MedPath

Acyline Advanced Drug Monograph

Published:Sep 30, 2025

Generic Name

Acyline

Drug Type

Small Molecule

Chemical Formula

C80H102ClN15O14

CAS Number

170157-13-8

A Comprehensive Monograph on the Investigational GnRH Antagonist: Acyline (DB11906)

1.0 Executive Summary

Acyline (developmental code name MER-104) is a potent, long-acting, synthetic peptide classified as a third-generation gonadotropin-releasing hormone (GnRH) antagonist.[1] As a small molecule oligopeptide, its primary mechanism of action is the direct, competitive inhibition of the gonadotropin-releasing hormone receptor (GnRHR) located on the anterior pituitary gland.[4] This blockade prevents endogenous GnRH from stimulating the receptor, resulting in an immediate, profound, and sustained suppression of the gonadotropins, luteinizing hormone (LH) and follicle-stimulating hormone (FSH).[6] The subsequent reduction in gonadotropin signaling leads to a rapid decrease in gonadal steroidogenesis, most notably causing serum testosterone levels in males to fall to castrate levels without the initial "flare" effect characteristic of GnRH agonists.[5]

The clinical development program for Acyline was ambitious, with male hormonal contraception as its lead indication, reaching Phase 3 clinical trials.[2] Its potent and long-lasting effect, with a single subcutaneous injection capable of suppressing testosterone for over two weeks, made it a promising candidate for a convenient, long-acting male contraceptive regimen.[7] The compound was also explored in Phase 2 trials for a range of other hormone-dependent conditions, including advanced prostate cancer, androgen deficiency, precocious puberty, and metabolic disorders such as Type 2 diabetes mellitus and obesity.[2]

A key challenge in Acyline's development was its peptide nature, which necessitated parenteral administration via subcutaneous injection.[1] In an effort to improve patient convenience and expand its therapeutic utility, particularly for chronic conditions like prostate cancer, an experimental oral formulation was developed using the GIPET® permeability enhancement technology.[6] While this formulation successfully achieved systemic absorption, a feat for an oral peptide, it was plagued by high inter-subject pharmacokinetic variability and a lack of dose-proportionality, rendering it clinically unpredictable.[6] This trade-off between the efficacy and predictability of the subcutaneous route and the convenience but unreliability of the oral route likely represented a significant developmental hurdle. Ultimately, despite demonstrating clear pharmacological potency and a favorable safety profile in clinical trials, Acyline was never commercially marketed and remains an investigational compound used for research purposes.[1] Its development history serves as an important case study on the evolution of GnRH antagonists and the persistent challenges of oral peptide drug delivery.

2.0 Drug Identification and Physicochemical Properties

2.1 Nomenclature and Unique Identifiers

The compound is generically known as Acyline.[4] Throughout its development and in scientific literature, it is frequently identified by its primary developmental code name, MER-104.[1] Additional synonyms have been used in specific contexts, including "Acyline oral" to denote its experimental tablet formulation and "GnRH antagonist - Merrion," referencing one of the companies involved in its development.[2]

For unambiguous identification in scientific and regulatory databases, Acyline is assigned several unique identifiers. Its DrugBank Accession Number is DB11906.[4] The Chemical Abstracts Service (CAS) Registry Number for the compound is 170157-13-8.[1] In public chemical databases, it is cataloged under PubChem Compound ID (CID) 16137348 and ChemSpider ID 17293858.[1]

2.2 Chemical Structure, Formula, and Classification

Acyline is classified as a small molecule, synthetic peptide, specifically an oligopeptide or polypeptide containing ten amino acid residues.[2] It is a GnRH analogue, meaning its structure is derived from the native GnRH decapeptide but has been modified to confer antagonist properties.[1] Within pharmacological and therapeutic classification systems, it belongs to the categories of "Gonadotropin-Releasing Hormone, antagonists & inhibitors," "Hormone Antagonists," and "Peptides".[4]

The chemical formula for Acyline is .[4] This corresponds to an average molecular weight of 1533.24 g/mol and a precise monoisotopic mass of 1531.7419207 Da.[4] The elemental analysis of the compound is approximately: C, 62.67%; H, 6.71%; Cl, 2.31%; N, 13.70%; and O, 14.61%.[10]

The structure of Acyline is that of a modified decapeptide with the following amino acid sequence: Ac-D-Nal-D-Cpa-D-Pal-Ser-Aph(Ac)-D-Aph(Ac)-Leu-Lys(iPr)-Pro-D-Ala-NH2.[13] This sequence includes several non-standard or modified amino acids, such as D-Naphthalen-2-ylalanine (D-Nal), D-4-Chlorophenylalanine (D-Cpa), D-Pyridin-3-ylalanine (D-Pal), and acetylated aminophenylalanine (Aph(Ac)), which are critical for its high binding affinity and antagonist activity at the GnRH receptor.[13] For computational and database indexing purposes, its structure is also represented by standard chemical identifiers:

  • InChI: InChI=1S/C80H102ClN15O14/c1-46(2)37-63(72(102)89-62(18-11-12-35-84-47(3)4)80(110)96-36-14-19-70(96)79(109)85-48(5)71(82)101)90-74(104)66(40-53-23-30-60(31-24-53)86-49(6)98)92-76(106)67(41-54-25-32-61(33-26-54)87-50(7)99)94-78(108)69(45-97)95-77(107)68(43-56-15-13-34-83-44-56)93-75(105)65(39-52-21-28-59(81)29-22-52)91- [4]
  • InChI Key: ZWNUQDJANZGVFO-YHSALVGYSA-N [10]
  • SMILES: O=C([C@H]1N(C([C@H](CCCCNC(C)C)NC([C@H](CC(C)C)NC([C@@H](CC2=CC=C(NC(C)=O)C=C2)NC([C@H](CC3=CC=C(NC(C)=O)C=C3)NC([C@H](CO)NC([C@@H](CC4=CC=CN=C4)NC([C@@H](CC5=CC=C(Cl)C=C5)NC([C@@H](CC6=CC=C7C=CC=CC7=C6)NC(C)=O)=O)=O)=O)=O)=O)=O)=O)=O)CCC1)N[C@H](C)C(N)=O [10]

2.3 Formulation, Solubility, and Stability

Acyline was primarily developed for parenteral administration via subcutaneous injection.[1] For this route, the drug was typically supplied as a lyophilized (freeze-dried) powder that was reconstituted with a suitable sterile diluent, such as bacteriostatic water, prior to administration.[1] An experimental oral formulation was also investigated, which consisted of Acyline incorporated into a solid tablet form utilizing the proprietary GIPET® (GI enhancing permeability technology) system, designed to facilitate gastrointestinal absorption of peptides.[1]

Regarding its physicochemical properties, Acyline exhibits poor aqueous solubility, a common characteristic of complex peptides. It is reported to be soluble in organic solvents like dimethyl sulfoxide (DMSO) but not soluble in water.[10] For research and formulation purposes, a salt form, Acyline TFA (trifluoroacetate), is also available, which typically offers enhanced water solubility and stability compared to the free base form.[14]

For long-term preservation, Acyline should be stored in a dry, dark environment at -20°C, under which conditions it has a shelf life of over five years.[10] For short-term storage (days to weeks), temperatures of 0-4°C are acceptable. The compound is stable enough to be shipped at ambient temperature as a non-hazardous chemical.[10]

Table 1: Acyline Identification and Chemical Properties

PropertyValue
Generic NameAcyline
Developmental Code NameMER-104
DrugBank IDDB11906
CAS Number170157-13-8
Chemical Formula
Average Molecular Weight1533.24 g/mol
Drug ClassGnRH Antagonist, Polypeptide, Oligopeptide
Amino Acid SequenceAc-D-Nal-D-Cpa-D-Pal-Ser-Aph(Ac)-D-Aph(Ac)-Leu-Lys(iPr)-Pro-D-Ala-NH2

3.0 Mechanism of Action and Pharmacological Target

3.1 The Gonadotropin-Releasing Hormone Receptor (GnRHR) as the Primary Target

The exclusive molecular target identified for Acyline is the human Gonadotropin-releasing hormone receptor (GnRHR).[2] The GnRHR is a crucial component of the hypothalamic-pituitary-gonadal (HPG) axis, the central regulatory system for reproduction and sex hormone production. This receptor is a G-protein coupled receptor (GPCR) predominantly expressed on the surface of gonadotrope cells in the anterior pituitary gland.[4]

Under normal physiological conditions, the hypothalamus secretes GnRH in a pulsatile manner. This endogenous GnRH binds to and activates the GnRHR, initiating a downstream signaling cascade via a phosphatidylinositol-calcium second messenger system.[4] This activation stimulates the synthesis and subsequent release of the two primary gonadotropic hormones: luteinizing hormone (LH) and follicle-stimulating hormone (FSH).[4] These pituitary hormones then travel through the bloodstream to the gonads (testes in males, ovaries in females), where they stimulate the production of sex steroids (e.g., testosterone and estrogen) and support gametogenesis.[15] The pulsatile nature of GnRH release is essential for maintaining this delicate hormonal balance.[15] Understanding this physiological pathway is fundamental to appreciating the profound and immediate impact of Acyline's inhibitory action.

3.2 Molecular Mechanism: Competitive Antagonism and Endocrine Suppression

Acyline functions as a direct, competitive antagonist of the GnRHR.[4] As a structural analogue of the native GnRH peptide, Acyline binds with high affinity to the same receptor sites on pituitary gonadotropes.[5] However, due to its specific structural modifications, this binding does not trigger the conformational change required for receptor activation and downstream signaling. Instead, it physically occupies the receptor, competitively inhibiting the binding of endogenous GnRH.[5]

This mechanism of competitive inhibition leads to an immediate, profound, and sustained blockade of the GnRH signaling pathway. By preventing GnRH from activating its receptor, Acyline effectively shuts down the primary stimulus for the synthesis and release of LH and FSH from the pituitary.[5] The direct and immediate consequence of this gonadotropin suppression is a rapid cessation of gonadal steroidogenesis. In males, this manifests as a swift decline in serum testosterone levels, which can reach the castrate range (typically defined as

 ng/dL or  nmol/L) within hours to days of administration.[1]

3.3 Comparative Pharmacology within the GnRH Modulator Class

The mechanism of Acyline places it in the class of GnRH antagonists, which are pharmacologically distinct from the other major class of GnRH modulators, the GnRH agonists (e.g., leuprolide, gonadorelin).[5] GnRH agonists also suppress the HPG axis, but they do so through a different, two-phase mechanism. Upon initial administration, agonists bind to and strongly activate the GnRHR, causing an initial surge in LH, FSH, and consequently, testosterone. This "flare effect" can last for days to weeks and can be clinically detrimental in hormone-sensitive conditions like advanced prostate cancer, where a temporary rise in testosterone can exacerbate symptoms.[5] Following this initial stimulation, the continuous presence of the agonist leads to receptor desensitization and downregulation, eventually resulting in a suppressed, hypogonadal state.[5]

Acyline and other GnRH antagonists circumvent this initial flare entirely. Their mechanism of immediate blockade provides a much more rapid and direct path to hormonal suppression.[5] This represents a significant clinical advantage, particularly for indications where an initial hormone surge is undesirable. Acyline's amino acid sequence shows its structural similarity to other marketed peptide GnRH antagonists such as Cetrorelix, Ganirelix, and Degarelix, which are used in indications ranging from assisted reproduction to advanced prostate cancer.[12]

The development of Acyline reflects a clear evolutionary progression within the GnRH antagonist class. The first generation of antagonists, while effective at avoiding the flare effect, were often limited by significant histamine-releasing properties, leading to adverse events like pruritic rashes and injection site reactions, and they possessed short biological half-lives that necessitated frequent, often daily, injections.[5] Acyline was engineered as a "third-generation" antagonist to overcome these specific limitations. The goal was to create a compound with improved potency, a longer duration of action to allow for less frequent dosing, and a minimized potential for histamine-related side effects. Clinical data for Acyline confirm the success of this approach, demonstrating a long half-life and only mild, transient local reactions, positioning it as a pharmacologically optimized agent within its class.[5]

4.0 Non-Clinical Pharmacology and Toxicology

4.1 In Vivo Efficacy in Animal Models

Preclinical studies in various animal models provided the foundational proof-of-concept for Acyline's potent biological activity. These studies consistently demonstrated its ability to effectively suppress the HPG axis. In male dogs, Acyline was shown to be a potent suppressor of both basal and GnRH-stimulated serum testosterone concentrations.[10] In the domestic cat, administration of Acyline during the follicular phase successfully prevented ovulation, confirming its ability to disrupt the female reproductive cycle, although it did not significantly affect luteal function during established pregnancies.[10]

Further mechanistic studies were conducted in genetically modified mouse models. In female Kiss1−/− and Gpr54−/− mice, which have disruptions in the kisspeptin signaling pathway that regulates GnRH, subcutaneous administration of Acyline (50 µg, twice daily for 5 days) resulted in the disruption of the vaginal estrous cycle and a reduction in uterine weights.[14] These physiological changes were accompanied by a decrease in serum LH concentrations, directly confirming Acyline's potent effect on pituitary gonadotropin release even in a model with altered upstream regulation.[14] In corresponding male mouse models, a single subcutaneous dose of Acyline (50 µg) was sufficient to cause a significant reduction in serum FSH concentrations.[14] Collectively, these animal studies established Acyline as a highly effective GnRH antagonist

in vivo, validating its mechanism of action and supporting its progression into human clinical trials.

4.2 In Vitro Receptor Binding and Functional Assays

The potent in vivo effects of Acyline are underpinned by its high affinity and functional antagonist activity at the molecular level, as demonstrated in in vitro assays. Studies utilizing human embryonic kidney (HEK293) cells engineered to express the human GnRH receptor provided quantitative measures of its potency. In a reporter gene assay designed to measure the inhibition of a GnRH-induced luciferase response, Acyline demonstrated a half-maximal inhibitory concentration () of 0.52 nM.[4] A separate assay, which measured the inhibition of GnRH-induced LH secretion, reported a similarly potent

 value of 0.69 nM.[14] These sub-nanomolar values indicate that Acyline is a highly potent functional antagonist, capable of blocking receptor signaling at very low concentrations.

In addition to functional antagonism, direct binding affinity to the GnRHR was also quantified. Binding studies reported a dissociation constant () of 2.5 nM.[4] The

 value represents the concentration of the drug required to occupy 50% of the receptors at equilibrium, with lower values indicating a higher binding affinity. This result confirms that Acyline binds strongly and tightly to its molecular target, which is a prerequisite for its potent and sustained biological effects.

4.3 Preclinical Safety Profile

The available documentation lacks detailed reports from formal, good laboratory practice (GLP) compliant preclinical toxicology studies, such as comprehensive assessments of cardiotoxicity, hepatotoxicity, or genotoxicity.[20] This represents a notable gap in the publicly accessible data for Acyline. However, inferences about its general safety can be drawn from the reported animal efficacy studies and early human trials.

In the non-clinical efficacy studies conducted in monkeys, dogs, cats, and mice, no significant or unexpected adverse effects were reported beyond the intended and predictable pharmacological consequences of profound gonadotropin and sex steroid suppression.[1] The primary adverse events observed in subsequent human trials were mild, transient injection site reactions.[7] While not explicitly detailed in the provided materials, it is standard practice for such local tolerability to be evaluated in preclinical animal models (e.g., in rabbits or minipigs) before human administration. The absence of reports of severe systemic toxicity in the animal studies suggests that Acyline possessed a favorable preliminary safety profile, which was a necessary prerequisite for its advancement into clinical development.

5.0 Clinical Pharmacology: Pharmacokinetics and Pharmacodynamics

5.1 Pharmacokinetics (PK)

5.1.1 Subcutaneous Administration Profile

The pharmacokinetic profile of Acyline following subcutaneous (s.c.) injection is characterized by rapid absorption and a remarkably long elimination half-life, which is the basis for its sustained pharmacodynamic effect. In a clinical study involving healthy male volunteers, a single s.c. injection of 300 µg/kg resulted in a peak serum concentration () of  ng/mL.[7] This peak was reached approximately 90 minutes (

) after administration, indicating swift entry into the systemic circulation.[7] The most notable feature of its subcutaneous profile is its long terminal elimination half-life (

), which was calculated to be 4.9 days.[7] This extended half-life allows the drug to remain at therapeutic concentrations for a prolonged period, enabling less frequent dosing.

Data from a lower dose of 75 µg/kg s.c. showed a similar pattern of rapid absorption, with a  of  ng/mL occurring at a  of 1 hour.[5] Following this dose, serum Acyline levels remained significantly elevated above baseline for at least 7 days and did not return to background levels until 14 to 17 days post-injection, further corroborating its long-acting nature.[5]

5.1.2 Oral Administration Profile (GIPET®-Enhanced Formulation)

In an effort to develop a more convenient dosage form, an experimental oral tablet of Acyline was created using the GIPET® permeability enhancement technology. A single-dose pharmacokinetic study in eight healthy men evaluated doses of 10, 20, and 40 mg.[6] The results demonstrated that the GIPET® system successfully facilitated gastrointestinal absorption of the peptide, with mean serum concentrations rising immediately after dosing for all three dose levels.[6]

However, the pharmacokinetic profile of the oral formulation was starkly different from the subcutaneous route and was marked by extreme variability. The time to peak concentration () was generally rapid, with mean values of  hours for the 10 mg dose and  hours for the 40 mg dose.[6] The peak concentrations (

) and overall exposure (Area Under the Curve, AUC) showed no clear dose-proportionality and were characterized by very large standard deviations, indicating a high degree of inter-subject variability.[6] For instance, the mean

 for the 40 mg dose ( ng/mL) was unexpectedly lower than that for the 20 mg dose ( ng/mL).[6] The terminal half-life for the oral formulation was also significantly shorter than the subcutaneous route, ranging from 7.4 to 10.7 hours.[6] Serum Acyline was undetectable in most subjects by 48 hours post-dose.[6]

This high degree of pharmacokinetic variability is a critical finding. While the GIPET® technology succeeded in overcoming the fundamental barrier of peptide absorption, it failed to deliver the drug in a predictable or consistent manner. For a potent hormonal agent like Acyline, where maintaining a specific therapeutic window is crucial for both efficacy (ensuring sufficient testosterone suppression) and safety (avoiding unintended consequences of over- or under-dosing), such erratic pharmacokinetics would be a major clinical and regulatory liability. The need for frequent dosing and the potential requirement for therapeutic drug monitoring to manage this variability would negate the convenience that the oral route was intended to provide, likely contributing to the discontinuation of this development path.

5.1.3 Analysis of Absorption, Distribution, Metabolism, and Elimination (ADME)

Detailed information regarding the broader ADME properties of Acyline is limited. Key data points for metabolism pathways, protein binding, volume of distribution, and routes of elimination are explicitly noted as "Not Available" in the DrugBank database.[4] This lack of comprehensive ADME data is common for investigational compounds that do not advance to late-stage regulatory submission. As a peptide-based therapeutic, it is reasonable to infer that Acyline is likely metabolized through proteolysis by peptidases in the plasma and tissues into smaller, inactive peptide fragments and constituent amino acids, which are then cleared or recycled. However, without specific experimental data, this remains a well-founded but unconfirmed hypothesis.

Table 2: Summary of Clinical Pharmacokinetic Parameters for Acyline

ParameterSubcutaneous (300 µg/kg)Oral (10 mg)Oral (20 mg)Oral (40 mg)
(ng/mL)
(hours)1.5
(hours)117.6 (4.9 days)
(ng·h/mL)Not Reported

Data are presented as mean ± standard deviation where available. Data sourced from.[6]

5.2 Pharmacodynamics (PD)

5.2.1 Dose-Response Relationship on Gonadotropin Suppression

The pharmacodynamic effects of Acyline are a direct consequence of its mechanism of action, resulting in potent suppression of pituitary gonadotropins. Clinical studies with subcutaneous administration established a clear dose-dependent relationship.[1] In the first-in-human study, escalating single doses produced progressively deeper suppression of both LH and FSH. At the highest tested dose in that study (75 µg/kg), LH levels were suppressed to a nadir of approximately 12.4% of baseline, while FSH was suppressed to 46.9% of baseline.[5] The consistently less pronounced suppression of FSH relative to LH is a known class effect for GnRH antagonists. This phenomenon is attributed to the significantly longer serum half-life of FSH (approximately 3-4 hours) compared to LH (approximately 60-80 minutes), as well as potentially different regulatory mechanisms for FSH synthesis and release.[5]

The experimental oral formulation also demonstrated effective gonadotropin suppression. Across all tested doses (10, 20, and 40 mg), serum LH and FSH were significantly suppressed, reaching a nadir approximately 12 hours after administration.[9] At this time point, the average suppression of LH was profound at approximately 70%, while FSH suppression was more modest at around 28%, again highlighting the differential effect on the two gonadotropins.[6]

5.2.2 Impact on Serum Testosterone and Duration of Action

The suppression of serum testosterone, the ultimate pharmacodynamic endpoint for many of Acyline's intended indications, closely mirrored the suppression profile of LH.[6] Following subcutaneous administration, the decline in testosterone was rapid and profound. A single high dose of 300 µg/kg s.c. was sufficient to suppress serum testosterone to castrate levels, and this profound suppression was maintained for an extended duration of 15 days.[7] This long duration of action is a key pharmacological feature of Acyline, supporting its potential for convenient, infrequent dosing regimens (e.g., twice-monthly). Furthermore, studies using serial injections of a lower dose (75 µg/kg every two days for five doses) demonstrated that the duration of testosterone suppression could be extended to over 20 days, showcasing the flexibility in maintaining a hypogonadal state.[7]

In contrast, the oral GIPET®-enhanced formulation produced a much more acute and transient effect on testosterone. Significant suppression was observed between 6 and 12 hours after dosing with all dose levels.[9] The higher doses of 20 mg and 40 mg were able to sustain testosterone suppression for a period of 12 to 24 hours.[9] This shorter duration of action is consistent with the much shorter pharmacokinetic half-life of the oral formulation.

5.2.3 Hormonal Recovery Post-Administration

A critical aspect of Acyline's pharmacodynamic profile is the reversibility of its effects upon drug clearance. Following the acute suppression induced by the oral formulation, all hormone concentrations (LH, FSH, and testosterone) returned to baseline levels within 48 hours of administration, demonstrating a rapid and complete recovery of the HPG axis.[9]

Recovery after the longer-acting subcutaneous administration was also demonstrated. In a study involving stallions, gonadotropin and testosterone levels returned to control values within nine days after the discontinuation of treatment.[25] In human studies with single high doses, hormone levels began to rise after the drug concentration fell below a therapeutic threshold, with a full return to baseline occurring over the subsequent weeks, consistent with the drug's 4.9-day half-life.[7] This predictable and reversible suppression is a crucial feature for indications like contraception, where a return to normal fertility is desired after treatment cessation.

6.0 Clinical Development and Efficacy

6.1 Overview of the Acyline Clinical Trial Program

The clinical development of Acyline was extensive, exploring its utility across a range of endocrine and metabolic indications. The program was anchored by its potent ability to induce a reversible state of medical castration.[2] The most advanced and primary focus of its development was for male hormonal contraception, for which the drug progressed to Phase 3 trials.[2]

Beyond contraception, Acyline was investigated in Phase 2 clinical trials for several hormone-dependent diseases. These included androgen-dependent conditions such as advanced prostatic cancer and precocious puberty.[2] The program also branched into metabolic disorders, with Phase 2 studies initiated for Type 2 diabetes mellitus, insulin resistance, and obesity, likely to explore the effects of profound sex steroid modulation on metabolic parameters.[2] Additionally, it was evaluated in Phase 2 for androgen deficiency and Phase 1 for hypogonadism.[2]

Key clinical trials registered on clinicaltrials.gov, such as NCT00161447 ("Male Hormonal Contraceptive Development-ACY-5") and NCT00156650, focused on dose-finding, pharmacokinetics, and pharmacodynamics in healthy male volunteers, often in combination with exogenous testosterone to mitigate the symptoms of hypogonadism.[8]

6.2 Analysis of Investigated Indications

6.2.1 Male Hormonal Contraception

This was the lead indication for Acyline, leveraging its ability to induce a profound and reversible suppression of spermatogenesis through the shutdown of the HPG axis.[2] The fundamental principle of hormonal male contraception is to suppress intratesticular testosterone to levels that cannot support sperm production, which requires potent suppression of LH and FSH. Clinical studies with Acyline demonstrated that it could achieve this goal effectively. A single 300 µg/kg subcutaneous dose was shown to reduce testosterone to castrate levels for 15 days, and repeated lower doses could extend this period to over 21 days.[7] This duration of action made a twice-monthly or monthly injection schedule a clinically feasible and convenient regimen.[7] To counteract the systemic side effects of low testosterone (e.g., decreased libido, loss of bone mineral density), contraceptive regimens involving Acyline were often co-administered with an exogenous androgen, such as testosterone, which maintains normal systemic functions without restoring spermatogenesis.[26]

6.2.2 Management of Androgen Deficiency and Hypogonadism

Acyline's investigation for hypogonadism (Phase 1) and androgen deficiency (Phase 2) appears paradoxical, as the drug itself induces a hypogonadal state.[2] However, this approach is used in certain clinical scenarios where precise control over the hormonal milieu is desired. By using Acyline to completely shut down the body's own erratic or insufficient endogenous testosterone production, clinicians can then administer a stable, controlled dose of exogenous testosterone. This "block and replace" strategy can provide more consistent serum testosterone levels than supplementation alone, potentially offering better symptom management for some patients.

6.2.3 Therapeutic Potential in Hormone-Dependent Diseases

The potent anti-androgenic effect of Acyline makes it a strong candidate for treating diseases driven by testosterone. It was investigated in Phase 2 trials for advanced prostate cancer, where androgen deprivation therapy (ADT) is a cornerstone of treatment.[2] For this indication, the rapid onset of action and avoidance of the testosterone flare associated with GnRH agonists would be significant clinical advantages.[5] The development of the experimental oral formulation was specifically hypothesized for use in prostate cancer, aiming to provide a more convenient alternative to long-acting injections.[6]

The same principle of hormone suppression applies to other conditions. Acyline reached Phase 2 for precocious puberty, a condition where premature activation of the HPG axis leads to early physical development.[2] By blocking the GnRH receptor, Acyline could effectively halt this premature puberty. Other GnRH modulators are standard of care for this condition, as well as for other gynecological diseases like endometriosis, for which Acyline also held theoretical potential.[18]

6.3 Synthesis of Efficacy Findings Across Clinical Studies

The primary measure of efficacy across the majority of Acyline's clinical studies was its pharmacodynamic effect: the degree and duration of hormonal suppression. The data from these trials are remarkably consistent in demonstrating that Acyline is a highly effective and potent suppressor of the HPG axis.[7] The first-in-human study successfully established a clear dose-response relationship for subcutaneous administration, showing that increasing doses led to deeper and more prolonged suppression of LH, FSH, and testosterone.[5] Subsequent studies with higher doses confirmed that a single injection could maintain testosterone at castrate levels for over two weeks, a duration sufficient for practical therapeutic and contraceptive regimens.[7] The efficacy of the oral formulation was demonstrated in its ability to produce acute, albeit transient, hormonal suppression.[9] While clinical outcome data (e.g., rates of contraception, prostate cancer progression) are not detailed in the provided materials, the consistent and profound hormonal suppression achieved by Acyline represents the successful fulfillment of its primary efficacy endpoint.

Table 3: Overview of Acyline's Clinical Development Program by Indication

Therapeutic AreaIndicationHighest Phase ReachedCountry/Location
ContraceptionContraceptionPhase 3-
EndocrinologyAndrogen DeficiencyPhase 2United States
Metabolic DiseaseDiabetes Mellitus, Type 2Phase 2United States
Metabolic DiseaseInsulin ResistancePhase 2United States
Metabolic DiseaseObesityPhase 2United States
OncologyProstatic CancerPhase 2United States
EndocrinologyPuberty, PrecociousPhase 2United States
EndocrinologyHypogonadismPhase 1United States

Table constructed from data in.[2]

7.0 Safety and Tolerability Profile

7.1 Analysis of Adverse Events from Clinical Trials

Across multiple clinical trials in healthy male volunteers, Acyline demonstrated a favorable safety and tolerability profile, with adverse events being generally mild and predictable.[7] In the study of the GIPET®-enhanced oral formulation, there were no treatment-related serious adverse events reported.[9] Importantly, routine laboratory safety assessments, including liver function tests (e.g., ALT, AST) and measures of renal function (e.g., creatinine), were unaffected by treatment, suggesting a lack of direct organ toxicity at the doses studied.[9]

7.2 Local Tolerability and Injection Site Reactions

The most frequently reported adverse events associated with Acyline were mild, localized, and transient reactions at the site of subcutaneous injection.[7] These reactions typically consisted of erythema (a blush or redness of the skin) and pruritus (itching).[7] These events were described as infrequent and were self-limiting, generally resolving within 90 minutes of the injection.[5] The mild nature of these reactions is a significant finding, as earlier-generation GnRH antagonists were often associated with more severe, histamine-mediated injection site reactions that limited their clinical utility.[5] The improved local tolerability of Acyline was a key feature supporting its development as a "third-generation" antagonist.

7.3 Systemic Side Effects Attributable to Pharmacological Action

The systemic side effects observed during treatment with Acyline were not unexpected toxicities but were rather the direct and predictable physiological consequences of its potent pharmacological action—the induction of a hypogonadal state.[8] By suppressing testosterone to castrate levels, Acyline produced symptoms consistent with androgen deprivation. These included decreased libido, fatigue, hot flashes (vasomotor symptoms), mood changes or irritability, and myalgia (muscle pain).[8]

These are well-characterized class effects for all potent GnRH modulators (both agonists and antagonists) that lead to medical castration. In the context of its development for male contraception, these side effects would be managed by the co-administration of exogenous testosterone, which would maintain normal physiological functions while the HPG axis remained suppressed.[26] For therapeutic indications like prostate cancer, these side effects are an accepted part of androgen deprivation therapy.

8.0 Regulatory and Developmental History

8.1 Chronology of Development and Key Milestones

Acyline's journey as an investigational drug began with its initial synthesis and characterization at the Salk Institute.[1] It entered the clinical development phase in the early 2000s, with the first peer-reviewed publication of a human study appearing in July 2002.[1] This pivotal study established its dose-dependent efficacy and safety profile in healthy men.[5] Subsequent research focused on optimizing its therapeutic potential. A key study published in 2004 investigated higher doses and repeat-dosing regimens, demonstrating that Acyline's long-acting properties could sustain testosterone suppression for over two weeks, making it a viable candidate for a twice-monthly injectable.[1]

Recognizing the limitations of parenteral administration for chronic use, development efforts then pivoted towards creating a more convenient oral dosage form. This culminated in a 2009 publication detailing the pharmacokinetics and pharmacodynamics of an experimental GIPET®-enhanced oral tablet.[1] Companies such as Merrion Pharmaceuticals were involved in this later stage of development, particularly focusing on the oral formulation for indications like prostate cancer.[10]

8.2 Regulatory Status: An Unapproved Investigational Agent

Despite its progression through a comprehensive clinical trial program, including reaching Phase 3 for its lead indication, Acyline was never marketed and has not received regulatory approval in any jurisdiction.[1] There is no public record of a New Drug Application (NDA) being submitted to the U.S. Food and Drug Administration (FDA) or a Marketing Authorisation Application (MAA) to the European Medicines Agency (EMA). A thorough review of FDA and EMA approval databases, press releases, and drug evaluation reports reveals no mention of Acyline, which stands in stark contrast to the readily available approval documentation for other drugs, including other GnRH modulators.[17] Consequently, Acyline remains an investigational compound. Its availability is restricted to research purposes only, supplied by specialized chemical and life science companies.[10]

8.3 Reasons for Discontinuation of Commercial Development

The provided materials do not contain an explicit, official statement detailing the reasons for the cessation of Acyline's commercial development. However, a critical analysis of the available clinical data and the broader pharmaceutical landscape allows for the formulation of several well-supported hypotheses.

First, and perhaps most critically, were the pharmacokinetic challenges of the oral formulation. The GIPET®-enhanced tablet, while a technical success in achieving bioavailability, exhibited high inter-subject variability and a lack of predictable dose-proportionality.[6] For a potent hormone-suppressing agent, this level of unpredictability is a significant clinical and regulatory flaw. The inability to ensure consistent therapeutic drug levels from patient to patient would compromise both efficacy and safety, making it difficult to gain regulatory approval and clinical acceptance. This failure to develop a reliable, convenient oral alternative to injections may have been a major blow to the program's long-term viability.

Second, the commercial viability and competitive landscape likely played a significant role. By the time Acyline was in later-stage development, the market for GnRH modulators for indications like prostate cancer was already well-established and highly competitive. It was dominated by long-acting GnRH agonists like leuprolide (Lupron Depot) and was seeing the entry of other GnRH antagonists like degarelix.[13] Acyline would have needed to demonstrate a substantial clinical advantage or a significant cost benefit to capture market share from these entrenched competitors. Its lead indication, male hormonal contraception, has also historically been a commercially challenging and high-risk market for pharmaceutical companies to enter, with complex societal and economic barriers to adoption.

Finally, the discontinuation may have been the result of a strategic shift within the developing company. Pharmaceutical development is resource-intensive, and companies often have to make difficult decisions to prioritize their pipelines. Merrion Pharmaceuticals or other stakeholders may have decided to allocate resources to other assets perceived to have a higher probability of success or a more favorable risk-reward profile, leading to the deprioritization and eventual termination of the Acyline program. It is likely that a combination of these technical, commercial, and strategic factors contributed to the decision to halt the development of Acyline before it could reach the market.

9.0 Expert Analysis and Future Perspectives

9.1 Critical Assessment of Acyline's Therapeutic Potential and Limitations

From a purely pharmacological and clinical-pharmacological perspective, Acyline stands as a potent and effective GnRH antagonist. The data from its clinical program unequivocally demonstrate its ability to induce rapid, profound, and sustained suppression of the HPG axis.[7] Its development as a third-generation antagonist successfully addressed key limitations of its predecessors, namely the problematic histamine-releasing side effects and short duration of action.[5] The achievement of a multi-day half-life following subcutaneous injection was a significant advancement, offering the potential for a much more convenient dosing schedule (e.g., twice-monthly) compared to daily injections, thereby improving the prospects for patient compliance in chronic treatment settings.[7]

However, Acyline's primary limitation was inextricably linked to its chemical nature as a peptide. Like most peptides, it suffered from poor oral bioavailability, necessitating parenteral administration.[1] While injections are acceptable for many therapeutic contexts, the lack of a viable oral option constrained its potential market and convenience, particularly for chronic conditions where patients strongly prefer oral medications. The ambitious attempt to overcome this limitation, while scientifically interesting, ultimately introduced a new and perhaps more fatal flaw: pharmacokinetic unpredictability.[6]

9.2 The Challenge of Peptide Drug Delivery and the GIPET® Experiment

Acyline's development history serves as a compelling case study on the formidable challenges of oral peptide drug delivery—a "holy grail" of pharmaceutical science. The GIPET®-enhanced oral formulation represented a sophisticated attempt to solve this multi-billion-dollar problem by using medium-chain fatty acids to transiently open tight junctions in the intestinal epithelium, allowing the large peptide molecule to pass into the bloodstream.[6]

The clinical trial of this formulation yielded a crucial lesson: achieving systemic absorption is only the first step. The ultimate clinical utility of an oral drug depends not just on whether it is absorbed, but on how reliably it is absorbed. The Acyline oral study demonstrated that even with an advanced permeation enhancer, the inherent variability of the human gastrointestinal tract (e.g., differences in transit time, pH, food effects, and mucosal state between individuals) can lead to highly erratic drug exposure.[6] This pharmacokinetic variability rendered the oral formulation clinically impractical. The story of oral Acyline is a cautionary tale that underscores the immense difficulty of translating oral peptide delivery technologies from controlled laboratory settings to the complex and variable reality of human physiology. It highlights that for a technology to be successful, it must deliver not just bioavailability, but also predictability and consistency.

9.3 Concluding Remarks on Acyline's Place in Endocrine Research

The trajectory of Acyline's development exemplifies the concept of the pharmaceutical "valley of death," where a compound with excellent scientific credentials and promising early clinical data fails to transition into a commercial product. Acyline possessed potent in vitro activity, demonstrated clear efficacy in animal models, and showed robust, safe, and long-lasting hormonal suppression in human trials via its injectable formulation.[4] It successfully met its primary pharmacological objectives.

However, the program faltered when faced with subsequent developmental hurdles. The pursuit of a superior delivery system led to a technically flawed oral formulation that introduced unacceptable clinical variability.[6] Concurrently, it faced a challenging commercial landscape with entrenched competitors in the prostate cancer market and a historically difficult-to-penetrate male contraception market.[13] This confluence of a critical technical setback in its next-generation formulation and a high-risk commercial environment likely sealed the program's fate, despite the intrinsic pharmacological merit of the molecule.

In conclusion, while Acyline never reached the pharmacy shelf, its contribution to endocrine research and pharmaceutical science is significant. It represents an important milestone in the evolution of GnRH antagonists, demonstrating that a long-acting, well-tolerated profile was achievable. The clinical data generated from its trials have provided valuable insights into the long-term suppression of the HPG axis and the physiological effects of profound hypogonadism. Furthermore, its development journey, particularly the experiment with the GIPET® oral formulation, offers enduring lessons on the complexities and high failure rates inherent in developing novel drug delivery systems for peptides. Acyline thus remains a valuable tool for researchers and a benchmark compound in the ongoing quest to modulate the GnRH system and conquer the challenges of peptide therapeutics.

Works cited

  1. Acyline - Wikipedia, accessed September 30, 2025, https://en.wikipedia.org/wiki/Acyline
  2. Acyline - Drug Targets, Indications, Patents - Patsnap Synapse, accessed September 30, 2025, https://synapse.patsnap.com/drug/e26e891378804a3d9cd429c6e007fd77
  3. en.wikipedia.org, accessed September 30, 2025, [https://en.wikipedia.org/wiki/Acyline#:~:text=Acyline%20(developmental%20code%20name%20MER,and%20testosterone%20levels%20in%20men.](https://www.google.com/url?q=https://en.wikipedia.org/wiki/Acyline%23:~:text%3DAcyline%2520(developmental%2520code%2520name%2520MER,and%2520testosterone%2520levels%2520in%2520men.&sa=D&source=editors&ust=1759222964593297&usg=AOvVaw1x63eNpWwY80xdW3VwqET2)
  4. Acyline: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed September 30, 2025, https://go.drugbank.com/drugs/DB11906
  5. Acyline: The First Study in Humans of a Potent, New Gonadotropin ..., accessed September 30, 2025, https://academic.oup.com/jcem/article/87/7/3215/2846910
  6. Oral administration of the GnRH antagonist acyline, in a GIPET ..., accessed September 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2721900/
  7. A single dose of the potent gonadotropin-releasing hormone antagonist acyline suppresses gonadotropins and testosterone for 2 weeks in healthy young men - PubMed, accessed September 30, 2025, https://pubmed.ncbi.nlm.nih.gov/15579744/
  8. Acyline induces long-term gonadotropin, testosterone suppression - | BioWorld, accessed September 30, 2025, https://www.bioworld.com/articles/581308-acyline-induces-long-term-gonadotropin-testosterone-suppression
  9. (PDF) Oral administration of the GnRH antagonist acyline, in a GIPET ®-enhanced tablet form, acutely suppresses serum testosterone in normal men: Single-dose pharmacokinetics and pharmacodynamics - ResearchGate, accessed September 30, 2025, https://www.researchgate.net/publication/26250329_Oral_administration_of_the_GnRH_antagonist_acyline_in_a_GIPET_R-enhanced_tablet_form_acutely_suppresses_serum_testosterone_in_normal_men_Single-dose_pharmacokinetics_and_pharmacodynamics
  10. Acyline | MER-104 | CAS#170157-13-8 | GnRH antagonist | MedKoo Biosciences, accessed September 30, 2025, https://www.medkoo.com/products/5180
  11. Acyline oral - Merrion - AdisInsight - Springer, accessed September 30, 2025, https://adisinsight.springer.com/drugs/800025851
  12. Gonadotropin-Releasing Hormone, antagonists & inhibitors - DrugBank, accessed September 30, 2025, https://go.drugbank.com/categories/DBCAT001838
  13. Gonadotropin-releasing hormone antagonist - Wikipedia, accessed September 30, 2025, https://en.wikipedia.org/wiki/Gonadotropin-releasing_hormone_antagonist
  14. Acyline | GnRH Antagonist | MedChemExpress, accessed September 30, 2025, https://www.medchemexpress.com/acyline.html
  15. Gonadorelin: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed September 30, 2025, https://go.drugbank.com/drugs/DB00644
  16. Acyline: The First Study in Humans of a Potent, New Gonadotropin-Releasing Hormone Antagonist | Request PDF - ResearchGate, accessed September 30, 2025, https://www.researchgate.net/publication/11271667_Acyline_The_First_Study_in_Humans_of_a_Potent_New_Gonadotropin-Releasing_Hormone_Antagonist
  17. FDA approves 6-month formulation of GnRH agonist - Urology Times, accessed September 30, 2025, https://www.urologytimes.com/view/fda-approves-6-month-formulation-gnrh-agonist
  18. (PDF) A Single Dose of the Potent Gonadotropin-Releasing Hormone Antagonist Acyline Suppresses Gonadotropins and Testosterone for 2 Weeks in Healthy Young Men - ResearchGate, accessed September 30, 2025, https://www.researchgate.net/publication/8147636_A_Single_Dose_of_the_Potent_Gonadotropin-Releasing_Hormone_Antagonist_Acyline_Suppresses_Gonadotropins_and_Testosterone_for_2_Weeks_in_Healthy_Young_Men
  19. The GnRH antagonist acyline prevented ovulation, but did not affect ovarian follicular development or gestational corpora lutea in the domestic cat - PubMed, accessed September 30, 2025, https://pubmed.ncbi.nlm.nih.gov/20171723/
  20. Predictive Toxicity: Toxicokinetics Evaluation in Preclinical Studies - porsolt, accessed September 30, 2025, https://www.porsolt.com/predictive-toxicity/
  21. Guidance for Industry - S6 Preclinical Safety Evaluation of Biotechnology-Derived Pharmaceuticals - FDA, accessed September 30, 2025, https://www.fda.gov/media/72028/download
  22. Preclinical Toxicology of New Drugs - DTIC, accessed September 30, 2025, https://apps.dtic.mil/sti/tr/pdf/ADA203402.pdf
  23. Acyline: the first study in humans of a potent, new gonadotropin-releasing hormone antagonist - PubMed, accessed September 30, 2025, https://pubmed.ncbi.nlm.nih.gov/12107227/
  24. Oral administration of the GnRH antagonist acyline, in a GIPET-enhanced tablet form, acutely suppresses serum testosterone in normal men: single-dose pharmacokinetics and pharmacodynamics - PubMed, accessed September 30, 2025, https://pubmed.ncbi.nlm.nih.gov/19479252/
  25. Acyline: The First Study in Humans of a Potent, New Gonadotropin-Releasing Hormone Antagonist | Request PDF - ResearchGate, accessed September 30, 2025, https://www.researchgate.net/publication/274440152_Acyline_The_First_Study_in_Humans_of_a_Potent_New_Gonadotropin-Releasing_Hormone_Antagonist
  26. Testosterone Completed Phase 1 / 2 Trials for Contraception Prevention | DrugBank Online, accessed September 30, 2025, https://go.drugbank.com/drugs/DB00624/clinical_trials?conditions=DBCOND0020891&phase=1%2C2&purpose=prevention&status=completed
  27. Achieve Life Sciences Announces FDA Acceptance of Cytisinicline New Drug Application for Treatment of Nicotine Dependence for Smoking Cessation, accessed September 30, 2025, https://ir.achievelifesciences.com/news-events/press-releases/detail/238/achieve-life-sciences-announces-fda-acceptance-of-cytisinicline-new-drug-application-for-treatment-of-nicotine-dependence-for-smoking-cessation
  28. Achieve Life Sciences Announces Submission of NDA to FDA for Cytisinicline as a Treatment of Nicotine Dependence for Smoking Cessation, accessed September 30, 2025, https://ir.achievelifesciences.com/news-events/press-releases/detail/229/achieve-life-sciences-announces-submission-of-ndato-fda-for-cytisinicline-as-a-treatment-of-nicotine-dependence-for-smoking-cessation
  29. FDA approves axatilimab-csfr for chronic graft-versus-host disease, accessed September 30, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-axatilimab-csfr-chronic-graft-versus-host-disease
  30. ADSTILADRIN - FDA, accessed September 30, 2025, https://www.fda.gov/vaccines-blood-biologics/cellular-gene-therapy-products/adstiladrin
  31. FDA D.I.S.C.O. Burst Edition: FDA approval of Adstiladrin (nadofaragene firadenovec-vncg) for patients with high-risk Bacillus Calmette-Guérin unresponsive non-muscle invasive bladder cancer with carcinoma in situ with or without papillary tumors, accessed September 30, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-disco-burst-edition-fda-approval-adstiladrin-nadofaragene-firadenovec-vncg-patients-high-risk
  32. EMA Backs 14 New Drugs, Confirms Atropine Rejection For Myopia, accessed September 30, 2025, https://insights.citeline.com/pink-sheet/product-reviews/eu-chmp/ema-backs-14-new-drugs-confirms-atropine-rejection-for-myopia-K542SMA5OJGBVKCW54GBULCPNU/
  33. European Medicines Agency (EMA), accessed September 30, 2025, https://www.ema.europa.eu/en/medicines
  34. Medicines for human use under evaluation | European Medicines Agency (EMA), accessed September 30, 2025, https://www.ema.europa.eu/en/medicines/medicines-human-use-under-evaluation
  35. New Treatment Options In Oncology: FDA And EMA Drug Approvals In Q4 2022, accessed September 30, 2025, https://www.aptitudehealth.com/oncology-news/fda-ema-drug-approvals-q4-2022/
  36. Gonadotropin-releasing hormone antagonists versus standard ..., accessed September 30, 2025, https://bmjopen.bmj.com/content/5/11/e008217
  37. ADT With GnRH Antagonists in Prostate Cancer - YouTube, accessed September 30, 2025, https://www.youtube.com/watch?v=NSXo6O7P_f4

Published at: September 30, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.