MedPath

Lutropin alfa Advanced Drug Monograph

Published:Aug 6, 2025

Generic Name

Lutropin alfa

Brand Names

Luveris, Pergoveris

Drug Type

Biotech

CAS Number

152923-57-4

A Comprehensive Monograph on Lutropin alfa (r-hLH): From Molecular Design to Clinical Application and Regulatory Landscape

Section 1: Introduction and Executive Summary

1.1. Overview of Lutropin alfa

Lutropin alfa is a highly purified preparation of recombinant human luteinizing hormone (r-hLH), a critical glycoprotein hormone in human reproduction.[1] As a product of modern biotechnology, it represents a significant advancement over earlier gonadotropin preparations derived from human sources. It is engineered to be structurally and functionally identical to the native luteinizing hormone (LH) produced by the pituitary gland.[2] Its primary role in medicine is as a targeted therapeutic agent for specific subsets of female infertility, where it serves to replace or supplement deficient endogenous LH activity. The development of Lutropin alfa provided clinicians with a preparation of defined LH activity, characterized by a consistent isoform profile and high batch-to-batch reliability, thereby enabling more precise and predictable ovarian stimulation protocols.[4]

1.2. Core Therapeutic Rationale

The central therapeutic application of Lutropin alfa is rooted in the fundamental physiology of ovarian follicular development, specifically the "two-cell, two-gonadotropin" hypothesis. This model posits that both follicle-stimulating hormone (FSH) and LH are indispensable for the production of estradiol, the key steroid hormone driving follicular maturation. Consequently, Lutropin alfa is not used as a monotherapy but is indicated for co-administration with a recombinant FSH preparation, most notably follitropin alfa.[5] Its use is specifically reserved for women diagnosed with severe LH and FSH deficiency, a condition known as hypogonadotropic hypogonadism, which renders them unable to achieve adequate follicular development with FSH stimulation alone.[3] By providing exogenous LH activity, Lutropin alfa restores the necessary physiological synergy with FSH, enabling the stimulation of follicular growth and maturation in this well-defined patient population.

1.3. Executive Summary of Key Findings

This monograph provides an exhaustive analysis of Lutropin alfa, synthesizing its biochemical, pharmacological, clinical, and regulatory characteristics. The key findings are summarized as follows:

  • Biochemical Profile: Lutropin alfa is a heterodimeric glycoprotein produced via recombinant DNA technology in Chinese Hamster Ovary (CHO) cells. This manufacturing process ensures exceptional purity (>99%) and a consistent isoform profile, which are key advantages over the inherent variability and potential for protein contaminants in older, urinary-derived gonadotropin preparations.[2]
  • Pharmacological Action: The drug functions as a specific agonist of the luteinizing hormone/choriogonadotropin receptor (LHCGR). Its mechanism of action is to stimulate ovarian theca cells to produce androgens, which are subsequently converted to estradiol by FSH-stimulated granulosa cells. This action is the cornerstone of the "two-cell, two-gonadotropin" model and is essential for normal follicular steroidogenesis and development.[2]
  • Clinical Application and Efficacy: Clinical evidence robustly supports the efficacy of Lutropin alfa for its niche indication: stimulating follicular development in women with severe LH deficiency. However, the translation of this surrogate endpoint into statistically significant improvements in ultimate clinical outcomes, such as pregnancy and live birth rates, proved challenging to demonstrate in clinical trials, a factor that profoundly influenced its regulatory trajectory.[10]
  • Regulatory Dichotomy: Lutropin alfa has experienced markedly different regulatory fates in major markets. It has maintained marketing authorization in the European Union (EU) since 2000 under the brand name Luveris®, where it is an established treatment for its indicated population. In contrast, in the United States (US), it received an accelerated approval in 2004 that was subsequently withdrawn in 2016. This withdrawal was not due to safety or efficacy concerns but resulted from the logistical infeasibility of completing a required postmarketing trial in the rare patient population.[11]
  • Market Evolution and Strategic Development: A pivotal evolution in the product's lifecycle has been the development and marketing of Pergoveris®, a fixed-dose combination of Lutropin alfa and follitropin alfa. This product simplifies the treatment regimen, enhances patient convenience, and represents a strategic shift toward positioning the therapy for a broader patient profile of "poor ovarian responders" in addition to its original orphan indication.[3]

Section 2: Physicochemical Properties and Molecular Profile

2.1. Identification and Nomenclature

Lutropin alfa is identified through a standardized set of chemical and regulatory classifiers that define its unique identity as a biopharmaceutical agent.

  • Generic Name: Lutropin alfa. The synonym "Lutropin alpha" is also frequently used.[16]
  • Brand Names: The primary brand name for the single-agent product is Luveris®. It is also a key active component of the combination product Pergoveris®.[1]
  • Database Identifiers:
  • DrugBank ID: DB00044 [3]
  • CAS Number: 152923-57-4 [1]
  • Regulatory Classification:
  • Anatomical Therapeutic Chemical (ATC) Code: G03GA07, which places it in the class of Gonadotropins.[17]
  • Chemical and Molecular Properties:
  • Chemical Formula: C1014​H1609​N287​O294​S27​ [3]
  • Average Molecular Weight: Approximately 30,000 Da [3]

2.2. Structural Elucidation

The biological function and specificity of Lutropin alfa are direct consequences of its complex glycoprotein structure, which is engineered to mimic that of endogenous human LH.

  • Heterodimeric Glycoprotein Structure: Lutropin alfa is a heterodimeric glycoprotein, meaning it is composed of two distinct, non-covalently linked polypeptide subunits, designated as the alpha (α) and beta (β) subunits.[2]
  • Alpha (α) Subunit: This subunit is composed of a 92-amino acid sequence. A key structural feature of the glycoprotein hormone family is that this α-subunit is common to several hormones, including FSH, human chorionic gonadotropin (hCG), and thyroid-stimulating hormone (TSH). Its primary role is in receptor activation after the β-subunit has conferred binding specificity.[4]
  • Beta (β) Subunit: This subunit consists of a 121-amino acid sequence. Unlike the α-subunit, the β-subunit is unique to LH and is responsible for the hormone's biological specificity. It dictates the molecule's ability to bind with high affinity to the specific Lutropin-choriogonadotropic hormone receptor (LHCGR), thereby initiating its physiological effects.[4] The distinct nature of the β-subunit is what differentiates the action of LH from that of FSH or TSH, despite their sharing a common α-subunit. This structural distinction is fundamental to its clinical application, as it mandates that to replicate the full physiological process of follicular development, the distinct signal provided by the LH β-subunit must be complemented by the signal from the FSH β-subunit, necessitating combination therapy.
  • Glycosylation: The protein backbone of Lutropin alfa is modified by the attachment of carbohydrate chains, a process known as glycosylation, which is critical for the hormone's folding, stability, and biological activity. The carbohydrate attachment occurs via N-linkage to asparagine residues. Specific N-glycosylation sites have been identified at positions Asn-52 and Asn-78 on the α–subunit and at Asn-30 on the β–subunit.[2] The resulting glycosylation pattern is very similar to that of LH naturally derived from the human pituitary gland. One subtle difference arising from its recombinant production in CHO cells is that the glycan structures are sialylated, whereas natural LH derived from urinary or pituitary sources also contains N-acetylgalactosamine and sulfated residues. However, this difference in glycan composition is considered to have little to no relevance to the molecule's overall biological activity.[5]

2.3. Recombinant Production and Pharmaceutical Formulation

The development of Lutropin alfa as a biopharmaceutical is predicated on its production via recombinant DNA technology, a process that confers significant advantages in purity, consistency, and safety over older, biologically-sourced preparations.

  • Manufacturing Process: Lutropin alfa is produced in a genetically modified Chinese Hamster Ovary (CHO) cell line. The process involves inserting the human genes that encode for the LH α- and β-chains into the CHO cells.[2] These engineered cells are then cultured in large-scale bioreactors. During this process, the cells secrete the fully assembled, heterodimeric Lutropin alfa glycoprotein directly into the cell culture medium.[2]
  • Purification and Purity: Following secretion, the culture medium is collected, and the Lutropin alfa protein is isolated and purified through a sophisticated series of chromatographic steps.[2] This highly controlled process yields a product with exceptional purity, typically greater than 99%, and ensures the removal of host cell proteins, DNA, and other process-related impurities.[8] This stands in stark contrast to urinary-derived gonadotropins (e.g., hMG), which are inherently less pure and can exhibit significant batch-to-batch variability in their hormonal composition and isoform profile.[4] The recombinant origin of Lutropin alfa is therefore a core component of its value proposition, offering a highly consistent and predictable therapeutic agent.
  • Pharmaceutical Form: Lutropin alfa is most commonly supplied as a sterile, white, lyophilized (freeze-dried) powder in vials, intended for reconstitution with an accompanying solvent (sterile water for injection) prior to subcutaneous administration.[6] In some markets, to enhance patient convenience, it is also available as a ready-to-use solution in a pre-filled injection pen.[12]
  • Excipients: The lyophilized powder formulation contains several excipients to ensure its stability, solubility, and physiological compatibility. These include sucrose (a cryoprotectant), dibasic sodium phosphate dihydrate and monobasic sodium phosphate monohydrate (to form a buffer system), polysorbate 20 (a surfactant to prevent protein aggregation), and L-methionine (an antioxidant). Phosphoric acid and/or sodium hydroxide are used to adjust the pH of the reconstituted solution to a range of 7.5 to 8.5.[2]

Section 3: Comprehensive Pharmacological Profile

3.1. Mechanism of Action (MOA)

The pharmacological effects of Lutropin alfa are mediated by its specific interaction with the Lutropin-choriogonadotropic hormone receptor (LHCGR), initiating a cascade of intracellular events that culminate in ovarian steroidogenesis.

  • Receptor Binding and Activation: Lutropin alfa functions as a potent agonist at the LHCGR.[3] The LHCGR is a member of the G protein-coupled receptor (GPCR) superfamily and is expressed on the cell membranes of ovarian theca cells, mature granulosa cells, and, in males, testicular Leydig cells.[3] The binding of Lutropin alfa to this receptor induces a conformational change that triggers downstream signaling.
  • Intracellular Signaling Pathways: Upon activation, the LHCGR couples with intracellular G proteins to initiate signaling cascades. The primary pathway involves the Gs protein, which activates the enzyme adenylate cyclase. This leads to an increase in intracellular cyclic adenosine monophosphate (cAMP), which in turn activates protein kinase A (PKA). The PKA pathway is the principal driver of steroidogenesis, upregulating the expression and activity of enzymes involved in hormone synthesis.[3] A secondary pathway involving the Gq/11 protein and phospholipase C activation is also implicated, particularly in the processes leading to ovulation.[8]
  • The "Two-Cell, Two-Gonadotropin" Model: The physiological role of Lutropin alfa in female fertility is best understood through this essential model of ovarian function.[2] The process requires the coordinated action of both LH and FSH on two different ovarian cell types:
  1. LH Action on Theca Cells: In the early to mid-follicular phase, LH (provided by Lutropin alfa) binds to LHCGRs on the theca cells surrounding the developing follicle. This stimulates the theca cells to take up cholesterol and convert it into androgens, primarily androstenedione and testosterone.[2]
  2. FSH Action on Granulosa Cells: These androgens then diffuse across the basement membrane into the adjacent granulosa cells. Within the granulosa cells, the enzyme aromatase—whose expression and activity are potently stimulated by FSH (e.g., follitropin alfa)—converts the androgens into estrogens, principally estradiol.[2]

This interdependent process makes it clear that LH action is a pharmacological prerequisite for FSH-mediated estradiol production. Administering Lutropin alfa alone would be physiologically insufficient for follicular maturation, as the androgens produced would not be efficiently converted to estradiol. This pharmacological reality mandates its clinical use exclusively in combination with an FSH preparation.

3.2. Pharmacodynamics (PD)

The pharmacodynamic effects of Lutropin alfa are the measurable physiological responses resulting from its mechanism of action, primarily centered on ovarian steroid production and follicular development.

  • Primary Effect: The principal and most direct pharmacodynamic effect of Lutropin alfa administration is a dose-dependent increase in the secretion of estradiol by the developing ovarian follicles.[2] This rise in serum estradiol is a key biomarker used to monitor patient response during treatment.
  • Role in Follicular Development: While FSH is the primary hormone responsible for the recruitment of a cohort of antral follicles and their initial growth, LH plays a critical and multifaceted supportive role. It is essential for providing the androgen substrate necessary for estrogen synthesis. Furthermore, as follicles mature, granulosa cells begin to express LHCGRs, making them responsive to LH. In the later stages of follicular development, LH action is believed to be important for final oocyte maturation and for improving oocyte quality.[4]
  • The LH "Therapeutic Window" and "Ceiling": The physiological response to LH is not linear. There appears to be a "therapeutic window" where optimal LH levels support the growth and health of the dominant follicle. Below this window, follicular development may be suboptimal due to insufficient androgen substrate. Above this window, a concept known as the "LH ceiling" suggests that excessively high intra-follicular androgen levels can become toxic and promote follicular atresia (degeneration).[4] This concept provides a strong rationale for the careful and precise dosing of Lutropin alfa to maintain LH activity within the optimal physiological range, supporting the dominant follicle while potentially contributing to the atresia of smaller, non-dominant follicles.

3.3. Pharmacokinetics (PK)

The pharmacokinetic profile of Lutropin alfa describes its absorption, distribution, metabolism, and excretion, which collectively determine the drug's concentration over time and underpin its recommended dosing regimen.

  • Absorption and Distribution: Lutropin alfa is administered via subcutaneous (SC) injection.[5] Following SC administration, it is absorbed into the systemic circulation, reaching peak serum concentrations ( Cmax​) in approximately 4 to 16 hours.[2] The mean absolute bioavailability is estimated to be 56%.[2] The steady-state volume of distribution ( Vss​) is approximately 10 to 14 L, suggesting that the drug is primarily distributed within the plasma and extracellular fluid compartments.[2]
  • Metabolism and Elimination: The drug exhibits a biphasic elimination pattern. After subcutaneous administration, Lutropin alfa is eliminated from the body with a mean terminal half-life (t1/2​) of approximately 18 hours.[2] This pharmacokinetic property is highly suitable for a once-daily dosing schedule, as it allows for the maintenance of therapeutic steady-state concentrations over a 24-hour interval without causing excessive accumulation. Upon repeated daily administration, a modest accumulation is observed, with an accumulation ratio of approximately 1.6.[2] The total body clearance is approximately 2 to 3 L/h.[2]
  • Excretion: Lutropin alfa is cleared primarily through metabolism. Less than 5% of an administered dose is excreted unchanged in the urine, indicating that renal excretion is a minor pathway of elimination.[2]
  • Drug-Drug Interactions: No clinically significant pharmacokinetic interactions have been observed when Lutropin alfa is co-administered with follitropin alfa.[2] Formal interaction studies with other drugs have not been conducted.
Table 1: Key Pharmacokinetic Parameters of Lutropin alfa
ParameterValue
Route of AdministrationSubcutaneous
Bioavailability~56% 3
Time to Peak Concentration (Tmax​)4–16 hours 2
Terminal Half-Life (t1/2​)~18 hours 2
Volume of Distribution (Vss​)10–14 L 2
Total Body Clearance2–3 L/h 2
Renal Excretion (unchanged)<5% 2

Section 4: Clinical Evidence and Therapeutic Application

4.1. Approved Indications and Patient Population

The clinical use of Lutropin alfa is highly specific, targeting a well-defined patient population with a rare endocrine disorder.

  • Primary Indication: Lutropin alfa, administered in combination with an FSH preparation, is indicated for the stimulation of follicular development in adult women diagnosed with severe Luteinising Hormone (LH) and Follicle-Stimulating Hormone (FSH) deficiency.[6] This condition is also known as hypogonadotropic hypogonadism.
  • Defining the Patient Population: The key diagnostic criterion that defines this patient population is a profoundly low level of endogenous LH. Across clinical trials and regulatory approvals, this is quantitatively specified as an endogenous serum LH level of less than 1.2 IU/L.[3] This precise biochemical marker identifies a small and specific subset of infertile women for whom FSH monotherapy is insufficient to achieve follicular growth. The rarity of this condition led to Lutropin alfa being granted orphan drug status in some jurisdictions, a designation that would profoundly shape its clinical development and regulatory history.[23]

4.2. Dosage, Administration, and Monitoring

The successful and safe use of Lutropin alfa requires a carefully managed and individualized treatment protocol under the guidance of a fertility specialist.

  • Initiation and Supervision: Treatment must be initiated and supervised by a physician with expertise in the management of fertility disorders.[6] While the first injection should be performed under medical supervision, patients who are well-motivated and adequately trained may self-administer subsequent doses.[6]
  • Recommended Dosage Regimen: The recommended starting regimen consists of 75 IU of Lutropin alfa administered subcutaneously once daily. This is given concurrently with a daily subcutaneous injection of follitropin alfa, typically at a starting dose of 75-150 IU.[6]
  • Dose Titration and Duration: Treatment must be tailored to the individual patient's ovarian response. The dose of follitropin alfa can be adjusted, usually at 7- to 14-day intervals and in increments of 37.5 IU to 75 IU, to achieve an optimal response. The total duration of stimulation in a single cycle may be extended for up to five weeks if necessary.[6]
  • Therapeutic Monitoring: Close monitoring of the patient's response is essential for both efficacy and safety. This is achieved through two primary methods:
  1. Transvaginal Ultrasound: To measure the number and size of developing follicles.[6]
  2. Serum Estradiol Measurements: To assess the functional activity of the follicles.[6]

This dual-monitoring approach allows the clinician to track follicular development, time the induction of ovulation correctly, and identify patients at risk of developing Ovarian Hyperstimulation Syndrome (OHSS).6

  • Induction of Final Maturation and Ovulation: When monitoring indicates that an optimal response has been achieved (typically defined by the presence of at least one follicle with a mean diameter ≥17 mm and an appropriate estradiol level), a single injection of human chorionic gonadotropin (hCG) at a dose of 5,000 to 10,000 IU, or recombinant hCG (r-hCG) at a dose of 250 mcg, is administered. This injection is given 24 to 48 hours after the final doses of Lutropin alfa and follitropin alfa to trigger the final stages of oocyte maturation and induce ovulation.[6]

4.3. Analysis of Clinical Trials

The clinical trial program for Lutropin alfa reflects the challenges of studying a drug for a rare disease and highlights a significant divergence in regulatory interpretation of clinical endpoints.

  • Pivotal Trials for European Approval: The marketing authorization in Europe was primarily supported by clinical trials conducted in the target population of women with severe LH and FSH deficiency. A key study involved a small cohort of 38 women. The main measure of effectiveness was the achievement of functional follicular development. In this study, 67% of the women (6 out of 9) who received the recommended 75 IU dose of Luveris® in combination with FSH successfully produced functional follicles.[12] This was considered sufficient evidence of efficacy for this specific, unmet medical need.
  • US Regulatory Trials and the Endpoint Controversy: The path to approval in the US was more contentious. The data submitted to the FDA from several studies (including 21008, 6253, 6905, 7798, and 8297) were scrutinized by the agency and its Reproductive Health Advisory Committee.[10]
  • The committee unanimously voted (15 to 0) that the data failed to demonstrate efficacy for the primary clinical endpoint of ovulation rate.
  • However, the committee was narrowly divided but ultimately voted in favor (11 to 3) that the data did demonstrate efficacy for the surrogate endpoint of follicular development.[10]

This debate over the appropriate endpoint was a critical juncture. While follicular development is a necessary precursor to pregnancy, it is not a guarantee of it. The FDA ultimately granted an accelerated approval based on this surrogate endpoint, but with a stringent requirement for a Phase 4 postmarketing study to confirm a tangible clinical benefit, defined as the pregnancy rate.10 The inability to conduct this confirmatory trial due to recruitment difficulties in this rare population was the direct cause of the drug's eventual withdrawal from the US market. This situation serves as a prominent example of the "surrogate endpoint trap," where an approval based on an intermediate marker is contingent on later validation of a true clinical outcome, a requirement that can be logistically prohibitive for orphan drugs.

  • Investigational Trials in Broader Populations: Beyond its primary indication, Lutropin alfa has been investigated in several other clinical contexts to explore the potential benefits of LH supplementation.
  • Poor Ovarian Responders and Advanced Reproductive Age: Several trials (e.g., NCT01075815, NCT01079949) have been conducted in women who respond poorly to standard ovarian stimulation or are of advanced reproductive age. The hypothesis in these studies was that LH supplementation might improve oocyte and embryo quality, thereby increasing implantation and pregnancy rates.[25]
  • Standard IVF/ICSI Protocols: Other studies, such as NCT00889512 and NCT00553293, explored various dosing regimens of r-hLH within standard IVF protocols for general infertility populations to determine if LH supplementation could optimize outcomes.[27]
  • Other Conditions: An observational study (NCT01457703) included Lutropin alfa as a drug of interest in studying hormonal alterations in obesity, indicating research interest in its role in other metabolic and reproductive contexts.[29]
Table 2: Summary of Key Lutropin alfa Clinical Trials
Trial IDPhaseCondition(s)Patient PopulationIntervention ArmsKey EndpointsStatus/Outcome
EMA Pivotal Studies (e.g., GF6253)IIISevere LH/FSH DeficiencyWomen with LH <1.2 IU/LLuveris® + FSH vs. FSH aloneFollicular DevelopmentCompleted; Supported EU approval 12
NCT00328926IVSevere LH/FSH DeficiencyWomen with LH <1.2 IU/LLuveris® (75 IU / 25 IU) + FSH vs. Placebo + FSHClinical PregnancyTerminated; Failure to complete led to US withdrawal 30
NCT01075815IIInfertility / Ovulation InductionWomen of advanced reproductive age (>35 years) undergoing IVF/ICSIr-FSH + r-LH vs. r-FSH aloneEmbryo Competence, Implantation RateCompleted 25
ESPART (NCT02047227)IIIInfertilityPoor ovarian responders undergoing ARTPergoveris® (r-FSH/r-LH) vs. GONAL-f® (r-FSH)Number of Oocytes RetrievedCompleted 31
NCT00889512N/AInfertility, Hypothalamic AmenorrheaWomen undergoing IVF without endogenous LH activityFixed dose r-LH vs. Increasing dose r-LH (both with FSH)Follicular Response, Oocyte QualityCompleted 27

Section 5: Safety, Tolerability, and Risk Management

5.1. Adverse Event Profile

The safety profile of Lutropin alfa is well-characterized and consistent with that of other gonadotropin therapies used for ovarian stimulation.

  • Common Adverse Effects (occurring in 1% to 10% of patients):
  • General and Administration Site Conditions: The most frequently reported adverse events are local reactions at the site of injection, including pain, erythema (redness), haematoma (bruising), swelling, and/or irritation.[12]
  • Nervous System Disorders: Headache is a common systemic side effect.[12]
  • Gastrointestinal Disorders: Nausea, vomiting, diarrhea, and abdominal pain or discomfort are frequently reported.[12]
  • Reproductive System and Breast Disorders: Mild to moderate Ovarian Hyperstimulation Syndrome (OHSS) and its associated symptoms, ovarian cysts, pelvic pain, and breast pain are common.[12]
  • Very Rare Adverse Effects (occurring in <0.01% of patients):
  • Vascular Disorders: Thromboembolic events (blood clots) have been reported very rarely, and these are typically associated with cases of severe OHSS.[6]
  • Immune System Disorders: Mild to severe hypersensitivity reactions, including rare cases of anaphylactic reactions and shock, have been observed.[6]

5.2. Ovarian Hyperstimulation Syndrome (OHSS): A Major Risk of Therapy

OHSS is the most significant and potentially serious complication of controlled ovarian stimulation with gonadotropins. It is crucial to recognize that OHSS is a class effect of this therapeutic approach, stemming from the supraphysiological ovarian response, rather than a specific toxicity of the Lutropin alfa molecule itself.[34] Adherence to recommended dosing and meticulous monitoring are the cornerstones of risk mitigation.[6]

  • Definition and Pathophysiology: OHSS is an exaggerated systemic response to ovarian stimulation, characterized by a spectrum of clinical signs and symptoms. The underlying mechanism involves increased vascular permeability, driven by vasoactive substances (such as vascular endothelial growth factor, VEGF) secreted by the hyperstimulated ovaries, leading to a fluid shift from the intravascular to the third space.[34]
  • Clinical Presentation:
  • Mild to Moderate OHSS: Symptoms include abdominal bloating, discomfort, or pain, nausea, vomiting, diarrhea, and mild to moderate ovarian enlargement seen on ultrasound.[12]
  • Severe OHSS: This is a serious and potentially life-threatening medical condition that can develop rapidly. It is characterized by severe abdominal pain, rapid weight gain, marked ovarian enlargement, and significant third-space fluid accumulation, leading to ascites (fluid in the abdomen) and pleural effusions (fluid around the lungs). This fluid shift can cause hypovolemia (low blood volume), haemoconcentration, electrolyte imbalances, dyspnea (shortness of breath), and oliguria (reduced urine output). Rare but severe complications include thromboembolic events and ovarian torsion (twisting of the ovary).[19]
  • Risk Mitigation Strategies:
  • Careful Patient Monitoring: Regular monitoring of follicular growth with transvaginal ultrasound and measurement of serum estradiol levels is the most critical step in identifying patients at risk.[6]
  • Withholding the hCG Trigger: If monitoring reveals an excessive ovarian response (e.g., very high estradiol levels or an excessive number of mature follicles), the final ovulatory trigger injection of hCG must be withheld. This is the single most effective measure to prevent the development of severe OHSS.[19]
  • Patient Counseling: Patients should be advised to refrain from intercourse if OHSS is suspected, as pregnancy can exacerbate the condition.[19]

5.3. Contraindications and Precautions

The use of Lutropin alfa is contraindicated in several situations where its administration would be unsafe or ineffective.

  • Absolute Contraindications:
  • Known hypersensitivity to gonadotropins (LH, FSH) or any of the excipients.[6]
  • Presence of tumors of the hypothalamus, pituitary gland, ovary, uterus, or breast, as these can be hormone-sensitive.[6]
  • Primary ovarian failure, a condition where the ovaries are unresponsive to gonadotropin stimulation.[19]
  • Ovarian enlargement or cysts not related to polycystic ovarian disease and of unknown origin.[19]
  • Gynaecological haemorrhages or abnormal vaginal bleeding of unknown origin.[36]
  • Pregnancy.[34]
  • Anatomical conditions that would make a normal pregnancy impossible, such as malformations of the sexual organs or significant fibroid tumors of the uterus.[19]
  • Warnings and Precautions:
  • Multiple Pregnancies: Gonadotropin therapy significantly increases the risk of multiple gestation (twins, triplets, etc.). As multiple pregnancy is associated with higher risks for both mother and babies, this potential outcome must be thoroughly discussed with the couple before initiating treatment.[34]
  • Ectopic Pregnancy: Women undergoing fertility treatments, particularly those with a history of tubal disease, have an increased risk of ectopic pregnancy.[7]
  • Pregnancy Loss: The rate of spontaneous abortion (miscarriage) is higher in patients undergoing ovarian stimulation for infertility than in the general population conceiving naturally, though it is comparable to rates in other infertile populations.[19]
  • Reproductive System Neoplasms: There have been isolated reports of both benign and malignant tumors of the ovary and other reproductive organs in women who have undergone multiple cycles of infertility treatment. A definitive causal link between gonadotropin therapy and an increased risk of these tumors has not been established.[7]

5.4. Drug Interactions

The potential for drug-drug interactions with Lutropin alfa is limited.

  • No formal drug-drug interaction studies have been conducted, with the exception of its intended use with follitropin alfa.[2]
  • Lutropin alfa should not be mixed in the same injection syringe with any other medicinal products, except for follitropin alfa (Gonal-f®). Studies have specifically shown that co-administration and mixing of these two products do not significantly alter the activity, stability, or pharmacokinetic properties of either active substance.[19]

Section 6: Regulatory History and Market Status

The regulatory journey of Lutropin alfa is a compelling case study in the complexities of orphan drug development, highlighting how differing regulatory philosophies and commercial realities can lead to divergent market outcomes for the same therapeutic agent.

6.1. European Medicines Agency (EMA) Trajectory

  • Initial Approval and Status: Luveris® was first granted a marketing authorization valid throughout the European Union on November 29, 2000.[19] The approval was for its specific indication of stimulating follicular development in women with severe LH and FSH deficiency (defined as serum LH <1.2 IU/L), based on clinical trials that demonstrated its efficacy in achieving the surrogate endpoint of functional follicle production.[12] The medicine remains authorized for use in the EU, with the marketing authorization held by Merck Europe B.V..[12]
  • Approval of Pergoveris®: Recognizing the clinical necessity of co-administering LH and FSH, a fixed-dose combination product, Pergoveris®, was developed. It received marketing authorization from the European Commission on June 25, 2007.[14] The approval was supported by the existing clinical data for Luveris® and Gonal-f®, along with bioequivalence studies confirming that the combined injection delivered the active substances comparably to separate injections.[14] Further enhancing its market position, a ready-to-use, pre-filled pen formulation of Pergoveris® was approved in May 2017, significantly improving patient convenience and ease of use.[15]

6.2. U.S. Food and Drug Administration (FDA) Trajectory

  • Accelerated Approval: In the United States, Luveris® was approved by the FDA on October 8, 2004.[23] This approval was granted under the agency's accelerated approval regulations (21 CFR part 314, subpart H), which allow for the marketing of drugs for serious conditions based on a surrogate endpoint that is reasonably likely to predict clinical benefit.[10]
  • Basis for Approval and Controversy: The basis for this accelerated approval was contentious. The submitted clinical trial data had failed to show a statistically significant benefit on the definitive clinical endpoints of ovulation or pregnancy rate. The FDA's Advisory Committee was divided, ultimately recommending approval based on the surrogate endpoint of follicular development, despite acknowledging the weakness of the evidence for more definitive outcomes.[10]
  • Postmarketing Commitment and Withdrawal: A mandatory condition of the accelerated approval was that the sponsor, EMD Serono, conduct a Phase 4 postmarketing study to verify and describe the clinical benefit of Luveris® on the pregnancy rate.[10] However, the company encountered significant difficulties in recruiting a sufficient number of patients from the extremely small and specific population of women with hypogonadotropic hypogonadism. Citing the infeasibility of completing this required trial, EMD Serono voluntarily requested that the FDA withdraw the approval for the New Drug Application (NDA) for Luveris®. The FDA officially withdrew its approval on April 12, 2016.[11] The company explicitly stated that this decision was a result of logistical and recruitment challenges and was not related to any new safety or efficacy concerns.[11] This sequence of events illustrates the profound impact that differing evidentiary standards and the practical challenges of orphan drug research can have on market access.

6.3. Manufacturers and Formulations

  • Primary Manufacturer: Lutropin alfa is developed and manufactured by Merck KGaA, Darmstadt, Germany, and its various global biopharmaceutical affiliates, including EMD Serono (in the US and Canada) and Merck Europe B.V..[3]
  • Available Formulations:
  • Luveris®: Available as a lyophilized powder in vials, typically containing 75 IU of Lutropin alfa, which is reconstituted with 1 mL of solvent for subcutaneous injection.[6] A solution in a pre-filled pen is also available in some markets.[12]
  • Pergoveris®: This fixed-dose combination product contains both follitropin alfa and Lutropin alfa. It is available in multiple formats, including powder and solvent for reconstitution and, more commonly, as a liquid solution in a multi-dose, pre-filled pen. The pen is available in various strengths (e.g., 300 IU FSH/150 IU LH; 450 IU FSH/225 IU LH; 900 IU FSH/450 IU LH) to allow for individualized dosing.[14] The introduction of Pergoveris®, particularly in the convenient pen formulation, represents a significant strategic evolution, simplifying treatment and potentially improving patient adherence.

Section 7: Comparative Analysis and Strategic Positioning

7.1. Lutropin alfa (Recombinant LH) vs. Human Menopausal Gonadotropin (hMG)

The choice between using a recombinant LH preparation like Lutropin alfa and a urinary-derived product like hMG is a central and ongoing debate in reproductive medicine. The decision involves a trade-off between purity, precision, composition, clinical evidence, and cost.

  • Source, Purity, and Composition: This is the most fundamental difference.
  • Lutropin alfa: It is a pure, single-molecule product (r-hLH) manufactured via recombinant technology. This ensures high purity (>99%), a consistent isoform profile, and negligible batch-to-batch variability, allowing for precise dosing.[4]
  • hMG (e.g., Menopur®): It is extracted and purified from the urine of postmenopausal women. It is a mixture of hormones, containing both FSH and LH activity. Critically, the LH activity in hMG is primarily derived from human chorionic gonadotropin (hCG), a molecule that binds to the same receptor as LH but has a much longer half-life and activates different intracellular signaling pathways.[44] Urinary preparations are inherently less pure and are subject to greater batch-to-batch variability.[21] This distinction frames a key clinical question: is a pure, precise LH signal preferable, or does the complex "soup" of hormones in hMG, particularly the potent and long-acting hCG component, offer a different, potentially beneficial biological stimulus?
  • Comparative Clinical Efficacy: The clinical evidence comparing these two approaches is extensive but lacks a definitive consensus, with outcomes often depending on the specific patient population, stimulation protocol, and endpoints measured.
  • Oocyte Yield: Some meta-analyses and trials suggest that protocols using r-hFSH with or without r-hLH may yield a higher number of retrieved oocytes compared to protocols using hMG.[44]
  • Pregnancy and Live Birth Rates: This is the most contested area. Several large meta-analyses have found that hMG is associated with a significantly higher live birth rate compared to r-FSH monotherapy.[47] Conversely, other analyses suggest that adding r-LH to r-FSH results in a higher pregnancy rate than other combinations.[44] A recent large retrospective study in an Asian population found that a combination of r-hFSH+r-hLH led to a significantly higher cumulative live birth rate and a lower miscarriage rate compared to a combination of r-hFSH+hMG.[48]
  • Specific Populations: There is a body of evidence suggesting that urinary gonadotropins, including hMG, may provide better outcomes in certain difficult-to-treat populations, such as poor ovarian responders and women of advanced maternal age.[47]
  • Safety and Cost-Effectiveness:
  • Safety: The risk of OHSS is generally considered comparable between the different gonadotropin types, although some individual trials have suggested a potentially higher risk with recombinant combinations.[47]
  • Cost: Urinary-derived preparations like hMG are consistently found to be more cost-effective than their recombinant counterparts. This cost difference is a significant factor in clinical decision-making, especially in healthcare systems with budget constraints.[47]
Table 3: Comparative Profile of Lutropin alfa (r-hLH) vs. Human Menopausal Gonadotropin (hMG)
FeatureLutropin alfa (r-hLH)Human Menopausal Gonadotropin (hMG)
SourceRecombinant DNA technology (CHO cells)Purified from urine of postmenopausal women
Purity & ConsistencyHigh (>99%); consistent isoform profile; low batch-to-batch variabilityLower purity; contains other urinary proteins; subject to batch-to-batch variability
LH Activity SourceLutropin alfa (recombinant human LH)Primarily human chorionic gonadotropin (hCG)
Pharmacology of LH-like MoleculeShorter half-life; distinct intracellular signalingLonger half-life; different intracellular signaling
Clinical Evidence (Pregnancy/Live Birth)Conflicting and context-dependent. Some studies show superiority, others do not.Conflicting and context-dependent. Some meta-analyses show higher live birth rates.
Cost-EffectivenessGenerally lower (higher cost)Generally higher (lower cost)
Key AdvantagePurity, precision, and consistency of dosing.Lower acquisition cost; potential benefit of long-acting hCG activity in certain patient groups.

7.2. Pergoveris®: The Fixed-Dose Combination

The development of Pergoveris® represents a strategic evolution of Lutropin alfa, aiming to simplify therapy and expand its clinical application.

  • Rationale and Positioning: The primary rationale for Pergoveris® is patient convenience. Since Lutropin alfa must be co-administered with FSH, combining them into a single, fixed-ratio product eliminates the need for mixing or separate injections.[14] This positions Pergoveris® as a premium, user-friendly option, particularly with the availability of the pre-filled pen, which is part of Merck's "Family of Pens™" designed for ease of use.[15]
  • Clinical Evidence (The ESPART Trial): The largest and most significant trial for Pergoveris® is the Phase III ESPART study (NCT02047227). This trial signaled a crucial strategic shift by evaluating the product not in its original orphan indication, but in the much broader and more common clinical challenge of "poor ovarian responders".[31]
  • The primary endpoint, the total number of oocytes retrieved, was found to be comparable between the Pergoveris® group and the r-FSH monotherapy group.[52]
  • However, post-hoc analyses revealed important nuances. Treatment with Pergoveris® was associated with a lower rate of total pregnancy outcome failure. Furthermore, a significant interaction was observed: patients with moderate or severe poor ovarian response had higher live birth rates with Pergoveris®, whereas those with a milder poor response profile fared better with r-FSH alone.[52]

This evidence suggests a move away from a strictly indication-based use (severe LH deficiency) toward a more personalized, patient-profile-based application, where Pergoveris® may be beneficial for specific subgroups of poor responders.

Section 8: Synthesis and Concluding Remarks

8.1. Final Synthesis

Lutropin alfa stands as a landmark achievement in reproductive biotechnology, offering a pure, precise, and consistent form of human luteinizing hormone. Its development provided an essential tool for treating a specific and challenging form of female infertility: hypogonadotropic hypogonadism. The story of Lutropin alfa is one of scientific precision confronting the complexities of clinical evidence generation and navigating divergent global regulatory landscapes. Its undisputed physiological necessity and efficacy in its narrow orphan indication are well-established. However, its journey, particularly the withdrawal from the US market, underscores the profound difficulties inherent in orphan drug development, where the rarity of the condition can make fulfilling the evidentiary demands of regulatory bodies a logistical and commercial impossibility. The subsequent evolution to the fixed-dose combination product, Pergoveris®, reflects a strategic adaptation, enhancing patient convenience and exploring broader clinical applications in more common infertility scenarios, such as poor ovarian response.

8.2. Expert Commentary on Clinical Role

From a clinical perspective, the role of Lutropin alfa in the modern fertility armamentarium is nuanced.

  • For the Indicated Population: In women with confirmed severe LH deficiency (serum LH <1.2 IU/L), the addition of exogenous LH activity is not optional; it is a physiological necessity. For this small but well-defined cohort, treatment with Lutropin alfa in combination with follitropin alfa (either as separate injections or as Pergoveris®) represents the unequivocal, evidence-based standard of care.
  • Outside the Primary Indication: The routine use of LH supplementation in general IVF populations or unselected poor responders remains a subject of intense debate. The clinical evidence does not support a universal benefit. The choice between adding recombinant LH (Lutropin alfa) or using a urinary-derived hMG preparation is not settled and must be individualized. The decision should be based on a careful consideration of patient-specific factors, including age, ovarian reserve biomarkers, previous response to stimulation, the specific stimulation protocol being used, and cost-effectiveness. The clinician must weigh the theoretical advantages of the purity and precision of r-hLH against the lower cost and different biological stimulus provided by the hCG component of hMG.
  • The Rise of Combination Products: The development of Pergoveris® is the most clinically relevant evolution of this therapy. By simplifying the treatment regimen, it addresses a key practical barrier for patients and may improve treatment adherence. Its demonstrated benefit in certain subgroups of poor responders suggests its role may continue to expand beyond its initial, narrow indication.

8.3. Future Directions and Recommendations

  • Recommendations for Clinicians:
  1. Accurate Diagnosis is Paramount: The primary clinical imperative is the accurate identification of patients with true hypogonadotropic hypogonadism, as they are the group that will derive the most definitive benefit from LH supplementation.
  2. Individualize Treatment: For patients outside this indication, the decision to supplement with LH activity should be made on a case-by-case basis, considering the totality of the conflicting evidence and patient-specific factors.
  3. Prioritize Safety: Regardless of the gonadotropin preparation chosen, meticulous monitoring of ovarian response through ultrasound and estradiol measurements remains the cornerstone of safe practice to minimize the risk of OHSS.
  • Recommendations for Future Research:
  1. Subgroup-Specific Trials: Future research should move beyond broad comparisons and focus on well-designed, adequately powered, head-to-head clinical trials in specific, well-phenotyped subgroups of patients (e.g., poor responders stratified by age or biomarkers) to definitively clarify who benefits most from r-hLH versus hMG.
  2. Mechanistic Studies: Further investigation into the distinct intracellular signaling pathways activated by LH versus hCG in human granulosa cells could provide a stronger biological basis for selecting one agent over the other in different clinical scenarios.
  3. Cost-Effectiveness Analyses: As fertility treatments represent a significant financial burden, rigorous cost-effectiveness analyses comparing different gonadotropin strategies in various healthcare systems are urgently needed to guide both clinical practice and health policy.

Works cited

  1. Lutropinalfa CAS#: 152923-57-4 - ChemicalBook, accessed August 6, 2025, https://amp.chemicalbook.com/ProductChemicalPropertiesCB61174010_EN.htm
  2. Luveris® (lutropin alfa for injection) - accessdata.fda.gov, accessed August 6, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2004/21322lbl.pdf
  3. Lutropin alfa: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed August 6, 2025, https://go.drugbank.com/drugs/DB00044
  4. Lutropin alpha, recombinant human luteinizing hormone, for the ..., accessed August 6, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2726078/
  5. Pharmacology Review(s) - accessdata.fda.gov, accessed August 6, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2004/021322s000_Luveris_pharmr.pdf
  6. AUSTRALIAN PRODUCT INFORMATION – LUVERIS® (lutropin alfa (rch)) 1 NAME OF THE MEDICINE - Medsinfo, accessed August 6, 2025, https://medsinfo.com.au/api/documents/Luveris_PI?format=pdf
  7. Luveris, INN-lutropin alfa, accessed August 6, 2025, https://ec.europa.eu/health/documents/community-register/2015/20150424131830/anx_131830_en.pdf
  8. Lutropin alfa, Recombinant human iuteinizing hormone - Therapeutic Proteins, accessed August 6, 2025, https://therapeutic.creativebiomart.net/lutropin-alfa-recombinant-human-iuteinizing-hormone.html
  9. Recombinant follitropin alfa/lutropin alfa in fertility treatment - PMC - PubMed Central, accessed August 6, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2819896/
  10. Untitled - accessdata.fda.gov, accessed August 6, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2004/021322s000_Luveris_medr.pdf
  11. FDA Withdraws NDA for Fertility Drug After EMD Serono Fails to Run Postmarket Trial, accessed August 6, 2025, https://www.raps.org/news-and-articles/news-articles/2016/4/fda-withdraws-nda-for-fertility-drug-after-emd-ser
  12. Luveris | European Medicines Agency (EMA), accessed August 6, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/luveris
  13. EMD Serono; Withdrawal of Approval of a New Drug Application for LUVERIS, accessed August 6, 2025, https://www.federalregister.gov/documents/2016/04/12/2016-08336/emd-serono-withdrawal-of-approval-of-a-new-drug-application-for-luveris
  14. Pergoveris | European Medicines Agency (EMA), accessed August 6, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/pergoveris
  15. 2017-05-10 Approval for Merck's Pergoveris® Pen for Fertility Treatment, accessed August 6, 2025, https://www.merckgroup.com/en/news/pergoveris-pen-ec-aproval-10-05-2017.html
  16. Lutropin alfa - PubChem, accessed August 6, 2025, https://pubchem.ncbi.nlm.nih.gov/substance/46507624
  17. lutropin alfa - Drug Central, accessed August 6, 2025, https://drugcentral.org/drugcard/4929
  18. LH | Ligand page | IUPHAR/BPS Guide to PHARMACOLOGY, accessed August 6, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=1159
  19. Luveris, INN-lutropin alfa - EMA, accessed August 6, 2025, https://www.ema.europa.eu/en/documents/product-information/luveris-epar-product-information_en.pdf
  20. Luveris®: Recombinant hLH for tailored therapy - Merck Healthcare, accessed August 6, 2025, https://hcp.merckgroup.com/en/fertility/portfolio/therapeutics/luveris.html
  21. Comparison between cycles of the same patients when using ..., accessed August 6, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6524176/
  22. Lutropin alfa - Australian Prescriber, accessed August 6, 2025, https://australianprescriber.tg.org.au/articles/lutropin-alfa.html
  23. FDA Approvals: ExAblate 2000, Luveris, OraDisc A, EpiLift, MammoSite - Medscape, accessed August 6, 2025, https://www.medscape.com/viewarticle/492108
  24. Lutropin Alfa Dosage Guide + Max Dose, Adjustments - Drugs.com, accessed August 6, 2025, https://www.drugs.com/dosage/lutropin-alfa.html
  25. Study Details | A Clinical Trial to Determine the Effect of Lutropin Alfa on Embryo Quality and Implantation Rate in Advanced Reproductive Age | ClinicalTrials.gov, accessed August 6, 2025, https://clinicaltrials.gov/study/NCT01075815
  26. A Phase II Study to Assess the Efficacy and Safety of Luveris® (Lutropin Alfa) in Mid Follicular Phase for Controlled Ovarian Stimulation (COS) in Advanced Reproductive Age | ClinicalTrials.gov, accessed August 6, 2025, https://clinicaltrials.gov/study/NCT01079949?term=NCT01079949%20NCT02333253%20NCT02640976%20NCT02823080%20NCT03809429%20NCT04537078&rank=6
  27. Study Details | The Luveris In Vitro Fertilization Trial | ClinicalTrials.gov, accessed August 6, 2025, https://clinicaltrials.gov/study/NCT00889512
  28. Nordic Luveris Study - ClinicalTrials.gov, accessed August 6, 2025, https://clinicaltrials.gov/study/NCT00553293
  29. Lutropin alfa Completed Phase N/A Trials for Obesity | DrugBank Online, accessed August 6, 2025, https://go.drugbank.com/drugs/DB00044/clinical_trials?conditions=DBCOND0015947&status=completed
  30. Luveris® (Lutropin Alfa for Injection) in Women With Hypogonadotropic Hypogonadism (Luteinizing Hormone [LH] Less Than [<] 1.2 International Unit Per Liter [IU/L]) | ClinicalTrials.gov, accessed August 6, 2025, https://clinicaltrials.gov/study/NCT00328926
  31. Phase 3 Study to Evaluate the Efficacy and Safety of Pergoveris® in Assisted Reproductive Technology (ESPART) | ClinicalTrials.gov, accessed August 6, 2025, https://www.clinicaltrials.gov/study/NCT02047227?term=CHORIOGONADOTROPIN%20ALFA%20AND%20CHORIONIC%20GONADOTROPIN&rank=6
  32. Phase 3 Study to Evaluate the Efficacy and Safety of Pergoveris® in Assisted Reproductive Technology (ESPART) | ClinicalTrials.gov, accessed August 6, 2025, https://clinicaltrials.gov/study/NCT02047227
  33. Luveris - NPS MedicineWise, accessed August 6, 2025, https://www.nps.org.au/medicine-finder/luveris-75-iu-powder-for-injection
  34. Lutropin Alfa: Fertility Uses, Side Effects, Dosage - MedicineNet, accessed August 6, 2025, https://www.medicinenet.com/lutropin_alfa/article.htm
  35. Luveris N-18, accessed August 6, 2025, https://ec.europa.eu/health/documents/community-register/2003/200303046360/anx_6360_en.pdf
  36. Luveris: Uses, Dosage & Side Effects - Drugs.com, accessed August 6, 2025, https://www.drugs.com/luveris.html
  37. Lutropin alfa, r-hLH injection - Cleveland Clinic, accessed August 6, 2025, https://my.clevelandclinic.org/health/drugs/19679-lutropin-alfa-r-hlh-injection
  38. Consumer Information for: LUVERIS - Drug and Health Products Portal, accessed August 6, 2025, https://dhpp.hpfb-dgpsa.ca/dhpp/resource/75446/consumer-information
  39. Luveris - Merck Europe B.V., accessed August 6, 2025, https://www.drugfuture.com/ema/epar.aspx?ProductNumber=EMEA/H/C/000292
  40. Pergoveris, INN-follitropin alfa and lutropin alfa - EMA, accessed August 6, 2025, https://www.ema.europa.eu/en/documents/product-information/pergoveris-epar-product-information_en.pdf
  41. The legally binding text is the original French version TRANSPARENCY COMMITTEE OPINION 25 June 2008 PERGOVERIS 150 IU/75 IU, po, accessed August 6, 2025, https://www.has-sante.fr/jcms/c_1002746/fr/pergoveris-ct-5472-version-anglaise
  42. Luveris (lutropin alfa) FDA Approval History - Drugs.com, accessed August 6, 2025, https://www.drugs.com/history/luveris.html
  43. Drug Approval Package: Luveris (Lutropin Alfa) NDA #021322 - accessdata.fda.gov, accessed August 6, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2004/021322s000_LuverisTOC.cfm
  44. Efficacy of Follicle-Stimulating Hormone (FSH) Alone ... - Frontiers, accessed August 6, 2025, https://www.frontiersin.org/journals/endocrinology/articles/10.3389/fendo.2017.00114/full
  45. Comparative study of highly purified HMG versus recombinant ..., accessed August 6, 2025, https://www.elsevier.es/en-revista-medicina-reproductiva-embriologia-clinica-390-articulo-comparative-study-highly-purified-hmg-S2340932015000389
  46. Gonal-F vs. Menopur Treatment for Infertility: Differences and Side Effects - RxList, accessed August 6, 2025, https://www.rxlist.com/gonal-f_vs_menopur/drugs-condition.htm
  47. Urinary Versus Recombinant Gonadotropins for Ovarian Stimulation ..., accessed August 6, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6094533/
  48. A retrospective, matched case-control study of recombinant LH versus hMG supplementation on FSH during controlled ovarian hyperstimulation in the GnRH-antagonist protocol - Frontiers, accessed August 6, 2025, https://www.frontiersin.org/journals/endocrinology/articles/10.3389/fendo.2022.931756/full
  49. Urinary hMG (Meropur) versus recombinant FSH plus recombinant LH (Pergoveris) in IVF: a multicenter, prospective, randomized controlled trial - PubMed, accessed August 6, 2025, https://pubmed.ncbi.nlm.nih.gov/20537626/
  50. Fertility Treatment Medication - Instituto Bernabeu, accessed August 6, 2025, https://www.institutobernabeu.com/en/blog/fertility-treatment-medication/
  51. Efficacy and Safety of Pergoveris in Assisted Reproductive Technology—ESPART: rationale and design of a randomised controlled trial in poor ovarian responders undergoing IVF/ICSI treatment | BMJ Open, accessed August 6, 2025, https://bmjopen.bmj.com/content/5/7/e008297
  52. Efficacy and safety of follitropin alfa/lutropin alfa in ART: a randomized controlled trial in poor ovarian responders - PMC, accessed August 6, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5850777/

Published at: August 6, 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.