structural modification of GnRH
`leading to increased potency was discovered by Fujino
`and co-workers (37), who examined the effect of struc-
`tural modifications at the C terminus of GnRH.Although
`the des-amide of GnRH (GnRHfree acid) exhibited very
`low GnRH potency in
`ovariectomized rats
`(51)
`and Pro?-GnRH showed only 10% of the potency of
`GnRHin vitro (34), replacement of the glycine amide
`terminus with alkyl amines produced nonapeptide alkyl
`amides with significantly greater ovulation-inducing po-
`tency than GnRHitself (37). Thus,
`[Pro*-ethylamide
`(NEt)]GnRH,the most potent analog of the series, was
`5 times more potent than GnRH and more potent than
`either the Pro*-methylamide (NHMe)or Pro’-propylam-
`ide (NHPr) modifications. The fluorinated ethylamide
`analog, [Pro*-NHCH,CF;]GnRH was subsequently re-
`ported by Coyetal. (52) to be twice as potent as [Pro®-
`NEt]GnRHin releasing LH when administered to im-
`mature male rats. The data of Fujino et al. (37), it was
`noted, suggested that the terminal glycine amide was not
`essential for high potency andthat the total chain length
`of the peptide played an important role in the binding
`affinity of the analog for the pituitary receptor. It was
`also suggested that the introduction of this Pro-alkylam-
`ide moiety may also increase the duration of action of
`these analogs by virtue of their greater resistance to
`enzymatic degradation (53). These two desired proper-
`ties, greater binding affinity and enzymatic resistance to
`proteolysis, were to becomethe basis for the rational
`
`MSN Exhibit 1025 - Page 6 of 27
`MSNv. Bausch - IPR2023-00016
`
`

`

`46
`
`KARTEN AND RIVIER
`
`Vol. 7, No. 1
`
`design and for the explanationofactivity, or lack thereof,
`of all the GnRH analogs regardless of the site of struc-
`tural modification. The concept that protection from
`renal elimination would also lead to prolonged action
`was eventually incorporated into the design of GnRH
`analogs.
`
`p-Xaa® modifications
`
`The second important structural modification of
`GnRH,discovered by Monahanetal. (38), was the re-
`placementof the Gly® residue with D-alanineyielding [D-
`Ala®]GnRH with a potency of approximately 350-450%
`that of GnRH both in vitro and in ovariectomized rats.
`The corresponding [L-Ala®]GnRH had only 4% of the
`potency of GnRH, and it was suggested that since the
`potencies determined in vivo were in agreement with the
`in vitro results, it was unlikely that the differences in the
`biological activities could be solely explained by differ-
`encesin clearance rates. Instead, the increased biological
`potency wasattributed to the conformationalstabilizing
`effect of the p-alanine which was favorable for binding
`(and activity) at the receptor. This study is also note-
`worthy for its suggestion that GnRH may conformation-
`ally contain a @-II type bend (involving Ser-Tyr—Gly-
`Leu) whichis stabilized in [pD-Ala®]GnRH andpreferable
`for binding at the receptor site. This point will be dis-
`cussed below when the contribution of conformational
`studies to the design of GnRH analogs, particularly
`antagonists,
`is reviewed. Also to be deferred for later
`discussion is our knowledge of the enzymatic degradation
`of peptides and its contribution to the design (if any) of
`GnRHanalogs. However,it should be noted that, regard-
`less of whether the Gly*-Leu’ bondor the Tyr®-Gly® bond
`is considered to be a major site of proteolytic cleavage,
`substitution of glycine by D-amino acids is likely to
`render either linkage more resistant to enzymatic deg-
`radation (26).
`
`Additive effects (or lack thereof) of Pro°-NEt and p-Xaa®
`modifications
`
`It is often assumed that the biological effect of com-
`bining several structural changes in one molecule will be
`additive (or, more correctly, multiplicative). Thus, ac-
`cording to this additivity rule (54),
`if one structural
`modification leads to a relative potency of a and asecond
`modification to a relative potency of b then the combi-
`nation of both structural modifications in a single mol-
`ecule would be expected to yield ari analog with a biolog-
`ical potency of a multiplied by b. The potential additivity
`of biological potency of the Pro?-NEt and p-Ala® modi-
`fications was immediately tested and, in rapid succession,
`two reports [Coyet al. (55) and Fujinoet al. (56)] on this
`important combination appeared. Infusion experiments
`
`with immature male rats showed that the three analogs,
`[p-Ala®,Pro*-NEt]GnRH,
`[p-Ala®]GnRH, and [Pro?-
`NEt]GnRH, had LH/FSH releasing potency ratios of
`12-16:7-8:2.5, respectively, compared with GnRH (55).
`Similar ovulation-inducing potency ratios were observed
`(56) among the three analogs, but the potencies relative
`to GnRH were much higher. These in vivo results cor-
`related with the in vitro results of Vale et al. (57) who,
`using the stimulation of LH secretion from rat pituitary
`cells in culture by GnRHagonists as an index of potency,
`noted that the combination of the two structural modi-
`fications yielded an analog with a potency approximately
`equal to the product of the potencies of the individual
`modifications. The in vitro results of Fujino et al. (56)
`did not agree with these findings. Although it is self-
`evident that comparisonsofbiological data by different
`groupsare valid only if the same bioassays are employed
`in precisely the same manner(48), nevertheless, many
`of the apparent disagreements in the data reported,
`regarding comparisons of potencies of superagonists, can
`be attributed to a disregard of this axiom. Thus, the
`validity of the extension of the additivity principle to the
`combination of a D-aromatic amino acid in position 6
`and the Pro’-NEt modification became a focal point of
`interest with the publication of apparently conflicting
`reports on this subject (57-59). The observation was
`made that agonists with D-aromatic amino acids such as
`[D-Trp*]GnRH and [p-Trp®,Pro*-NEt]GnRH are much
`more potent (36 times and 144 timesthe in vitro potency
`of GnRH,respectively) than the corresponding substi-
`tutions with aliphatic amino acids, such as [D-Ala’]
`LHRHand [p-Ala®,Pro®-NEt]GnRH,which are approx-
`imately 4 times and 14 times, respectively, the potency
`of GnRH (57). The subsequent binding studies of Perrin
`et al.
`(50) showed increased binding potencies for the
`Pro’-NEt modifications as compared with [p-Ala®]
`GnRHor [D-Trp*]GnRH;however, when compared with
`the biological potencies, in stimulating LH secretion in
`vitro, the increases were far less dramatic. The data for
`the D-Trp® analogs are in accord with the subsequent
`binding studies of Barronetal. (60) but are not in accord
`with the earlier in vitro data reported by Coyet al. (58).
`Jn vivo measurements in immature rats comparing [D-
`Leu®]GnRH and[p-Leu®,Pro®-NEt]GnRH (Leuprolide),
`using integrated levels of LH over a 6-h period after
`injection, showed a similar additive effect (61), but the
`additive effect was reported not to hold true for the
`corresponding D-aromatic amino acid modifications in
`this same assay system (59). Thus, in male rats, the Pro?-
`NEt modification was reported to decrease the potency
`of [D-Phe*]GnRH and [p-Trp*]GnRH by a factor of
`nearly 2 (59). However, it has been more recently re-
`ported that, using estrus suppression (62) as an index of
`agonist activity, [D-Trp*]GnRH and [p-Trp®,Pro®-NEt]
`MSNExhibit 1025 - Page 7 of 27
`MSNv. Bausch - IPR2023-00016
`
`|
`
`

`

`February, 1986
`
`GnRH ANALOG DESIGN
`
`47
`
`GnRH were equipotent. Postcoital comparisonsin rats
`(Naqvi, R. and M. Lindberg, unpublished observations)
`also indicated that the two analogs were equipotent. In
`women,it was reported that the two analogs were equi-
`potent with respect to the sc dose required for maximal
`LHrelease (63). Barronetal. (60) showed that MCRsin
`pregnant women were similar for both analogs. They
`concluded that since the NEt residue, which is reported
`to protect the peptide from postproline-cleaving enzyme
`activity, did not lead to a prolonged survival
`time in
`pregnant women,degradation by this enzyme in human
`tissues contributes minimally to GnRH clearance. Sup-
`port for this conclusion was found in reports that both
`analogs were equipotent in vivo (42, 64). Thus, the over-
`whelming evidence points to i7 vivo equipotency for the
`two D-Trp® analogs irrespective of the in vitro results
`and binding studies supporting the additive effects on
`the biological potency of the p-Trp* and Pro®-NEt mod-
`ifications.
`
`Hydrophobic modifications at position 6
`
`The trend toward seeking more potent agonists with
`increasing hydrophobic character resulted in the addition
`of two more superagonists to the growinglist of analogs
`available for clinical exploration.
`[N’-Bzl-p-His*,Pro’-
`NEt]GnRH (Histerelin) was designed by Rivier etal.
`(22) to have the characteristics of high water solubility
`at acidic pH and greater lipophilic character in vivo,
`while retaining high biological potency. A correlation
`was noted between the in vitro potencies of certain
`position 6 superagonists and their HPLC retention times
`at physiological pH. These correlations included [N’-
`Bzl-p-His®,Pro°-NEt]GnRH and_
`[p-Trp*,Pro®’-NEt]
`GnRH, with the former being slightly more potent than
`the latter, in vitro. In vivo results showed a similar trend
`(48).
`It had previously been observed that the incorporation
`of D-amino acids, with larger and more lipophilic side
`chains than in [D-Leu®|GnRH, such as [D-Trp*]GnRH,
`tesulted in more potent agonists (57, 59). Nadasdi and
`Medzihradszky (65) proposed a quantitative correlation
`to exist between the potency of position 6 substituted
`GnRH analogs and the calculated hydrophobicity of the
`amino acid side chain. It
`is accepted that increased
`lipophilicity of drugs is generally associated with greater
`Tetention of the drug in the body and, therefore, pro-
`longed duration of action (66). The retention may be the
`Tesult of enhanced renal reabsorbtion or fat storage of
`honionized fat-soluble compounds. Protection of the
`drug from renal excretion, through plasma protein bind-
`Ing, will also affect its duration of action. Plasma protein
`inding generally increases, in a given series of analogs,
`With increasing hydrophobicity (66). With this in mind,
`
`Nestoret al. (62, 67) postulated that analogs with greater
`hydrophobicity could have an extended biological half-
`life resulting from a whole body depoteffect. They would
`attribute this effect to a decreased rate of clearance of
`the analog from thecirculation and the increased binding
`capacity of the analog for hydrophobic plasmacarrier
`proteins (67). The results of the study on a wide range
`of hydrophobic analogs showed that the most potent
`ones were found in a hydrophobicity range, as measured
`by their retention time on RP-HPLC(68), greater than
`that of the p-Trp® analogs. As an example, [D-Nal(2)*]
`GnRH(Nafarelin acetate), the most potentofthis series,
`was reported to be 200 times more potent than GnRHin
`suppressing estrus in rats. It was pointed out, however,
`that analogs with greater hydrophobicity than Nafarelin
`acetate were less potent and this includes the analog
`incorporating the Pro®-NEt modification into Nafarelin
`acetate
`(62).
`Interestingly,
`[p-Nal(2)*]GnRH and
`[D-Nal(1)°]GnRH wereisolipophilic but the latter was
`4-fold less potent. Other examples (68, 69) also bear
`witness to the inadequacy of using hydrophobicity alone
`as a prediction of agonist potency (28).
`Nafarelin acetate, which was reported to be twice as
`potent as [p-Trp*]GnRH,[p-Trp*,Pro°-NEt]GnRH,or
`[N*-Bzl-p-His®,Pro*-NEt]GnRH in estrus suppression
`comparisons (26), became the last superagonist to be
`madeavailableforclinical exploration. The improvement
`of the pharmacokinetics with the more hydrophobic ag-
`onist may appear to be supported by the comparisonsof
`the reported half-lives of GnRH [t,, = 8 min, constant
`infusion, (44)], [D-Trp*]GnRH [t, ~ 30 min, constant
`infusion,
`(44)], and [pD-Nal(2)*]GnRH [t,, = 2.4 h, se
`administration, (67)]; however, comparison of the three
`peptides underidentical conditions is not available. The
`considerably longer plasma elimination half-lives re-
`ported for Nafarelin acetate in rats, monkeys, and hu-
`mans than reported for GnRH or
`[p-Leu®,Pro®-NEt]
`GnRH (Leuprolide) were attributed, at least in part, to
`the more extensive plasma binding of Nafarelin acetate
`(70).
`N*, N*'-dialkyl-D-homoarginines were incorporated
`into position 6 of GnRH agonists (67, 71) as a result of
`successful GnRH antagonist investigations with this un-
`natural amino acid. The most potent, [N*,N*’-diethyl-
`D-Har®,Pro®-NEt]GnRH, was only slightly less potent
`than [D-Nal(2)°]GnRH in the rat estrus suppression
`assay.
`
`Other C-terminally modified analogs
`
`Another structural modification which has generally
`led to increases in potency, in combination with D-amino
`acids in position 6, is the a-aza-Gly’°(-NHNHCO-) sub-
`stitution. A series of a-aza analogs of GnRH weresyn-
`
`MSNExhibit 1025 - Page 8 of 27
`MSNv.Bausch - IPR2023-00016
`
`

`

`48
`
`KARTEN AND RIVIER
`
`Vol. 7, No, 1
`
`thesized by Duttaetal. (72, 73) with the expectation that
`the presence of an a-aza residue might be conformation-
`ally favorable, leading to higher binding affinity, and be
`moreresistant to enzymatic degradation. Replacement
`of amino acids in position 6, 9, and 10 of GnRHby a-
`aza amino acids alone did not confer any potency advan-
`tage but when the a-aza-Gly’ modification was com-
`bined with, for example, the p-Ser(Bu')® substitution,
`the resulting analog,
`[p-Ser(Bu')®, a-aza-Gly""]GnRH,
`currently undergoing clinical development, was consid-
`ered to be atleast 5 times more potent
`than [D-
`Ser(Bu')®,Pro®-NEt]GnRH, Buserelin, (74), using induc-
`tion of ovulation as a measurementof potency (72, 73).
`(Buserelin has been, clinically, the most extensively stud-
`ied GnRH analog.) It was unclear, to Dutta et al., which
`individual factor was primarily responsible for this en-
`hancementof biological potency. [2-D-Nal®, a-aza-Gly*”]
`GnRH was reported to be slightly more potent than
`Nafarelin acetate and approximately 2.5 times more po-
`tent than the corresponding Pro*-NEt modification in
`the estrus suppression assay (26). However, Nestor (26)
`noted that if the w-aza-Gly” substitution conferred high
`potency byvirtue of its enhanced resistance to the post-
`proline-cleaving enzyme in rat plasma, then the rele-
`vance of this substitution to human therapy was less
`clear since the amount of postproline-cleaving enzyme
`present in human plasma was reported to be 2-5 times
`less than in rat plasma (75, 76). Does the statement of
`Nestor also imply that any C-terminal amide modifica-
`tions generally would not confer any advantage over the
`parent Gly’’-NH, function in humans? Onthe basis of
`the human data available on [D-Trp*]GnRH and [D-
`Trp®,Pro*-NEt]GnRH (60, 63, 64), the answer would
`appearto be yes, although systematic comparisons would
`have to be made.
`
`Ser(Bu') groups at position 6 and 7 not only failed to
`enhance,but actually decreased, the ovulation-inducing
`potency of Buserelin.
`
`Conformationally constrained and backbone modifications
`
`With the recognition that the biological activity of [p-
`Ala®, N-Me-Leu’]GnRHwasconsistent with thatof a 8-
`turn conformation for residues 5-8 of GnRH,Freidinger
`et al.
`(78) introduced a y-lactam as a conformational
`constraint into the 6,7 position of GnRH and found that
`the resulting analog was 9 times more potent than GnRH
`in vitro and, by iv injection in ovariectomized rats, 2.4
`times more potent than GnRH. Further exploration of
`the y-lactam modification has not been made with ago-
`nists, per se, but this modification has been tried with
`GnRHantagonists. Various attempts to obtain a confor-
`mationally restricted agonist through cyclic analog de-
`sign have yielded inactive analogs (79) or agonists with
`low biological potency (22, 80). Spatola (81) has reviewed
`the effect of peptide backbone modification on structure-
`activity relationships. Backbone modifications, as new
`approaches to GnRH agonist design, resulted in rela-
`tively little in vitro potency in the cases of peptide bond
`reversals at the 5-6 or 5-6 and 6-7 position (retro-inverso
`analogs) (82) or in the cases of substitution of a thiom-
`ethylene (-CH.S-) group for the peptide linkage at the
`5-6, 6-7, or 9-10 position of GnRH. Thelatter substi-
`tution at 9-10 had 10% ofthe in vitro potency compared
`to that of GnRH, indicating the necessity of more precise
`conformational requirements for residues 5-8 than for
`residues 9-10 (23).
`Before closing the discussion on the current stage of
`development of the GnRH agonists, it is necessary to
`comment upon efforts to increase the potency of the
`agonists by modifying other aminoacid residues.
`
`Position 7 modifications
`
`Miscellaneous modifications
`
`[N-Me-Leu’]GnRH was found to be equipotent with
`GnRH and [p-Ala®,N-Me-Leu’]GnRH was found to be
`at least as active as [D-Ala®]GnRHinvitro (77). In fact,
`the N-Me-Leu’ modification has been incorporated into
`[p-Trp®,Pro®-NEt]GnRH
`yielding
`[D-Trp*®, N-Me-
`Leu’,Pro*-NEt]GnRH,an analog currently undergoing
`clinical development (63). Generally, the effect of the N-
`Me-Leu’ modification in enhancing the potency of the
`parent peptide depends on the D-aminoacid at position
`6 and the bioassay used to compare their potencies (45).
`The introduction of the bulky alkyl side chain, the tert-
`butyl group, as an ether into serine, which proved to be
`successful in significantly enhancing potency when in-
`corporated into the 6-position (e.g. Buserelin), also en-
`hanced potency at the 7-position, of nonapeptide NEt
`analogs of GnRH (74). The combination of two D-
`
`in GnRH is the only
`The 1-L-Nal® substitution (83)
`modification in position 3 which resulted in a peptide
`with

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