Comprehensive Report on Chorionic Gonadotropin (Human) (DrugBank ID: DB09126)
I. Introduction to Chorionic Gonadotropin (Human) (DB09126)
A. Overview and Clinical Significance
Chorionic Gonadotropin (Human), commonly referred to as hCG, is a polypeptide hormone naturally synthesized by the syncytiotrophoblast cells of the placenta during pregnancy.[1] As a therapeutic agent, specifically identified by DrugBank ID DB09126, it is formulated as a highly purified, pyrogen-free preparation derived from the urine of pregnant females.[3] The hormone's primary physiological role is the maternal recognition of pregnancy and maintenance of the corpus luteum, ensuring continued progesterone secretion crucial for sustaining the early stages of gestation.[1]
The clinical utility of exogenous hCG stems from its ability to mimic the biological actions of endogenous luteinizing hormone (LH) and, to a lesser extent, follicle-stimulating hormone (FSH).[3] This LH-like activity allows hCG to stimulate gonadal steroid hormone production. Consequently, its principal therapeutic applications include the management of prepubertal cryptorchidism (undescended testes not due to anatomical obstruction) and hypogonadotropic hypogonadism (characterised by deficient pituitary gonadotropin secretion) in selected male populations. In females, hCG is instrumental in inducing final follicular maturation and ovulation in appropriately selected infertile women, often as a critical component of assisted reproductive technology (ART) protocols.[4] The importance of hCG in reproductive medicine is underscored by its direct action on gonadal cells, promoting steroidogenesis and gametogenesis where endogenous gonadotropic support is insufficient or mistimed.
The transition from urinary-derived hCG to recombinant hCG formulations reflects a significant trend in biopharmaceutical development. This evolution aims for enhanced purity, improved batch-to-batch consistency, and potentially reduced immunogenicity.[8] While these advancements are notable, comparative clinical studies often indicate that the differences in primary clinical outcomes, such as live birth rates, may not always be statistically significant between the two forms, suggesting that the inherent biological activity of the hormone, once administered, can be a dominant factor for certain endpoints.[10] The inherent nature of hCG as a placental hormone vital for pregnancy maintenance provides a direct biological rationale for its therapeutic applications in mimicking or augmenting natural reproductive processes. Its endogenous function in sustaining the corpus luteum and progesterone output is directly leveraged in fertility treatments to trigger ovulation and support the luteal phase, while its action on testicular Leydig cells to stimulate androgen production is a direct extension of its LH-like properties.[1]
B. Scope of the Report: Focus on Urinary-Derived hCG (DB09126) with Comparative Insights
This report will primarily concentrate on urinary-derived Chorionic Gonadotropin (Human), as identified by DrugBank ID DB09126 and CAS Number 9002-61-3. Given the substantial body of research and clinical relevance of recombinant choriogonadotropin alfa (e.g., Ovidrel®, DrugBank ID DB00097) [13], this document will incorporate comparative data and highlight distinct characteristics of recombinant forms where pertinent. This approach is intended to provide a comprehensive understanding, particularly in discussions concerning pharmacokinetics, product purity, immunogenicity, and clinical efficacy. Such distinction is vital, as the available literature frequently addresses both urinary and recombinant forms, sometimes without explicit differentiation.
C. Historical Context and Evolution of hCG Preparations
Historically, therapeutic hCG was exclusively obtained through extraction and purification from the urine of pregnant women, a practice dating back to early research on gonadotropins.[1] This method, while effective, presented challenges related to product purity, potential for protein contaminants, and batch-to-batch variability.[9] The advent of recombinant DNA technology revolutionized the production of glycoprotein hormones, leading to the development of choriogonadotropin alfa. This recombinant form is produced in cell cultures (e.g., Chinese Hamster Ovary cells) and offers a higher degree of purity and consistency.[1] This technological advancement has also influenced administration routes, with recombinant hCG often formulated for subcutaneous injection, enhancing patient convenience compared to the traditional intramuscular route for many urinary-derived products.[8]
II. Physicochemical Characteristics and Molecular Structure
A. General Chemical Properties
Chorionic Gonadotropin (Human) (CAS Number: 9002-61-3) is classified as a biotech drug and is fundamentally a glycoprotein hormone.[1] In its purified, lyophilized form, it typically appears as a white or yellowish-white amorphous powder.[17] It exhibits solubility in water, a characteristic essential for its formulation as an injectable medication.[17] The FDA Unique Ingredient Identifier (UNII) for Chorionic Gonadotropin (Human) is 20ED16GHEB.[1]
B. Molecular Structure and Composition
Human chorionic gonadotropin is a heterodimeric glycoprotein, meaning it is composed of two distinct, non-covalently associated polypeptide subunits: an alpha (α) subunit and a beta (β) subunit.[1]
The alpha (α) subunit consists of 92 amino acids and has an approximate molecular mass of 14.5 kDa.[1] A critical structural feature of the α-subunit is its high degree of homology; it is essentially identical to the alpha subunits found in other human pituitary gonadotropins, namely luteinizing hormone (LH), follicle-stimulating hormone (FSH), and thyroid-stimulating hormone (TSH).[1] The amino acid sequence for the common alpha chain is APDVQDCPECTLQENPFFSQPGAPILQCMGCCFSRAYPTPLRSKKTMLVQKNVTSESTCCVAKSYNRVTVMGGFKVENHTACHCSTCYYHKS.[4] This shared α-subunit structure, while facilitating common receptor interaction motifs when paired with various β-subunits, also presents a practical challenge in clinical diagnostics. Specifically, it can lead to cross-reactivity in immunoassays designed to measure individual gonadotropin levels, potentially complicating the interpretation of results when hCG is administered exogenously or is endogenously elevated.[21]
The beta (β) subunit is unique to hCG and is responsible for conferring its specific biological activity and receptor binding specificity.[1] It contains 145 amino acids, with an approximate molecular mass of 22.2 kDa.[1] Some sources mention 149 amino acids, which may refer to the precursor or a slightly different isoform, but 145 amino acids is more consistently reported for the mature protein.[16] The amino acid sequence for the beta chain is SKEPLRPRCRPINATLAVEKEGCPVCITVNTTICAGYCPTMTRVLQGVLPALPQVVCNYR DVRFESIRLPGCPRGVNPVVSYAVALSCQCALCRRSTTDCGGPKDHPLTCDDPRFQDSSS SKAPPPSLPSPSRLPGPSDTPILPQ.[4]
The total molecular mass of the heterodimeric hCG molecule is approximately 36.7 kDa.[1] However, due to extensive and variable glycosylation, the apparent molecular weight can differ. For instance, choriogonadotropin alfa (recombinant) has a reported chemical formula of C1105H1770N318O336S26 and a molar mass of 25719.77 g/mol, reflecting its highly glycosylated state.[1] Urinary-derived hCG preparations can exhibit a molecular weight of approximately 38,600 Da.[18] This variability is a key characteristic of glycoprotein hormones.
Glycosylation is a significant post-translational modification of hCG. The β-subunit features a distinctive C-terminal peptide (CTP) that is not present in the β-subunit of LH. This CTP contains several serine residues that are sites for O-linked glycosylation.[1] These O-linked carbohydrate chains on the CTP are crucial as they protect the hormone from rapid metabolic degradation and renal clearance, thereby significantly extending hCG's circulatory half-life compared to LH. This prolonged duration of action is fundamental to hCG's physiological role in maintaining early pregnancy and also contributes to its pharmacological efficacy and risk profile, notably the potential for ovarian hyperstimulation syndrome (OHSS) due to sustained gonadal stimulation.[1] Both subunits also possess N-linked carbohydrate moieties; for recombinant hCG, these are typically at ASN-52 and ASN-78 on the α-subunit, and ASN-13 and ASN-30 on the β-subunit.[13] The patterns of glycosylation can vary between hCG produced by the placenta, the pituitary gland (which produces small amounts of hCG with different glycosylation), and recombinant manufacturing processes.[1] These "glycoforms" can exhibit differences in biological activity, receptor binding affinity, and activation of intracellular signaling pathways, which may translate to variations in clinical efficacy and safety.[1] Urinary hCG, being a pool of naturally produced molecules, inherently contains a heterogeneous mixture of these glycoforms, contrasting with the more defined composition of specific recombinant products.[3] This heterogeneity in urinary preparations can contribute to batch-to-batch variability and potentially influence clinical responses.[9]
The three-dimensional structure of hCG reveals that the two subunits associate to form a compact molecule with a small hydrophobic core. The exterior is predominantly hydrophilic.[1] Each subunit incorporates a cystine-knot motif, characterized by specific disulfide bond arrangements, from which extended hairpin loops emerge. The interface between the α and β subunits is extensive and features two inter-chain β-sheets. A unique structural element is a disulfide-tethered "arm" extending from the β-subunit, which "embraces" the α-subunit, contributing to the stability and conformation of the heterodimer.[20] The carboxy-terminal peptide of the β-subunit, rich in O-linked sugars, is structurally disordered.[20]
C. Source and Purification (for DB09126)
Chorionic Gonadotropin (Human) with DrugBank ID DB09126 is sourced from the urine of pregnant females.[1] The manufacturing process involves extensive purification steps to yield a highly purified, pyrogen-free preparation suitable for therapeutic use. The potency of the final drug product is standardized using a biological assay procedure to ensure consistent activity.[3]
D. Forms of hCG
Several forms of hCG exist physiologically and can be detected. These include intact, regular hCG (the predominant form in pregnancy and in urinary-derived drug products), hyperglycosylated hCG (prominent in early pregnancy, invasive molar pregnancies, and choriocarcinoma), the free β-subunit of hCG (also a tumor marker), and various degradation products such as nicked hCG and hCG missing the C-terminal peptide.[1] The pituitary gland also produces small amounts of hCG, but its glycosylation pattern differs from that of placental hCG.[1]
Table 1: Physicochemical Properties of Chorionic Gonadotropin (Human) (DB09126)
Property | Details | Key Sources |
---|
DrugBank ID | DB09126 | User Query, 4 |
CAS Number | 9002-61-3 | User Query, 1 |
UNII | 20ED16GHEB | 1 |
Type | Biotech, Glycoprotein Hormone | User Query, 1 |
Source (for DB09126) | Human Pregnancy Urine | User Query, 1 |
Appearance | White or yellowish-white, amorphous powder | 17 |
Solubility | Soluble in water | 17 |
Molecular Composition | Heterodimer of α and β subunits | User Query, 1 |
Alpha (α) Subunit | 92 amino acids; approx. 14.5 kDa; identical to α-subunits of LH, FSH, TSH | 1 |
Beta (β) Subunit | 145 amino acids (unique to hCG); approx. 22.2 kDa; contains C-terminal peptide (CTP) with O-linked glycosylation | 1 |
Overall Molecular Mass | Approx. 36.7 kDa (native); urinary hCG approx. 38.6 kDa; variable due to extensive N- and O-linked glycosylation | 1 |
Key Structural Features | Cystine-knot motif in each subunit; β-subunit CTP crucial for extended half-life; extensive glycosylation | 1 |
Purification (DB09126) | Highly purified, pyrogen-free preparation from urine; standardized by biological assay | User Query, 3 |
III. Mechanism of Action and Cellular Pharmacology
A. LHCG Receptor (LHCGR) Interaction and Primary Physiological Roles
Chorionic Gonadotropin (Human) exerts its biological effects through specific binding to the Luteinizing Hormone/Chorionic Gonadotropin Receptor (LHCGR). The LHCGR is a member of the G protein-coupled receptor (GPCR) superfamily, characterized by seven transmembrane domains.[1] These receptors are predominantly expressed on the surface of target cells in the gonads: specifically, on granulosa, theca, and luteal cells of the ovary, and on Leydig cells of the testis.[30] Expression of LHCGR has also been identified in various extragonadal tissues, including the uterus, sperm, seminal vesicles, prostate, skin, breast, adrenals, and thyroid, suggesting broader physiological roles beyond reproduction, although these are less well characterized.[27] The presence of LHCGR in tissues like the endometrium, where hCG can modulate immune responses (e.g., by inducing T-cell apoptosis and influencing cytokine production), points to functions in maternal immunotolerance and implantation.[1]
The pharmacological action of hCG is virtually identical to that of pituitary-derived LH, as both hormones bind to and activate the same LHCGR.[4] Some evidence also suggests that hCG possesses a minor degree of FSH-like activity.[4]
In Females:
The primary roles of hCG in female reproductive physiology, and consequently its therapeutic applications, revolve around ovulation and luteal support.
- Ovulation Induction: In natural cycles, a mid-cycle surge of LH is the proximate trigger for final follicular maturation, resumption of oocyte meiosis, and the rupture of the dominant ovarian follicle (ovulation). Exogenously administered hCG mimics this LH surge. In clinical settings, particularly in ovulation induction protocols or ART, hCG is administered after ovarian stimulation with FSH or human menopausal gonadotropins (hMG) to ensure the timely maturation and release of oocytes.[13]
- Corpus Luteum Maintenance: Following ovulation, hCG promotes the formation, maintenance, and function of the corpus luteum. The corpus luteum is critical for producing progesterone, a steroid hormone essential for preparing the endometrium for embryo implantation and sustaining the early stages of pregnancy.[1] Endogenously, placental hCG takes over this role from pituitary LH once pregnancy is established.
In Males:
In males, hCG's actions are primarily directed at testicular steroidogenesis and its consequences.
- Stimulation of Androgen Production: hCG binds to LHCGR on the Leydig cells within the testes, stimulating these cells to synthesize and secrete androgens, predominantly testosterone.[3] Testosterone is vital for the development and maintenance of male secondary sexual characteristics, libido, and overall male reproductive health.
- Induction of Testicular Descent: In cases of prepubertal cryptorchidism where there is no anatomical blockage, hCG is believed to promote testicular descent by stimulating Leydig cell testosterone production, thereby mimicking the hormonal milieu that normally facilitates this process during puberty.[3]
- Support of Spermatogenesis: Adequate intratesticular testosterone, stimulated by hCG (acting as an LH surrogate), is essential for the initiation and maintenance of spermatogenesis within the seminiferous tubules. This is particularly relevant in the treatment of hypogonadotropic hypogonadism, where endogenous LH production is deficient.[6]
B. Intracellular Signaling Pathways
The binding of hCG to the LHCGR initiates a complex array of intracellular signaling cascades, which are fundamental to its diverse physiological and pharmacological effects. These pathways involve the activation of heterotrimeric G proteins, generation of second messengers, and modulation of various protein kinases and transcription factors.[2] The sophisticated integration of these signals ultimately dictates the cellular response.
1. Canonical Gs/cAMP/PKA Pathway:
This is considered the principal and most extensively characterized signaling pathway activated by LHCGR.
- Activation: Upon hCG binding, LHCGR couples to the stimulatory G protein (Gαs). Activated Gαs stimulates the enzyme adenylyl cyclase, which catalyzes the conversion of ATP to cyclic AMP (cAMP).[26]
- Downstream Effects: The rise in intracellular cAMP levels leads to the activation of Protein Kinase A (PKA). PKA, a serine/threonine kinase, then phosphorylates a multitude of downstream target proteins. These include enzymes involved in steroidogenesis (e.g., Cholesterol Side-chain Cleavage Enzyme (P450scc), Steroidogenic Acute Regulatory Protein (StAR)) and transcription factors such as CREB (cAMP response element-binding protein). Phosphorylated CREB translocates to the nucleus and binds to cAMP response elements (CREs) in the promoter regions of target genes, thereby regulating their expression.[27]
- Physiological Relevance: This pathway is paramount for the acute stimulation of steroidogenesis in both ovarian cells (leading to the production of progesterone and estrogen precursors) and testicular Leydig cells (leading to testosterone synthesis).[2] It also plays roles in follicular maturation and luteinization.
2. Gq/11/Phospholipase C (PLC) Pathway:
LHCGR can also couple to Gαq/11 proteins, initiating an alternative signaling cascade.
- Activation: Activated Gαq/11 stimulates Phospholipase Cβ (PLCβ). PLCβ then hydrolyzes phosphatidylinositol 4,5-bisphosphate (PIP2), a membrane phospholipid, into two second messengers: inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG).[27]
- Downstream Effects: IP3 diffuses into the cytoplasm and binds to IP3 receptors on the endoplasmic reticulum, triggering the release of stored calcium ions (Ca2+) into the cytosol. The increased intracellular Ca2+ can activate various calcium-dependent enzymes and processes. DAG, in conjunction with Ca2+, activates members of the Protein Kinase C (PKC) family.[27]
- Physiological Relevance: The Gq/11-PLC-Ca2+/PKC pathway is involved in modulating steroidogenesis, often synergizing with or fine-tuning the cAMP/PKA pathway. It also contributes to the regulation of cell proliferation, differentiation, and survival in gonadal cells.[27]
3. Mitogen-Activated Protein Kinase (MAPK)/ERK Pathway:
hCG binding to LHCGR can also lead to the activation of Mitogen-Activated Protein Kinase (MAPK) cascades, most notably the Extracellular signal-Regulated Kinase (ERK1/2) pathway.
- Activation Mechanisms: ERK1/2 activation can occur through several mechanisms downstream of LHCGR, including cross-talk from the cAMP/PKA pathway, activation via the Gq/11-PKC pathway, or through β-arrestin-mediated scaffolding of signaling complexes.[27] For example, hCG has been shown to induce leptin expression in trophoblast cells via a MAPK-dependent mechanism.[47]
- Downstream Effects: Activated ERK1/2 can phosphorylate a wide range of cytoplasmic and nuclear substrates, including transcription factors, thereby influencing gene expression.
- Physiological Relevance: The MAPK/ERK pathway is critically involved in regulating cellular processes such as proliferation, differentiation, survival, and apoptosis. In gonadal cells, it plays a role in modulating steroidogenesis and supporting cell growth and viability.[26]
4. Phosphatidylinositol 3-Kinase (PI3K)/AKT Pathway:
The PI3K/Protein Kinase B (AKT) signaling pathway is another important cascade activated by hCG through LHCGR.
- Activation: Activation of PI3K leads to the generation of phosphatidylinositol (3,4,5)-trisphosphate (PIP3), which serves as a docking site for proteins containing pleckstrin homology (PH) domains, such as AKT (also known as Protein Kinase B) and phosphoinositide-dependent kinase 1 (PDK1). This recruitment to the membrane facilitates the phosphorylation and activation of AKT by PDK1 and other kinases like mTORC2.[26]
- Downstream Effects: Activated AKT phosphorylates numerous substrates involved in regulating cell survival (e.g., by inhibiting pro-apoptotic proteins like BAD and Forkhead box O (FOXO) transcription factors), cell proliferation (e.g., by phosphorylating cell cycle regulators like GSK3β and activating mTORC1), and cellular metabolism.
- Physiological Relevance: The PI3K/AKT pathway is crucial for promoting cell survival by inhibiting apoptosis, stimulating cell growth and proliferation, and regulating glucose metabolism. In the context of hCG action, it has been shown that hCG stimulates theca-interstitial cell proliferation via a cAMP-dependent activation of AKT and its downstream target, mammalian target of rapamycin complex 1 (mTORC1).[44]
5. β-Arrestin Mediated Signaling and Receptor Regulation:
β-arrestins play a dual role in LHCGR signaling: desensitization/internalization and G protein-independent signaling.
- Desensitization and Internalization: Following prolonged or strong agonist (hCG) binding, LHCGR becomes phosphorylated by GPCR kinases (GRKs). Phosphorylated LHCGR then recruits β-arrestins (β-arrestin 1 and 2).[27] β-arrestin binding sterically hinders further G protein coupling (desensitization) and targets the receptor for internalization via clathrin-coated pits (endocytosis). Internalized receptors can then be either dephosphorylated and recycled back to the plasma membrane or targeted for lysosomal degradation, which determines the long-term responsiveness of the cell.[27] The adaptor protein APPL1 is involved in gonadotropin receptor signaling from endosomal compartments.[27]
- Scaffolding and Non-Canonical Signaling: Beyond their role in desensitization, β-arrestins can act as scaffolding proteins, assembling and activating distinct signaling complexes, including components of the MAPK/ERK pathway, independently of G protein activation. This "non-canonical" signaling from internalized receptors can contribute to sustained cellular responses.[27]
6. Biased Agonism (hCG vs. LH):
A significant concept in LHCGR pharmacology is "biased agonism" or "functional selectivity." Although hCG and LH bind to the same orthosteric site on LHCGR, they can stabilize different receptor conformations, leading to preferential activation of certain downstream signaling pathways or G protein subtypes over others. This results in quantitatively or qualitatively distinct cellular responses.26
- Differential Pathway Activation: Evidence suggests that hCG is generally a more potent and sustained activator of the cAMP/PKA pathway and steroidogenesis (particularly progesterone synthesis) compared to LH. hCG also tends to induce more robust β-arrestin recruitment and subsequent receptor internalization and downregulation.[26] In contrast, LH may show a preference for activating PI3K/AKT and ERK pathways, which are more associated with cell proliferation, differentiation, and survival, with a potentially less intense acute steroidogenic response.[26]
- Structural Basis: These differences in signaling bias are thought to be mediated by subtle differences in the interaction of hCG and LH with the LHCGR, particularly involving the extracellular hinge region of the receptor and variations in the glycosylation patterns of the hormones themselves.[28] The unique C-terminal peptide of hCG, with its extensive glycosylation, not only contributes to its longer half-life but may also influence receptor interaction and signaling outcomes.
- Clinical Implications: This biased agonism has profound clinical implications. hCG's potent and sustained activation of cAMP makes it an effective agent for inducing a robust ovulatory trigger and providing initial luteal phase support. However, this same characteristic, coupled with its longer half-life, contributes significantly to the risk of OHSS due to prolonged and potentially excessive ovarian stimulation. LH or its analogs, which might elicit a more balanced or different profile of signaling, could theoretically be favored in situations requiring more nuanced gonadal stimulation or where specific cellular responses (e.g., proliferation over acute steroidogenesis) are desired, though the clinical translation of these nuanced differences is still an area of active research.[5]
The complex interplay between these multiple signaling pathways (cAMP/PKA, PLC/Ca2+/PKC, MAPK/ERK, PI3K/AKT, β-arrestin) activated by the hCG-LHCGR interaction underscores the sophisticated mechanisms by which cells integrate hormonal signals. The balance and crosstalk between these pathways, which can be modulated by cell type, receptor density, the presence of co-receptors (like FSHR forming heterodimers with LHCGR), and other growth factors, ultimately determine the specific physiological or pharmacological outcome. This intricate signaling network allows for a fine-tuned cellular response rather than a simple "on/off" switch, highlighting the complexity of gonadotropin action.
C. Pharmacodynamics
The pharmacodynamic effects of Chorionic Gonadotropin (Human) are a direct consequence of its interaction with LHCGR and the subsequent activation of intracellular signaling pathways, leading to specific physiological responses in target gonadal tissues.
- Onset and Duration of Action: Following intramuscular (IM) administration of urinary hCG (e.g., Pregnyl®), an increase in serum hCG concentrations can be detected within 2 hours. Peak serum concentrations are typically reached within 6 hours, and these elevated levels persist, exerting biological effects, for approximately 36 hours. Subsequently, serum hCG levels begin to decline around 48 hours post-administration, becoming undetectable after about 72 hours.[18] This relatively prolonged duration of action, especially when compared to the much shorter half-life of endogenous LH, is a key characteristic of hCG and is largely attributable to its molecular structure, particularly the glycosylated C-terminal peptide of the β-subunit which protects it from rapid degradation.
- Effects on Ovulation: When used for ovulation induction, hCG administration (typically 5,000 to 10,000 IU) triggers the final maturation of the ovarian follicle and induces ovulation approximately 36 to 48 hours after the injection.[24] This timing is critical for coordinating with timed intercourse or assisted reproductive procedures like intrauterine insemination (IUI) or oocyte retrieval for in vitro fertilization (IVF).
- Effects on Steroidogenesis: hCG potently stimulates gonadal steroid hormone production.
- In Males: Administration of hCG leads to a significant increase in serum testosterone levels due to its action on testicular Leydig cells. Studies have shown comparable increases in serum testosterone, inhibin, and 17β-estradiol following intravenous (IV), IM, or subcutaneous (SC) administration of hCG.[3] This androgenic effect is responsible for the development of secondary sexual characteristics in hypogonadal males.
- In Females: hCG stimulates the corpus luteum to produce and secrete progesterone, and to a lesser extent, estradiol. This luteal support is crucial for endometrial receptivity and the maintenance of early pregnancy.[1]
- Effects on Testicular Descent: In prepubertal boys with cryptorchidism not caused by an anatomical obstruction, hCG administration may induce testicular descent. This effect is mediated by the hCG-stimulated increase in testosterone, which mimics the hormonal changes of puberty that normally facilitate descent. The response can be temporary, with the testes sometimes re-ascending after treatment, although permanent descent is achieved in some cases.[3]
- Other Potential Physiological Effects: Beyond its primary gonadal actions, hCG has been implicated in modulating immune responses, particularly at the maternal-fetal interface. For example, hCG may contribute to local maternal immunotolerance by inducing apoptosis in T cells within the endometrium, potentially facilitating trophoblast invasion and embryo implantation.[1]
IV. Pharmacokinetics (ADME)
The pharmacokinetic profile of Chorionic Gonadotropin (Human) describes its absorption, distribution, metabolism, and excretion (ADME) following administration. These characteristics differ slightly between urinary-derived hCG (DB09126) and recombinant choriogonadotropin alfa (DB00097), primarily due to formulation and purity differences.
A. Absorption
Chorionic Gonadotropin (Human) is administered parenterally, as it is a polypeptide hormone that would be degraded in the gastrointestinal tract if given orally.
- Routes of Administration: Urinary-derived hCG (e.g., Pregnyl®, Novarel®) is typically administered via the intramuscular (IM) route.[3] Recombinant forms like choriogonadotropin alfa (e.g., Ovidrel®) are commonly administered subcutaneously (SC), offering greater convenience for self-administration.[13] Some sources suggest urinary hCG may also be administered SC.[6]
- Bioavailability:
- For recombinant hCG (choriogonadotropin alfa), the mean absolute bioavailability following a single SC injection is approximately 40%.[13] This indicates that a significant portion of the administered dose does not reach systemic circulation, a factor that must be considered in dosing.
- For urinary hCG, IM or SC administration results in a comparable bioavailability of 40-50%.[48]
- Time to Peak Concentration (Tmax):
- Urinary hCG (e.g., Pregnyl®, IM): Peak serum concentrations are generally observed within 6 hours post-injection.[18]
- Recombinant hCG (e.g., Ovidrel®, SC): The time to reach peak plasma concentration is typically longer, ranging from 12 to 24 hours.[33]
B. Distribution
Once absorbed into the systemic circulation, hCG distributes to its target tissues.
- Tissue Distribution: The hormone primarily distributes to the gonads – the testes in males and the ovaries in females – where LHCGRs are highly expressed.[29]
- Volume of Distribution (Vd):
- For recombinant hCG (Ovidrel®), reported Vd values vary; some sources indicate 5.9 ± 1.0 L [13], while others state 21.4 L.[33] This discrepancy may reflect differences in study populations, specific product formulations, or calculation methods.
- Protein Binding: Specific plasma protein binding information for hCG is generally not extensively detailed or is listed as "Not Available".[13] As a large glycoprotein, its binding characteristics may differ from small molecule drugs.
C. Metabolism
hCG undergoes metabolic degradation in the body.
- Primary Site of Catabolism: The liver is the primary organ responsible for the catabolism of intact hCG.[2]
- Renal Degradation: The kidneys also play a role in hCG metabolism. Specifically, the β-subunit of hCG is degraded into a core fragment within the kidney.[2] This β-core fragment is a major urinary metabolite and is often the target analyte in urine-based pregnancy tests.
- Detailed metabolic pathways for choriogonadotropin alfa are often cited as "Not Available" in drug summaries.[13]
D. Excretion
The elimination of hCG and its metabolites occurs primarily via the kidneys.
- Urinary Excretion: Approximately 10-20% of an administered dose of hCG is excreted unchanged in the urine within 24 hours.[2] This renal excretion of active hormone is the basis for its original extraction from the urine of pregnant women and is also the reason why exogenous hCG administration can lead to false-positive pregnancy test results.[18] The renal handling of hCG implies that kidney function can significantly influence its clearance. Impaired renal function could lead to reduced clearance, prolonged exposure, and potentially higher systemic concentrations of hCG, which might increase the risk of adverse effects such as OHSS or necessitate dose adjustments, a common consideration for drugs with significant renal elimination pathways.[21]
E. Half-Life
The elimination half-life of hCG is biphasic, reflecting an initial distribution phase followed by a slower terminal elimination phase.
- Urinary hCG (e.g., Novarel®, Pregnyl®):
- Initial half-life (t½α): Reported to be between 5.6 and 11 hours.[18]
- Terminal half-life (t½β): Ranges from 23 to 37.2 hours.[18]
- Recombinant hCG (choriogonadotropin alfa, e.g., Ovidrel®):
- Initial half-life (t½α): Approximately 4.0 to 4.5 ± 0.5 hours.[13]
- Mean terminal half-life (t½β): About 29 ± 6 hours.[13]
The relatively long terminal half-life of hCG (around 23-37 hours for urinary forms and ~29 hours for recombinant forms), especially when compared to the much shorter half-life of endogenous LH, is a critical pharmacokinetic characteristic. This extended presence in the circulation underpins its sustained biological activity, which is essential for its therapeutic efficacy in indications requiring continuous gonadal stimulation, such as corpus luteum maintenance or prolonged androgen production. However, this same pharmacokinetic property – the prolonged action – is also a key factor contributing to the risk of OHSS, as it can lead to sustained and potentially excessive ovarian stimulation.[1]
Pharmacokinetic studies indicate that hCG follows linear kinetics. After IV administration, its disposition can be described by a bi-exponential model, while after IM or SC administration, a first-order absorption, one-compartment model is often applicable.[48]
F. Comparative Pharmacokinetics (Urinary vs. Recombinant)
Studies comparing urinary-derived hCG (uHCG) and recombinant hCG (rHCG) have generally found their pharmacokinetic and pharmacodynamic profiles to be similar.[48]
- Bioequivalence: Pharmacokinetic bioequivalence has been demonstrated between liquid and freeze-dried formulations of rHCG, and also between freeze-dried rHCG and freeze-dried uHCG.[48]
- Area Under the Curve (AUC): One study noted that after single IV doses, the AUC for uHCG was approximately 29% lower than that for rHCG.[48] This finding, if consistent, could suggest potential differences in total systemic exposure for a given dose, which might have clinical implications for dosing or when comparing outcomes from trials using different hCG sources, despite the general label of "similar" pharmacokinetics. A lower AUC for uHCG might imply that either a higher dose is needed to achieve the same systemic levels as rHCG, or that there are differences in the purity or proportion of biologically active hormone in urinary preparations.
- Multiple Dosing: During multiple SC dosing regimens, the amount of circulating hCG has been observed to increase by approximately 1.7-fold, suggesting some accumulation with repeated administration.[48]
Table 2: Summary of Key Pharmacokinetic Parameters (Urinary vs. Recombinant hCG)
Parameter | Urinary hCG (DB09126 - e.g., Pregnyl®, Novarel®) | Recombinant hCG (DB00097 - e.g., Ovidrel®) | Key Sources |
---|
Typical Route(s) | IM | SC | 3 |
Bioavailability (IM/SC) | 40-50% | ~40% (SC) | 13 |
Tmax (IM/SC) | ~6 hours (IM) | 12-24 hours (SC) | 18 |
Initial Half-life (t½α) | 5.6 - 11 hours | 4.0 - 4.5 hours | 13 |
Terminal Half-life (t½β) | 23 - 37.2 hours | ~29 ± 6 hours | 13 |
Primary Metabolism Route | Liver (intact hCG), Kidney (β-subunit) | Assumed similar (liver, kidney) | 2 |
Primary Excretion Route | Urine (10-20% unchanged) | Urine (~10% unchanged) | 2 |
Volume of Distribution (Vd) | Not well specified | 5.9 L or 21.4 L (varies by source) | 13 |
V. Therapeutic Applications
Chorionic Gonadotropin (Human) has well-defined roles in reproductive medicine and endocrinology, stemming from its ability to mimic LH activity. Its applications span female infertility, male hypogonadism, and specific pediatric conditions, alongside diagnostic uses.
A. Approved Indications (FDA)
The Food and Drug Administration (FDA) has approved urinary-derived Chorionic Gonadotropin (Human) (DB09126) for the following indications:
1. Female Infertility (Ovulation Induction, Assisted Reproductive Technology - ART):
- Mechanism and Use: hCG is employed as an LH surrogate to trigger final follicular maturation and induce ovulation in anovulatory women or as part of controlled ovarian hyperstimulation protocols for ART procedures such as IVF.[6] It is typically administered one day after the completion of ovarian stimulation with follicle-stimulating hormone (FSH) or human menopausal gonadotropins (hMG).[3]
- Patient Selection and Monitoring: This therapy is indicated for women whose anovulation is secondary (e.g., hypothalamic-pituitary dysfunction, PCOS) and not due to primary ovarian failure. Treatment requires careful monitoring of ovarian response through transvaginal ultrasound and serum estradiol measurements, conducted by physicians experienced in infertility management, to optimize outcomes and minimize risks like OHSS.[3]
2. Male Hypogonadotropic Hypogonadism:
- Mechanism and Use: In males with hypogonadism secondary to pituitary insufficiency, hCG stimulates testicular Leydig cells to produce testosterone. This promotes virilization (development of secondary sexual characteristics) and can support spermatogenesis, especially when used in combination with FSH or hMG if endogenous FSH is also deficient.[3]
- Clinical Goal: Treatment aims to achieve normal serum testosterone levels, induce and maintain secondary sexual characteristics, and, if desired, initiate or restore fertility.
3. Prepubertal Cryptorchidism (Not Due To Anatomical Obstruction):
- Mechanism and Use: hCG is used in prepubertal boys (typically aged 4-9 years) with undescended testes where no physical obstruction to descent exists.[3] The hormone is thought to stimulate endogenous testosterone production, which can promote testicular descent in cases where descent would have naturally occurred at puberty.[3]
- Predictive Value and Efficacy: hCG administration may help predict whether future surgical intervention (orchiopexy) will be necessary. While testicular descent following hCG treatment can be permanent in some individuals, in many cases, the response is temporary.[3]
- Controversy and Current Recommendations: The utility of hCG for cryptorchidism is a subject of considerable debate. Despite being an FDA-approved indication and historical practice, numerous systematic reviews and meta-analyses have concluded that hCG is no more effective than placebo in achieving testicular descent.[66] Success rates reported in literature are highly variable (0-50.8% for unilateral, 0-22.4% for bilateral).[66] Consequently, major pediatric urology and endocrinology guidelines (e.g., Nordic consensus, EAU, AUA) generally do not recommend hormonal therapy as a primary treatment for cryptorchidism, favoring early orchiopexy (typically between 6-12 months of age).[65] This discrepancy between regulatory approval and evidence-based guidelines presents a clinical dilemma, possibly reflecting a lag in regulatory updates or a very narrow, poorly defined niche for its use (e.g., for truly retractile testes or as a diagnostic predictor).
B. Off-Label and Investigational Uses (Critical Assessment)
Beyond its approved indications, hCG has been explored or used for various other conditions, often without robust scientific backing.
- Weight Loss/Obesity: A prominent off-label use of hCG has been for weight loss, typically in conjunction with severely calorie-restricted diets. However, there is no substantial scientific evidence to support its efficacy for this purpose. Regulatory bodies like the FDA and professional organizations explicitly state that hCG is not effective for weight loss, does not cause a more "normal" distribution of fat, and does not decrease hunger associated with such diets.[5] The persistence of this misuse, despite strong disclaimers, highlights challenges in combating unproven therapies driven by popular trends and poses potential public health and safety risks due to inappropriate drug exposure without benefit.
- Kaposi Sarcoma: Isolated reports have suggested local regression of Kaposi sarcoma lesions with hCG treatment, but this is not an established therapy.[49]
- Sepsis (Investigational): Preliminary research and hypotheses suggest potential benefits of hCG or hCG-like microbial molecules in sepsis, possibly due to anti-inflammatory properties. This remains an area of investigation.[70]
- Resistant Ovary Syndrome (ROS): In ROS, where ovaries are hyporesponsive to gonadotropins despite normal follicle reserves, both urinary and recombinant gonadotropins, including hCG as an ovulatory trigger, have been used experimentally in COS protocols with some success, although treatment remains largely empirical due to the rarity of the condition.[53]
- Other unlisted uses may exist but require rigorous evaluation for evidence of efficacy and safety.[6]
C. Diagnostic Uses
hCG plays a vital role in several diagnostic contexts:
- hCG Stimulation Test (Assessment of Leydig Cell Function): This dynamic endocrine test is used to evaluate the functional capacity of testicular Leydig cells to produce testosterone in response to LH-like stimulation. It is particularly valuable in pediatric endocrinology for investigating disorders of sex development, micropenis, ambiguous genitalia, anorchia versus cryptorchidism, and differentiating hypogonadotropic hypogonadism from constitutional delay of puberty.[17] The test involves administering hCG (dosages vary by age/protocol, e.g., a 3-day test) and measuring baseline and stimulated testosterone levels.[71] A defined post-hCG testosterone response (e.g., a cut-off like 1.1 ng/mL in one study) can help predict future Leydig cell function and the potential need for long-term hormone replacement therapy.[71] This functional assessment provides more insight than static hormone measurements alone, especially when basal levels are low, as in prepubertal boys.
- Tumor Marker: Serum hCG, particularly its free β-subunit or hyperglycosylated forms, serves as an important biomarker for certain malignancies. Elevated levels are characteristic of gestational trophoblastic diseases (e.g., hydatidiform mole, choriocarcinoma), germ cell tumors of the testis or ovary, and occasionally other non-trophoblastic cancers that ectopically produce hCG.[1] Monitoring hCG levels is crucial for diagnosis, staging, assessing treatment response, and detecting recurrence in these conditions.
- Pregnancy Diagnosis and Monitoring: The detection of hCG in a woman's serum or urine is the cornerstone of pregnancy diagnosis. Quantitative serum hCG measurements, often performed serially, are essential for monitoring early pregnancy viability, assessing gestational age, diagnosing ectopic pregnancy, evaluating threatened abortion, and managing pregnancy loss.[1] Typically, hCG levels rise exponentially in early pregnancy, doubling approximately every 24-48 hours during the first 8 weeks.[2]
VI. Dosage, Administration, and Formulations
The dosage and administration of Chorionic Gonadotropin (Human) are highly dependent on the specific clinical indication, patient characteristics (age, weight), and the treating physician's preference and experience.[3]
A. Dosage Regimens (Indication-Specific)
The following regimens primarily pertain to urinary-derived hCG (e.g., Pregnyl®, Novarel®), which is the focus of DB09126.
1. Prepubertal Cryptorchidism (Not Due To Anatomical Obstruction; typically ages 4-9 years):
Several regimens have been advocated, reflecting variability in clinical practice 3:
- 4,000 USP units IM three times weekly for three weeks.
- 5,000 USP units IM every other day for four injections.
- 500 to 1,000 USP units IM for a total of 15 injections administered over a period of six weeks.
- 500 USP units IM three times weekly for four to six weeks. If this course of treatment is not successful, another course may be initiated one month later, using 1,000 USP units per injection. Dosages for the hCG stimulation test in pediatric patients also vary based on age or body surface area/weight.[72]
2. Hypogonadotropic Hypogonadism in Males:
Regimens aim to stimulate testosterone production and, if desired, spermatogenesis 3:
- For Virilization:
- 500 to 1,000 USP units IM three times a week for three weeks, followed by the same dose twice a week for three weeks.
- 4,000 USP units IM three times weekly for six to nine months. Subsequently, the dosage may be reduced to 2,000 USP units three times weekly for an additional three months.
- For Induction of Spermatogenesis: This often requires longer treatment durations and is typically combined with FSH or hMG preparations if endogenous FSH is also deficient. Common hCG dosages range from 1,000 to 2,000 USP units IM two to three times per week, potentially for several months (2-3 months or longer to see effects on spermatogenesis).[33] Recent reviews on physiological pubertal induction suggest potentially lower, gradually increasing doses of hCG, sometimes initiated before adding FSH.[42]
3. Induction of Ovulation and Pregnancy (in anovulatory, infertile women pretreated with menotropins/FSH):
hCG is administered to trigger final follicular maturation and ovulation 3:
- Urinary hCG (e.g., Pregnyl®, Novarel®): A single dose of 5,000 to 10,000 USP units IM, administered one day following the last dose of menotropins or FSH. A dosage of 10,000 USP units is often recommended in the labeling for menotropins.
- Recombinant hCG (e.g., Ovidrel®): A single dose of 250 mcg SC, administered one day following the last dose of the follicle-stimulating agent.[33] A 250 mcg dose of r-hCG is considered approximately equivalent to 5,000-7,000 IU of urinary hCG.[11] This dosing difference (IU for urinary vs. mcg for recombinant) may reflect variations in purity, specific activity, and historical dosing conventions. The higher IU dosage for urinary hCG might have been established to ensure sufficient delivery of active hormone, considering potential impurities and variability inherent in biological extracts, whereas purer recombinant products allow for more precise mass-based dosing.
B. Administration
- Route of Administration: Urinary-derived hCG (DB09126, e.g., Pregnyl®, Novarel®) is primarily approved for IM injection.[3] Recombinant choriogonadotropin alfa (e.g., Ovidrel®) is typically administered via SC injection.[6] Some clinical settings or patient preferences might lead to SC administration of urinary hCG, though this is less standard.[6]
- Reconstitution: Urinary hCG products are supplied as a sterile lyophilized powder in vials (commonly 5,000 or 10,000 USP units). Prior to administration, this powder must be reconstituted using the provided sterile diluent. This diluent is often Bacteriostatic Water for Injection containing a preservative such as benzyl alcohol (e.g., 0.9%).[3] The powder should be agitated gently after adding the diluent until the solution is complete and clear.[21]
- Self-Administration: Patients, particularly those undergoing fertility treatments, may be instructed by healthcare professionals on how to prepare and self-administer hCG injections, especially SC forms. Comprehensive training and adherence to sterile techniques are crucial. Proper disposal of used needles and syringes in a designated sharps container is mandatory to prevent injury and contamination.[6]
C. Marketed Formulations and Brand Names
- Urinary-derived hCG (DB09126 and similar):
- Brand Names: Pregnyl®, Novarel®, HCG, Profasi®.[1] Generic versions are also available.
- Formulation: Typically supplied as a sterile lyophilized powder for injection in multi-dose or single-dose vials, commonly containing 5,000 USP units or 10,000 USP units per vial. These are often packaged as kits that include the vial of hCG powder and a separate vial of sterile diluent.[3]
- Recombinant Choriogonadotropin Alfa (DB00097):
- Brand Names: Ovidrel®, Ovitrelle®.[6]
- Formulation: Available as a solution for injection in pre-filled syringes (e.g., 250 mcg in 0.5 mL) or as a powder for solution that requires reconstitution.[13] The pre-filled syringe formulation offers enhanced convenience and potentially reduces the risk of dosing errors associated with reconstitution, which can be a significant advantage for patient compliance and preference, particularly in self-administration scenarios.
D. Storage and Stability
Proper storage is crucial to maintain the potency and safety of hCG preparations.
- Unopened Lyophilized Powder (Urinary hCG): Dry powder vials should generally be stored at controlled room temperature, typically 20°C to 25°C (68°F to 77°F), with excursions permitted between 15°C and 30°C (59°F and 86°F).[19] Some sources suggest refrigeration at 2°C to 8°C for the powder.[17] The Canadian monograph for Pregnyl® specifies storage at 15°C - 30°C for the dry powder.[18] It is imperative to follow the specific storage instructions provided on the product labeling.
- Reconstituted Urinary hCG Solution: Once reconstituted, urinary hCG solutions must be refrigerated at 2°C to 8°C (36°F to 46°F). The stability period after reconstitution varies depending on the brand and the specific diluent used (particularly if it contains a preservative like benzyl alcohol):
- Novarel® (reconstituted with Bacteriostatic Water for Injection preserved with 0.9% benzyl alcohol): Stable for up to 30 days when refrigerated.[19]
- Pregnyl® (reconstituted with Bacteriostatic Water for Injection preserved with 0.9% benzyl alcohol): Stability is reported as up to 60 days when refrigerated by some US sources.[3] However, the Canadian Pregnyl® monograph states stability for 28 days when refrigerated.[18] This variability underscores the critical importance of adhering to the product-specific labeling for reconstituted stability, as incorrect storage can lead to loss of potency or microbial contamination, particularly with multi-dose vials.
- Generic hCG solutions (reconstituted): Storage and stability will depend on the specific manufacturer's instructions, typically ranging from 30 to 60 days under refrigeration.[57]
- Recombinant hCG (e.g., Ovidrel®): Pre-filled syringes of Ovidrel® should be stored protected from light and refrigerated at 2°C to 8°C. Prior to use, they may be stored at room temperature (up to 25°C or 77°F) for no more than 30 days, but should not be re-refrigerated once brought to room temperature. Specific product labeling should always be consulted.[38]
- General Precautions: Reconstituted solutions should not be frozen. Any solution that is discolored or contains particulate matter should not be used and should be discarded.[7]
Table 3: Approved Indications and Recommended Dosage Regimens for Chorionic Gonadotropin (Human) (Urinary-Derived, e.g., Pregnyl®, Novarel®)
Indication | Typical Patient Population | Dosage Range (IM) | Frequency & Duration | Key Clinical Notes/Sources |
---|
Female Infertility - Ovulation Induction/ART (after FSH/hMG pretreatment) | Anovulatory, infertile women (secondary anovulation, not primary ovarian failure) | 5,000 - 10,000 USP units | Single dose, 1 day after last FSH/hMG dose | 10,000 USP units often recommended. Requires careful ovarian monitoring. 3 |
Male Hypogonadotropic Hypogonadism - Virilization | Males with hypogonadism secondary to pituitary deficiency | 500 - 1,000 USP units | 3x/week for 3 weeks, then 2x/week for 3 weeks | Alternative: 4,000 USP units 3x/week for 6-9 months, then 2,000 USP units 3x/week for 3 months. 3 |
Male Hypogonadotropic Hypogonadism - Spermatogenesis Induction | Males with hypogonadism secondary to pituitary deficiency, desiring fertility | 1,000 - 2,000 USP units | 2-3x/week, often for 3-6 months or longer | Typically used in combination with FSH/hMG. 33 |
Prepubertal Cryptorchidism (not due to anatomical obstruction) | Boys aged 4-9 years | 1. 4,000 USP units <br> 2. 5,000 USP units <br> 3. 500 - 1,000 USP units <br> 4. 500 USP units (then 1,000 USP units if needed) | 1. 3x/week for 3 weeks <br> 2. Every other day for 4 injections <br> 3. Total 15 injections over 6 weeks <br> 4. 3x/week for 4-6 weeks; repeat course after 1 month if needed. | Efficacy controversial; guidelines often favor orchiopexy. May be temporary descent. 3 |
VII. Safety Profile and Risk Management
The use of Chorionic Gonadotropin (Human) is associated with a range of potential adverse effects and requires careful patient selection and monitoring to mitigate risks.
A. Contraindications
The administration of hCG is contraindicated in individuals with [5]:
- Known prior hypersensitivity to hCG or any of its excipients (e.g., benzyl alcohol in some diluents).
- Precocious puberty, or in children being treated for cryptorchidism if signs of precocious puberty develop (Novarel® has this as an explicit contraindication).
- Presence of androgen-dependent tumors, such as prostatic carcinoma in males, or tumors of the ovary, breast, or uterus in females. Also contraindicated in tumors of the hypothalamus or pituitary gland.
- In females being treated for infertility: high serum FSH levels indicating primary ovarian failure, uncontrolled non-gonadal endocrinopathies (e.g., thyroid, adrenal, or pituitary disorders), abnormal vaginal bleeding of undetermined origin, malformations of the reproductive organs incompatible with pregnancy, or fibroid tumors of the uterus incompatible with pregnancy.
- Pregnancy: hCG should not be used if a woman is already pregnant.
- Active thromboembolic disorders (a specific contraindication listed for some recombinant hCG products like Ovidrel® [76]).
B. Warnings and Precautions
Significant warnings and precautions are associated with hCG therapy:
- Ovarian Hyperstimulation Syndrome (OHSS): This is the most serious and potentially life-threatening complication of ovarian stimulation in females, particularly when hCG is used to trigger ovulation after treatment with gonadotropins (FSH/hMG).[5] OHSS is characterized by a spectrum of symptoms, from mild ovarian enlargement and abdominal discomfort to severe forms involving marked ovarian enlargement, ascites, pleural effusion, hemoconcentration, electrolyte imbalances, oliguria, dyspnea, and an increased risk of thromboembolic events. The development of OHSS is a direct consequence of hCG's potent and sustained stimulation of multiple ovarian follicles, leading to the release of vasoactive substances (e.g., VEGF) that increase vascular permeability. The long pharmacokinetic half-life of hCG exacerbates this risk by prolonging ovarian stimulation.
- Risk Factors: Include polycystic ovary syndrome (PCOS), a high number of developing follicles (>20-25), high or rapidly rising serum estradiol levels prior to hCG administration, young age, and a history of OHSS.
- Monitoring and Prevention: Careful monitoring of ovarian response with transvaginal ultrasound (to assess follicular number and size) and serum estradiol levels is crucial to identify patients at high risk. If signs of excessive ovarian response are present (e.g., estradiol >2500-3500 pg/mL, or numerous follicles), hCG administration should be withheld to prevent OHSS. Alternative strategies, such as using a GnRH agonist to trigger ovulation (in GnRH antagonist cycles) or cycle cancellation, may be considered.
- Management: OHSS typically manifests after hCG administration and can worsen if pregnancy occurs. Management of established OHSS is primarily supportive and may require hospitalization for severe cases. It includes bed rest, fluid and electrolyte management, analgesia, and thromboprophylaxis. Diuretics are generally avoided in the acute phase as they can exacerbate intravascular volume depletion.
- Thromboembolic Events: Both arterial and venous thromboembolic events (e.g., deep vein thrombosis, pulmonary embolism, stroke, myocardial infarction) have been reported in association with gonadotropin therapy, including hCG. These events can occur with or without OHSS.[5] Patients with known risk factors for thrombosis (e.g., personal or family history of thrombophilia, severe obesity, prolonged immobilization) should be counseled about this risk and managed with caution. The interconnectedness of OHSS and thromboembolism is significant, as hemoconcentration during severe OHSS is a major predisposing factor. However, the occurrence of thromboembolic events independent of OHSS suggests that hCG itself might have prothrombotic effects or that underlying patient factors play a role.
- Multiple Births: Ovulation induction with gonadotropins followed by hCG significantly increases the risk of multiple pregnancies (e.g., twins, triplets, or higher-order multiples).[7] Multiple gestation is associated with increased risks for both the mother (e.g., preeclampsia, gestational diabetes, postpartum hemorrhage) and the fetuses/neonates (e.g., prematurity, low birth weight, congenital anomalies, perinatal mortality). Patients should be counseled about this risk before starting treatment. Careful monitoring during ovarian stimulation aims to achieve monofollicular or, at most, bifollicular development to minimize this risk in non-ART ovulation induction cycles.
- Anaphylaxis and Hypersensitivity Reactions: Serious systemic hypersensitivity reactions, including anaphylaxis, have been reported with urinary-derived hCG products. These reactions can be life-threatening and require immediate medical attention. Localized reactions at the injection site can also occur.[5]
- Precocious Puberty in Children: In boys treated for cryptorchidism, the androgen secretion induced by hCG can lead to signs of precocious puberty, such as penile and testicular enlargement, pubic hair growth, voice deepening, acne, and rapid linear growth. If these signs appear, hCG therapy should be discontinued.[7] Premature epiphyseal closure is a concern.
- Fluid Retention: hCG-induced androgen production can cause sodium and water retention, leading to edema. Therefore, hCG should be used with caution in patients whose conditions might be aggravated by fluid retention, such as those with pre-existing cardiac or renal disease, epilepsy, migraine, or asthma.[6]
- Ovarian Cysts and Torsion: hCG therapy can lead to the enlargement of pre-existing ovarian cysts or the development of new ones. Rupture of ovarian cysts with resultant hemoperitoneum has occurred. Ovarian torsion, a surgical emergency, has also been reported following gonadotropin treatment and may be related to OHSS, ovarian enlargement, pregnancy, or prior abdominal surgery.[6]
- Ectopic Pregnancy: Women undergoing ART have a slightly increased risk of ectopic pregnancy. Early ultrasound confirmation of intrauterine pregnancy is important.[14]
- Spontaneous Abortion and Congenital Abnormalities: The incidence of spontaneous abortion (miscarriage) and congenital abnormalities may be slightly higher in pregnancies conceived following gonadotropin therapy and ART compared to spontaneously conceived pregnancies. However, this may be related to underlying parental factors (e.g., maternal age, sperm quality, genetic factors) or the ART procedures themselves, rather than a direct teratogenic effect of hCG.[14] Animal studies with combined gonadotropin and hCG at doses to induce superovulation have reported defects in offspring.[21]
- Tumorigenicity: There have been sporadic reports of testicular tumors in young men receiving hCG for secondary infertility; however, a causal relationship has not been definitively established.[22] Some studies suggest a possible increased risk of ovarian cancer with multiple cycles of fertility treatments, but this remains an area of ongoing research and debate.[56]
- Benzyl Alcohol in Diluent: Some urinary hCG formulations are supplied with a diluent containing benzyl alcohol as a preservative. Large amounts of benzyl alcohol (≥99 mg/kg/day) have been associated with a potentially fatal "gasping syndrome" in neonates and premature infants, characterized by metabolic acidosis, respiratory distress, gasping respirations, CNS dysfunction, hypotension, and cardiovascular collapse. Therefore, formulations containing benzyl alcohol should be used with caution, or preferably avoided, in this population.[3]
- Obesity: hCG is not effective as an adjunctive therapy in the treatment of obesity and should not be used for this purpose.[5]
C. Adverse Effects
The adverse effects of hCG can be categorized as common or serious [6]:
Common Adverse Effects:
- Local: Pain, bruising, redness, swelling, irritation, or inflammation at the injection site.
- Systemic: Headache, irritability, restlessness, fatigue, depression, changes in emotions or mood.
- Fluid Balance: Edema, mild swelling, or water weight gain.
- Hormonal (Males): Gynecomastia (breast enlargement).
- Hormonal (Females, due to androgenic effects or ovarian stimulation): Acne, facial hair growth, mild abdominal or pelvic pain/bloating, nausea, vomiting, upset stomach, diarrhea (these gastrointestinal symptoms can also be early warning signs of OHSS).
- Hormonal (Children): Early signs of puberty if treated for cryptorchidism (see Warnings).
Serious Adverse Effects (requiring immediate medical attention):
- Ovarian Hyperstimulation Syndrome (OHSS): Severe pelvic or abdominal pain, severe nausea/vomiting, rapid weight gain, significant abdominal distension, shortness of breath, oliguria (decreased urination), diarrhea.
- Thromboembolic Events: Signs include pain, warmth, redness, numbness, or tingling in an arm or leg; confusion; extreme dizziness; severe headache; sudden shortness of breath; chest pain.
- Allergic Reactions/Anaphylaxis: Hives, skin rash, itching, angioedema (swelling of the face, lips, tongue, or throat), difficulty breathing, wheezing, chest tightness.
- Precocious Puberty (in boys): As listed under Warnings.
- Rupture of Ovarian Cysts / Hemoperitoneum.
- Serious Pulmonary Conditions: Atelectasis, acute respiratory distress syndrome (ARDS).
- Neurological (potential signs of stroke): Weakness on one side of the body, slurred speech, sudden vision changes, sudden severe headache.
D. Overdose Information
Acute overdose with hCG is not expected to produce life-threatening symptoms.7 However, chronic overdosage or administration of excessively high doses could lead to an exaggeration of its pharmacological effects, most notably severe OHSS in females or pronounced signs of androgen excess (e.g., severe acne, priapism, aggressive behavior) in males.
Management of suspected overdose involves discontinuing the drug and providing symptomatic and supportive care. In cases of accidental ingestion or significant overdosage, contacting a poison control center is advisable.69
E. Drug Interactions
Clinically significant drug interactions with hCG primarily involve pharmacodynamic interactions or interference with laboratory tests.
1. Drug-Drug Interactions:
- Gonadotropin-Releasing Hormone (GnRH) Analogs (Agonists and Antagonists): hCG is often used sequentially with GnRH agonists (e.g., leuprolide) or GnRH antagonists (e.g., ganirelix, cetrorelix) in ART protocols. These GnRH analogs are used to control the pituitary-ovarian axis and prevent premature LH surges. While not a direct interaction with hCG's mechanism, their use dictates the timing and necessity of hCG administration. Some data suggest that ganirelix might decrease the effects of hCG, potentially requiring dose adjustments or special monitoring if used concurrently, though this is typically a sequential administration.[80] The interaction is more about the coordinated regulation of the reproductive cycle.
- Androgen-Dependent Neoplasms: The use of hCG is contraindicated in patients with androgen-dependent neoplasms (e.g., prostate cancer) because hCG stimulates endogenous androgen production, which could exacerbate these conditions.[78]
- Drugs Affecting Fluid Balance: Caution is advised when hCG is used concomitantly with other drugs that can cause fluid retention, as hCG itself can induce edema due to androgenic effects.[78]
- Lack of Extensive Studies: For many urinary-derived hCG products like Pregnyl®, product monographs may state that formal drug interaction studies have not been conducted, and therefore, interactions with other commonly used medicinal products cannot be entirely excluded.[18] Patients should always inform their healthcare provider of all medications, including over-the-counter drugs, vitamins, and herbal supplements they are taking.[7]
- Questionable Interactions: Some sources [58] list interactions with hormonal contraceptives, cyclosporine, rifampin, and warfarin. These are not typically recognized interactions for hCG and should be viewed with extreme caution unless corroborated by authoritative pharmacological sources. The risk of thromboembolism with hCG therapy itself means that if a patient is on anticoagulants like warfarin, their anticoagulation status would need very careful monitoring, but this is a management issue related to an adverse effect of hCG, not a direct drug-drug interaction altering hCG's primary action.
2. Drug-Laboratory Test Interactions:
- Interference with Gonadotropin Immunoassays: Due to the identical α-subunit shared by hCG, LH, FSH, and TSH, and the significant homology between the β-subunits of hCG and LH, exogenous hCG administration can interfere with immunoassays used to measure endogenous gonadotropin levels, particularly LH. This can lead to falsely elevated or misinterpreted LH (and sometimes FSH) results.[21] It is crucial for physicians to inform the laboratory if a patient is receiving hCG therapy when gonadotropin levels are requested, and laboratories should be aware of the potential for cross-reactivity with their specific assays.
- False Positive Pregnancy Tests: Administered exogenous hCG is detected by both serum and urine pregnancy tests. Therefore, pregnancy tests performed too soon after hCG administration (up to 10-14 days, depending on the dose and individual clearance) can yield a false positive result, as the test will detect the exogenous hormone rather than hCG produced by an actual pregnancy.[18]
Table 4: Clinically Significant Drug Interactions and Laboratory Test Interferences for Chorionic Gonadotropin (Human)
Interacting Agent/Test | Nature of Interaction | Clinical Significance/Management | Key Sources |
---|
Drug-Drug Interactions | | | |
Gonadorelin / GnRH Antagonists (e.g., Ganirelix) | Potential decreased effect of hCG if used concurrently (though typically sequential). Pharmacodynamic interaction related to ovarian stimulation protocols. | Usually administered sequentially in ART. If concurrent use is considered, dose adjustment or special testing may be needed. Monitor ovarian response. | 34 |
Androgen-dependent tumor therapies | hCG stimulates androgen production. | Contraindicated in patients with prostatic or other androgen-dependent neoplasms. | 78 |
Drugs causing fluid retention | Additive effect on fluid retention. | Use with caution in patients with cardiac/renal disease, epilepsy, migraine, asthma. Monitor for edema. | 78 |
Drug-Laboratory Test Interactions | | | |
Immunoassays for LH (and potentially FSH) | Cross-reactivity due to shared α-subunit and β-subunit homology (for LH). | Can cause falsely elevated or misinterpreted LH/FSH levels. Inform laboratory if patient is on hCG. | 21 |
Pregnancy Tests (Serum/Urine) | Exogenous hCG is detected by the test. | Can cause false positive pregnancy results for up to 10-14 days after last hCG dose. Delay testing or use serial quantitative hCG to confirm endogenous production. | 18 |
VIII. Regulatory Status
The regulatory status of Chorionic Gonadotropin (Human) varies by region and specific formulation (urinary-derived vs. recombinant).
A. FDA Approval Status
Urinary-derived Chorionic Gonadotropin (Human) (DB09126), available under brand names such as Pregnyl® and Novarel®, is approved by the U.S. Food and Drug Administration (FDA).[3] Its approved indications include:
- Prepubertal cryptorchidism not due to anatomical obstruction.
- Selected cases of hypogonadotropic hypogonadism in males.
- Induction of ovulation and pregnancy in the anovulatory, infertile woman in whom the cause of anovulation is secondary and not due to primary ovarian failure, and who has been appropriately pretreated with human menotropins.[4]
Recombinant choriogonadotropin alfa (e.g., Ovidrel®, DB00097) is also FDA-approved for female infertility indications.13
The FDA explicitly states that hCG has not been demonstrated to be effective adjunctive therapy in the treatment of obesity, and there is no substantial evidence that it increases weight loss beyond that resulting from caloric restriction, causes a more attractive or "normal" distribution of fat, or decreases hunger associated with calorie-restricted diets.5
B. EMA Approval Status
Choriogonadotropin alfa (recombinant hCG), under brand names like Ovitrelle®, is authorized for use in the European Union by the European Medicines Agency (EMA).13 Ovitrelle® is indicated to trigger final follicular maturation and luteinization in women undergoing superovulation prior to ART, and for ovulation induction in anovulatory or oligo-ovulatory women after stimulation of follicular development.76
The EMA has also authorized Ngenla® (somatrogon), a long-acting human growth hormone modified by combining it with part of human chorionic gonadotropin, for growth hormone deficiency. The hCG component in Ngenla® serves to extend its half-life but does not exert gonadotropic effects itself.82
Information on the specific EMA approval status of urinary-derived hCG products (analogous to DB09126) requires searching the EMA database or national registers, as they might be authorized via national procedures rather than centrally through the EMA.83
C. Other Regulatory Bodies (Brief Mention if data available)
Urinary-derived hCG products like Pregnyl® are also approved in other regions, such as Canada.[18] The specific indications and approved formulations may vary by country.
D. Controlled Substance Classification
Chorionic Gonadotropin (Human) is not classified as a controlled substance under the U.S. Controlled Substances Act (CSA).[3] However, it is listed by the World Anti-Doping Agency (WADA) as a prohibited substance for athletes, as it can be misused to increase testosterone production.[3]
IX. Clinical Trials and Recent Developments
The clinical development and ongoing research for Chorionic Gonadotropin (Human), both urinary-derived (DB09126) and recombinant forms, span several decades. Numerous clinical trials have evaluated its efficacy and safety across its primary indications.
A. Overview of Clinical Trial Landscape for DB09126
DrugBank records indicate that Chorionic Gonadotropin (Human) (DB09126) has been involved in a substantial number of clinical trials across various phases: 4 Phase 0, 13 Phase 1, 26 Phase 2, 42 Phase 3, and 76 Phase 4 trials.[4] These trials cover its indicated conditions, such as female infertility (ovarian response, ovulation induction) and male hypogonadism.[4] For example, completed Phase 4 trials include studies on hCG priming for poor responders in IVF (NCT00870025) and biosimilarity studies for female infertility (NCT04605107).[84]
B. Efficacy and Safety in Approved Indications: Key Trial Data
1. Female Infertility (Ovulation Induction/ART):
hCG is a cornerstone in ART for triggering final oocyte maturation.
- Mechanism: It mimics the natural LH surge, leading to granulosa cell luteinization, resumption of oocyte meiosis, and follicular rupture approximately 36-40 hours later.[24]
- Efficacy: Studies have consistently shown its effectiveness in achieving oocyte maturation and enabling successful fertilization and pregnancy when used appropriately after ovarian stimulation with FSH/hMG.[9] Clinical pregnancy rates vary depending on patient population and ART protocol but are generally considered comparable between urinary and recombinant hCG forms when equivalent biological doses are used.[9]
- Safety: The primary safety concern is OHSS, which can be severe. The long half-life of hCG contributes to this risk by providing sustained luteal stimulation.[5] Other risks include multiple pregnancies and thromboembolic events.[7]
2. Male Hypogonadotropic Hypogonadism (Virilization and Spermatogenesis):
- Mechanism: hCG stimulates Leydig cells to produce testosterone, leading to virilization. For spermatogenesis, concurrent FSH activity (either endogenous if partially deficient or exogenous via hMG/rFSH) is usually required to support Sertoli cell function.[39]
- Efficacy:
- Testosterone Production: hCG effectively increases serum testosterone levels.[48] A 2022 study (retrospective analysis of 28 men) showed that hCG monotherapy was effective in maintaining testosterone levels in men with previous exogenous testosterone use and led to a significant decrease in hematocrit.[61]
- Spermatogenesis: The time to achieve spermatogenesis with gonadotropin therapy (hCG often combined with FSH/hMG) varies widely, typically ranging from 3 to 24 months.[39] Success rates are higher with combined hCG+FSH therapy (around 86%) compared to hCG monotherapy (around 40-50%).[41] Factors like initial testicular volume and history of cryptorchidism can influence outcomes.[39] A 2024 review by Alexander et al. on gonadotropin therapy for mini-puberty induction in male infants with CHH reported that gonadotropins induced statistically significant increases in penile length and inhibin B, and led to testicular descent in 73% of patients, though data heterogeneity was a limitation.[86] A 2024 systematic review and meta-analysis by Alexander et al. on gonadotropins for pubertal induction in males with HH found significant increases in testicular volume, penile size, and testosterone, with higher spermatogenesis rates with hCG + FSH.[41]
- Safety: Adverse effects can include gynecomastia and acne due to increased androgen/estrogen levels. Precocious puberty is a risk if used in prepubertal boys outside of cryptorchidism protocols.[21]
3. Prepubertal Cryptorchidism:
- Efficacy: As discussed in Section V.A.3, the efficacy of hCG for inducing testicular descent is highly controversial. While it is an approved indication and some older studies or case series reported success [62], robust meta-analyses (e.g., Wei et al., 2018) found hCG to be no more effective than placebo, with success rates ranging from 0% to 50.8% for unilateral and 0% to 22.4% for bilateral cryptorchidism.[66]
- Current Recommendations: Most current pediatric urology and endocrinology guidelines (Nordic, EAU, AUA) do not recommend hormonal treatment (hCG or GnRH) for cryptorchidism, favoring early orchiopexy (surgical correction) as the standard of care, ideally between 6 and 12 (or at latest 18) months of age.[65]
- Assessment Timeline: If hCG is used, response (testicular descent) is typically assessed at the end of the treatment course and sometimes a few months later to check for permanency.[66] However, given the low efficacy, prolonged follow-up for descent alone post-hCG is less common if surgery is planned.
C. Comparative Efficacy: Urinary-Derived hCG (DB09126) vs. Recombinant hCG (DB00097)
1. Purity, Immunogenicity, and Consistency:
- Urinary-Derived hCG (uHCG): Extracted from the urine of pregnant women, uHCG preparations inherently contain a mixture of hCG isoforms and other urinary proteins, even after purification.[3] This can lead to batch-to-batch variability in specific activity and a higher potential for immunogenic reactions or allergic responses due to protein contaminants.[9] Prolonged treatment with urinary gonadotropins (including hCG) may lead to the development of anti-hCG antibodies, which could potentially reduce treatment efficacy over time, particularly in male hypogonadism.[41]
- Recombinant hCG (rHCG; choriogonadotropin alfa): Produced using recombinant DNA technology in cell lines (e.g., CHO cells), rHCG offers a highly purified product with a consistent isoform profile and specific activity.[8] This higher purity generally leads to a better safety profile regarding local injection site reactions and potentially lower immunogenicity.[8] rHCG allows for precise dosing by mass (micrograms) rather than biological units (IU).[9]
2. Clinical Outcomes (Live Birth Rate, OHSS, etc.):
Numerous clinical trials and meta-analyses have compared uHCG and rHCG for ovulation induction in ART.
- Live Birth Rate / Ongoing Pregnancy Rate: Most systematic reviews and meta-analyses, including Cochrane reviews (e.g., Youssef et al., 2016, which incorporated earlier versions), conclude that there is no significant difference in live birth rates or ongoing pregnancy rates between rHCG and uHCG when used for final oocyte maturation in IVF/ICSI cycles.[10] A 2022 review by Alsamarai et al. suggested purified urinary hCG might lead to higher pregnancy rates in IVF-ICSI compared to rHCG, attributing this to potentially more favorable progesterone profiles with uHCG, though this contrasts with the broader consensus of no difference from larger meta-analyses.[55]
- Ovarian Hyperstimulation Syndrome (OHSS) Rate: Similarly, the incidence of OHSS (all severities) is generally found to be comparable between rHCG and uHCG groups in large meta-analyses.[10]
- Oocyte Quality and Number: The number of oocytes retrieved and oocyte maturation rates are also generally similar between the two forms.[8] Some individual studies or smaller analyses might show slight variations, but overall, major differences are not consistently reported.
- Injection Site Reactions: rHCG, due to its higher purity, is often associated with a lower incidence of local injection site reactions compared to uHCG.[10]
- Male Hypogonadism: While rHCG is considered safer and of better quality [41], direct large-scale comparative trials of uHCG vs. rHCG specifically for long-term spermatogenesis induction or virilization in males are less abundant than in female ART. EAU guidelines state comparable effects but note "No data in men" for rHCG standard dosage in their tables.[43] A 2002 study by Handelsman et al. using r-hCG in older men with partial androgen deficiency showed hormonal and body composition effects, noting previous reports on urinary hCG were for different populations.[49]
3. Cost-Effectiveness Considerations:
Urinary-derived hCG preparations are generally less expensive than recombinant hCG products.9 Given the similar efficacy for key outcomes like live birth rates in ART, the lower cost of uHCG can be a significant factor in its selection, particularly in resource-limited settings or where healthcare costs are a major consideration for patients.9 However, the potential for fewer local reactions and the convenience of pre-filled syringes with rHCG might be valued by some patients and clinicians despite the higher cost.
4. Current Clinical Practice Guidelines and Expert Opinions (ASRM, EAU, Endocrine Society):
- ASRM (American Society for Reproductive Medicine): A 2020 ASRM committee opinion stated that gonadotropin products from urinary extracts or recombinant technology have similar effectiveness and safety for ovulation induction. A systematic review found no difference in live birth or OHSS rates between urinary and recombinant gonadotropins in women with PCOS.[11] For triggering ovulation, 5,000-10,000 IU of urinary hCG or 250 mcg of recombinant hCG are considered options.[11] Aetna's 2023/2024/2025 policy and CarelonRx's 2025 criteria, referencing ASRM, do not differentiate preference for ART based on efficacy or safety between urinary and recombinant hCG, listing both as medically necessary.[51]
- EAU (European Association of Urology): The 2023-2025 EAU guidelines on Sexual and Reproductive Health state that recombinant hCG and LH formulations offer comparable effects to urinary-derived preparations for male hypogonadism when fertility is desired. They recommend hCG administration with FSH for better outcomes. While noting the lower cost of extractive (urinary) hCG, the table lists "No data in men" for standard dosage of recombinant hCG, suggesting less established specific dosing for r-hCG in this context within their guideline summary.[43]
- General Consensus: The overall consensus from major guidelines and systematic reviews is that while rHCG offers advantages in purity, consistency, and potentially fewer local reactions, its efficacy in terms of major clinical outcomes like live birth rates and OHSS rates in ART is largely comparable to that of uHCG. The choice often comes down to cost, availability, patient/physician preference, and specific product characteristics (e.g., route of administration, pre-filled syringes).
D. Recent Advances and Future Directions (Post-2022 Research)
1. Novel Signaling Pathway Insights (LHCGR):
Research continues to unravel the complexities of LHCGR signaling.
- Biased Agonism: Recent reviews (2023, 2024) emphasize the concept of biased agonism, where LH and hCG, despite binding to the same receptor, can differentially activate downstream pathways (Gs, Gq/11, Gi, β-arrestins), influenced by hormone glycosylation and receptor dimerization (e.g., with FSHR).[26] hCG tends to be a stronger activator of cAMP and steroidogenesis, while LH may favor proliferative/anti-apoptotic signals via AKT/ERK.[26] These differences have clinical relevance, for example, in the context of dual triggers in ART (GnRH agonist + low-dose hCG) to balance oocyte maturation quality with luteal support and OHSS risk reduction.[26]
- Non-canonical Pathways and Endosomal Signaling: The role of β-arrestins in scaffolding non-G protein-mediated signaling (e.g., MAPK activation) and the importance of receptor internalization and signaling from endosomal compartments (involving proteins like APPL1) are areas of active investigation.[27] These pathways contribute to the duration and diversity of cellular responses to hCG/LH.
- Gene Regulation: FSH-induced Lhcgr gene expression in granulosa cells involves PKA and PI3K/AKT pathways, with β-catenin and SF1 playing roles at the promoter level.[45] Understanding these regulatory mechanisms is crucial for optimizing ovarian response.
- Anti-apoptotic Signaling: Both LH and hCG can exert anti-apoptotic effects in gonadal cells, often mediated via the PI3K/AKT pathway.[27] However, at very high concentrations or in specific contexts, gonadotropins might also induce pro-apoptotic signals.[27]
2. New Therapeutic Targets or Formulations:
- Low-Molecular-Weight Allosteric Modulators of LHCGR: Development of orally active, small molecule allosteric agonists or modulators of LHCGR (e.g., thienopyrimidines) is a promising area. These could offer more convenient administration, potentially different signaling profiles, and reduced risk of issues like OHSS or antibody formation compared to injectable protein hormones.[28]
- Long-Acting Gonadotropins: While not directly hCG, the development of long-acting FSH analogs (e.g., corifollitropin alfa) [42] reflects a trend towards reducing injection burden in fertility treatments. Similar principles could theoretically be applied to LH/hCG analogs.
- Biosimilars: The emergence of biosimilar gonadotropins, including hCG, is increasing, potentially offering more cost-effective treatment options.[85]
3. Updates on Long-Term Safety or Efficacy:
- Male Hypogonadism: A 2024 review by Yilmaz et al. on physiological pubertal induction in males with HH highlighted that consistent evidence suggests rFSH/hCG combination therapy is more effective than hCG alone for spermatogenesis and testicular volume. It also noted that while urinary and recombinant hCG show no efficacy difference, rHCG is purer and has a better safety profile regarding immunogenicity.[42] The median time to achieve sperm in ejaculate with gonadotropin therapy was reported as 7.1 months in one meta-analysis cited.[42]
- Resistant Ovary Syndrome: A 2025 review on ROS concluded that both recombinant and urinary gonadotropins may be effective in COS for these patients, and both r-hCG and u-hCG have been proven effective as triggers if dominant follicles are observed.[53]
- Recurrent Pregnancy Loss (RPL): A 2024 review on immune mechanisms in RPL mentions that urinary chorionic gonadotropin is used for pregnancy confirmation in RPL studies, but does not focus on its therapeutic use in this context.[94] hCG's immunomodulatory roles are known, but its direct therapeutic application in RPL is still being explored with limited consensus.[32]
- No major new long-term safety signals for established urinary hCG products have emerged in the very recent (2023-2025) snippets beyond well-known risks like OHSS. The focus remains on optimizing protocols and patient selection.
X. Conclusion
A. Summary of Key Findings
Chorionic Gonadotropin (Human) (DB09126), a urinary-derived glycoprotein hormone, plays a significant, albeit specific, role in reproductive medicine and endocrinology. Its primary mechanism involves mimicking endogenous LH by activating the LHCGR, leading to stimulation of gonadal steroidogenesis and gamete maturation. Key findings from this comprehensive review include:
- Structure and Function: hCG is a heterodimer with a common α-subunit and a unique, heavily glycosylated β-subunit containing a C-terminal peptide responsible for its extended half-life. This structure underpins its sustained biological activity and differentiates it from LH.
- Signaling Complexity: Activation of LHCGR engages multiple intracellular pathways (cAMP/PKA, PLC/Ca2+/PKC, MAPK/ERK, PI3K/AKT, β-arrestin), with evidence of biased agonism between hCG and LH, potentially leading to different physiological outcomes.
- Pharmacokinetics: Urinary hCG has a terminal half-life of approximately 23-37 hours, with IM administration leading to peak levels within 6 hours. About 10-20% is excreted unchanged in urine.
- Therapeutic Uses: Approved indications include female infertility (ovulation induction/ART), male hypogonadotropic hypogonadism, and prepubertal cryptorchidism. Its use in cryptorchidism is highly controversial, with current guidelines favoring surgical correction due to low efficacy of hormonal treatment. Off-label use for weight loss is unsubstantiated and not recommended. Diagnostic uses include the hCG stimulation test and as a tumor marker.
- Safety Profile: The most significant risks are OHSS in females, thromboembolic events, and multiple pregnancies. Precocious puberty can occur in children. Contraindications primarily relate to hormone-sensitive tumors and primary gonadal failure.
- Urinary vs. Recombinant hCG: Recombinant hCG offers higher purity and consistency, potentially lower immunogenicity, and more convenient SC administration (often in pre-filled syringes). However, for key clinical outcomes in ART (live birth, OHSS rates), large meta-analyses generally show no significant difference in efficacy compared to urinary hCG. Cost is often lower for urinary preparations.
B. Overall Clinical Utility and Limitations of Chorionic Gonadotropin (Human) (DB09126)
Urinary-derived Chorionic Gonadotropin (Human) remains a clinically useful agent, particularly in settings where cost is a significant consideration and its established efficacy in ovulation induction and male hypogonadism treatment is valued. Its long history of use provides extensive clinical experience.
However, its limitations are notable:
- Variability and Purity: As a biological extract, it has inherent variability and potential for impurities compared to recombinant products, which may influence immunogenicity and consistency of effect.
- Risk of OHSS: The potent and sustained action, while beneficial for triggering ovulation, is a primary contributor to the risk of OHSS, requiring careful patient selection and monitoring.
- Controversial Efficacy in Cryptorchidism: Its continued approval for cryptorchidism contrasts with current evidence-based guidelines that largely dismiss its therapeutic value for this indication.
- Administration Route: IM administration is less convenient than SC options available with recombinant forms.
The development of recombinant hCG has addressed some of these limitations (purity, convenience), but the fundamental biological actions and associated risks like OHSS remain similar due to the shared mechanism of action via the LHCGR. The choice between urinary and recombinant hCG often involves a balance of cost, convenience, physician/patient preference, and specific clinical context, as major efficacy differences are not consistently demonstrated for primary ART outcomes.
C. Future Research Perspectives
Future research in the field of hCG and LHCGR signaling should continue to explore:
- Elucidation of Biased Agonism: Further understanding the molecular determinants of biased agonism at the LHCGR and how different signaling profiles translate to specific physiological effects could lead to the design of more targeted gonadotropin therapies with improved efficacy/safety ratios.
- Low-Molecular-Weight LHCGR Modulators: Continued development and clinical testing of oral, small-molecule allosteric modulators of LHCGR could revolutionize gonadotropin therapy by offering non-injectable options with potentially tailored signaling and reduced side effects.
- Extragonadal LHCGR Functions: Investigating the physiological significance and therapeutic potential of LHCGR expression in extragonadal tissues may uncover novel applications for hCG or related compounds.
- Optimizing Protocols for Male Hypogonadism: Refining gonadotropin regimens (hCG with or without FSH) for male hypogonadism, particularly for physiological pubertal induction and optimizing long-term fertility outcomes, requires further well-designed, standardized clinical trials. This includes clarifying the optimal timing, dosage, and duration of therapy, and comparing urinary versus recombinant preparations in this specific population.
- Personalized Medicine Approaches: Identifying biomarkers or genetic factors (e.g., LHCGR polymorphisms) that predict patient response or risk of adverse effects (like OHSS) to hCG therapy could enable more personalized treatment strategies.
- Long-Term Outcomes in Cryptorchidism: While current evidence disfavors hCG for cryptorchidism, long-term follow-up from any well-conducted trials assessing its impact on fertility or testicular cancer risk, if used, would be valuable, though ethical considerations for such trials are now paramount given the preference for surgery.
In conclusion, Chorionic Gonadotropin (Human) is a hormone with a complex profile and enduring, though evolving, therapeutic importance. Continued research is essential to optimize its use and develop novel approaches to hormonal therapy in reproductive and endocrine disorders.
Works cited
- Human chorionic gonadotropin - Wikipedia, accessed June 4, 2025, https://en.wikipedia.org/wiki/Human_chorionic_gonadotropin
- Human Chorionic Gonadotropin - StatPearls - NCBI Bookshelf, accessed June 4, 2025, https://www.ncbi.nlm.nih.gov/books/NBK532950/
- Chorionic Gonadotropin: Package Insert / Prescribing Info - Drugs.com, accessed June 4, 2025, https://www.drugs.com/pro/chorionic-gonadotropin.html
- Chorionic Gonadotropin (Human): Uses, Interactions, Mechanism of Action - DrugBank, accessed June 4, 2025, https://go.drugbank.com/drugs/DB09126
- Pregnyl® (chorionic gonadotropin) for injection - accessdata.fda.gov, accessed June 4, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/017692s028s036s043lbl.pdf
- HCG Injection (Human Chorionic Gonadotropin): Uses & Side Effects - Cleveland Clinic, accessed June 4, 2025, https://my.clevelandclinic.org/health/drugs/18559-human-chorionic-gonadotropin-hcg-injection
- HCG (Injection) Uses, Dosage, Side Effects - Drugs.com, accessed June 4, 2025, https://www.drugs.com/hcg.html
- The efficacy of recombinant versus urinary HCG in ART outcome - ResearchGate, accessed June 4, 2025, https://www.researchgate.net/publication/235768487_The_efficacy_of_recombinant_versus_urinary_HCG_in_ART_outcome
- To Compare the Effectiveness of Recombinant Human Chorionic Gonadotropin (rhCG) with Urinary (u) hCG during Controlled Ovarian H - PAJOG, accessed June 4, 2025, https://pajog.com/images/pajog/issue-image/All_issue/65c1ef08b2f4320240206Article_PDF_FileTo%20Compare%20the%20Effectiveness%20of%20Recombinant%20Human%20Chorionic%20Gonadotropin%20(rhCG).pdf
- Recombinant versus urinary human chorionic gonadotrophin for final oocyte maturation triggering in IVF and ICSI cycles - PMC - PubMed Central, accessed June 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7133782/
- Use of exogenous gonadotropins for ovulation induction in ... - ASRM, accessed June 4, 2025, https://www.asrm.org/practice-guidance/practice-committee-documents/use-of-exogenous-gonadotropins-for-ovulation-induction-in-anovulatory-women-a-committee-opinion-2020/
- Recombinant versus urinary gonadotrophins for triggering ovulation in assisted conception | Human Reproduction | Oxford Academic, accessed June 4, 2025, https://academic.oup.com/humrep/article/20/8/2061/618553
- Choriogonadotropin alfa: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed June 4, 2025, https://go.drugbank.com/drugs/DB00097
- Ovidrel (Choriogonadotropin Alfa Injection): Side Effects, Uses, Dosage, Interactions, Warnings - RxList, accessed June 4, 2025, https://www.rxlist.com/ovidrel-drug.htm
- (PDF) Efficacy, efficiency and effectiveness of gonadotropin therapy for infertility treatment, accessed June 4, 2025, https://www.researchgate.net/publication/279275175_Efficacy_efficiency_and_effectiveness_of_gonadotropin_therapy_for_infertility_treatment
- www.chemicalbook.com, accessed June 4, 2025, https://www.chemicalbook.com/ProductChemicalPropertiesCB8353322_EN.htm#:~:text=Glycoprotein%20hormone%20consisting%20of%20a,corpus%20luteum%20during%20early%20pregnancy.
- Chorionic Gonadotropin | 9002-61-3 - ChemicalBook, accessed June 4, 2025, https://www.chemicalbook.com/ChemicalProductProperty_EN_CB8353322.htm
- www.organon.com, accessed June 4, 2025, https://www.organon.com/canada-en/wp-content/uploads/sites/5/2021/05/PREGNYL-PM_E.pdf
- www.accessdata.fda.gov, accessed June 4, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/017016s156lbl.pdf
- 1HCN: STRUCTURE OF HUMAN CHORIONIC ... - RCSB PDB, accessed June 4, 2025, https://www.rcsb.org/structure/1hcn
- Chorionic Gonadotropin - DailyMed, accessed June 4, 2025, https://dailymed.nlm.nih.gov/dailymed/fda/fdaDrugXsl.cfm?setid=f93b2baa-03ac-4650-b35a-20b818d712d6
- Novarel (Chorionic Gonadotropin for Injection): Side Effects, Uses, Dosage, Interactions, Warnings - RxList, accessed June 4, 2025, https://www.rxlist.com/novarel-drug.htm
- Gonadotropins and Their Analogs: Current and Potential Clinical Applications, accessed June 4, 2025, https://www.researchgate.net/publication/326133705_Gonadotropins_and_Their_Analogs_Current_and_Potential_Clinical_Applications
- Drugs in use - IVF-Worldwide, accessed June 4, 2025, https://ivf-worldwide.com/education/ivf-drug-in-use.html
- Pregnyl (Chorionic Gonadotropin for Injection): Side Effects, Uses, Dosage, Interactions, Warnings - RxList, accessed June 4, 2025, https://www.rxlist.com/pregnyl-drug.htm
- Using GnRH agonists in combination with hCG in antagonist ART ..., accessed June 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10718548/
- Recent advances in understanding gonadotropin signaling - PMC, accessed June 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8130412/
- Hormonal and Allosteric Regulation of the Luteinizing Hormone ..., accessed June 4, 2025, https://www.imrpress.com/journal/FBL/29/9/10.31083/j.fbl2909313/htm
- Chorionic Gonadotropin (Human) | Drug Lookup | Pediatric Care ..., accessed June 4, 2025, https://publications.aap.org/pediatriccare/drug-monograph/18/5628/Chorionic-Gonadotropin-Human
- Luteinizing hormone/choriogonadotropin receptor - Wikipedia, accessed June 4, 2025, https://en.wikipedia.org/wiki/Luteinizing_hormone/choriogonadotropin_receptor
- Differences in Signal Activation by LH and hCG are ... - Frontiers, accessed June 4, 2025, https://www.frontiersin.org/journals/endocrinology/articles/10.3389/fendo.2015.00140/full
- Human Chorionic Gonadotrophin: New Pleiotropic Functions for an “Old” Hormone During Pregnancy - PMC - PubMed Central, accessed June 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7083149/
- Ovidrel, Novarel (choriogonadotropin alfa) dosing, indications, interactions, adverse effects, and more - Medscape Reference, accessed June 4, 2025, https://reference.medscape.com/drug/ovidrel-novarel-choriogonadotropin-alfa-342751
- Chorionic Gonadotropin, Human Intramuscular: Uses, Side Effects, Interactions, Pictures, Warnings & Dosing - WebMD, accessed June 4, 2025, https://www.webmd.com/drugs/2/drug-3582/chorionic-gonadotropin-human-intramuscular/details
- Fertility Injectables | Cigna, accessed June 4, 2025, https://static.cigna.com/assets/chcp/pdf/coveragePolicies/pharmacy/ph_1012_coveragepositioncriteria_infertility_injectables.pdf
- Dual triggering for final oocyte maturation. A narrative review - Frontiers, accessed June 4, 2025, https://www.frontiersin.org/journals/endocrinology/articles/10.3389/fendo.2025.1556732/full
- What is Choriogonadotropin used for? - Patsnap Synapse, accessed June 4, 2025, https://synapse.patsnap.com/article/what-is-choriogonadotropin-used-for
- Chorionic Gonadotropin (Hcg) Dosage Guide + Max Dose ..., accessed June 4, 2025, https://www.drugs.com/dosage/chorionic-gonadotropin-hcg.html
- Fertility treatments for men with hypogonadotropic hypogonadism - ProBiologists, accessed June 4, 2025, https://www.probiologists.com/article/fertility-treatments-for-men-with-hypogonadotropic-hypogonadism
- Recovery of spermatogenesis following testosterone replacement therapy or anabolic-androgenic steroid use - PMC, accessed June 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4854084/
- Is There a Role for Hormonal Therapy in Men with ... - PubMed Central, accessed June 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11721639/
- A Current Perspective on Delayed Puberty and Its Management - PMC, accessed June 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11629716/
- EAU Guidelines on Sexual and Reproductive Health - Uroweb, accessed June 4, 2025, https://uroweb.org/guidelines/sexual-and-reproductive-health/chapter/male-hypogonadism
- Human Chorionic Gonadotropin Stimulates Theca-Interstitial Cell ..., accessed June 4, 2025, https://academic.oup.com/mend/article/24/9/1782/2706154
- FOSL2 regulates gonadotropin-dependent folliculogenesis through feedback amplification of FSH/FSHR signaling | bioRxiv, accessed June 4, 2025, https://www.biorxiv.org/content/10.1101/2025.03.11.642212v2.full-text
- Lhcgr Expression in Granulosa Cells: Roles for PKA-Phosphorylated β-Catenin, TCF3, and FOXO1 | Molecular Endocrinology | Oxford Academic, accessed June 4, 2025, https://academic.oup.com/mend/article/27/8/1295/2615203
- Review: Leptin gene expression in the placenta - Regulation of a ..., accessed June 4, 2025, https://ri.conicet.gov.ar/bitstream/handle/11336/11683/CONICET_Digital_Nro.14774_B.pdf?sequence=3&isAllowed=y
- Pharmacokinetics and pharmacodynamics of recombinant human ..., accessed June 4, 2025, https://pubmed.ncbi.nlm.nih.gov/12470572/
- Double-Blind, Placebo-Controlled, Randomized Clinical Trial of Recombinant Human Chorionic Gonadotropin on Muscle Strength and Physical Function and Activity in Older Men with Partial Age-Related Androgen Deficiency - Oxford Academic, accessed June 4, 2025, https://academic.oup.com/jcem/article/87/7/3125/2846669
- Uses of Pregnyl | Vinmec, accessed June 4, 2025, https://www.vinmec.com/eng/blog/uses-of-pregnyl-en
- www.carelonrx.com, accessed June 4, 2025, https://www.carelonrx.com/ms/pharmacyinformation/Infertility-and-HCG-Agents.pdf
- Infertility - Medical Clinical Policy Bulletins | Aetna, accessed June 4, 2025, https://es.aetna.com/cpb/medical/data/300_399/0327.html
- Infertility management in resistant ovary syndrome: a review - Frontiers, accessed June 4, 2025, https://www.frontiersin.org/journals/endocrinology/articles/10.3389/fendo.2025.1560981/full
- Induction of final follicular maturation and early luteinization in women undergoing ovulation induction for assisted reproduction treatment—recombinant HCG versus urinary HCG - Oxford Academic, accessed June 4, 2025, https://academic.oup.com/humrep/article/15/7/1446/2915299
- Recombinant Human Chorionic Gonadotropin Versus ... - Ovid, accessed June 4, 2025, https://qa-hub2.ovid.com/journals/mtmj/fulltext/10.4103/mj.mj_11_22~recombinant-human-chorionic-gonadotropin-versus-purified
- Chorionic gonadotropin (subcutaneous route, intramuscular route ..., accessed June 4, 2025, https://www.mayoclinic.org/drugs-supplements/chorionic-gonadotropin-subcutaneous-route-intramuscular-route-injection-route/description/drg-20062846
- HCG (Human chorionic gonadotropin): Uses, FAQs, Warnings ..., accessed June 4, 2025, https://www.goodrx.com/hcg/what-is
- Human Chorionic Gonadotropin: Uses, Side Effects, Dosage & Interactions, accessed June 4, 2025, https://www.apollohospitals.com/medicines/human-chorionic-gonadotropin/
- Chorionic gonadotropin (hcg) - brand name list from Drugs.com, accessed June 4, 2025, https://www.drugs.com/ingredient/chorionic-gonadotropin-hcg.html
- Gonadotropins for pubertal induction in males with hypogonadotropic hypogonadism: systematic review and meta-analysis | European Journal of Endocrinology | Oxford Academic, accessed June 4, 2025, https://academic.oup.com/ejendo/article/190/1/S1/7486652
- The Safety of Human Chorionic Gonadotropin Monotherapy Among ..., accessed June 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9271319/
- Successful treatment of unilateral cryptorchid boys risking infertility with LH-RH analogue, accessed June 4, 2025, https://www.scielo.br/j/ibju/a/f3N6jW56pkLDwHYKHttGCcm/?lang=en
- Harmonisation of Dynamic Endocrine Testing in Paediatrics (HDET-Paeds): an Australasian collaboration - AACB, accessed June 4, 2025, https://aacb.asn.au/common/Uploaded%20files/aacb/SRAC/HEDT-Paeds%20-%20Summary%20of%20literature%20review%20methodology.pdf
- Harmonisation of Dynamic Endocrine Testing in Paediatrics (HDET-Paeds): an Australasian collaboration, accessed June 4, 2025, https://www.aacb.asn.au/common/Uploaded%20files/aacb/SRAC/HEDT-Paeds%20-%20Summary%20of%20literature%20review%20methodology.pdf
- Systematic review and meta-analysis comparing outcomes following orchidopexy for cryptorchidism before or after 1 year of age | BJS Open | Oxford Academic, accessed June 4, 2025, https://academic.oup.com/bjsopen/article/2/1/1/6060727
- Efficacy and safety of human chorionic gonadotropin for treatment of ..., accessed June 4, 2025, https://www.researchgate.net/publication/324522532_Efficacy_and_safety_of_human_chorionic_gonadotropin_for_treatment_of_cryptorchidism_A_meta-analysis_of_randomised_controlled_trials_Hormone_treatment_of_cryptorchidism
- Hypogonadism and Cryptorchidism - PMC - PubMed Central, accessed June 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6974459/
- Evaluation of the role of human chorionic gonadotropin therapy in the management of retractile testes in children - ResearchGate, accessed June 4, 2025, https://www.researchgate.net/publication/366343484_Evaluation_of_the_role_of_human_chorionic_gonadotropin_therapy_in_the_management_of_retractile_testes_in_children
- Choriogonadotropin Alfa,Humrec Syringe - Uses, Side Effects, and More - WebMD, accessed June 4, 2025, https://www.webmd.com/drugs/2/drug-20305/choriogonadotropin-alfa-human-recombinant-subcutaneous/details
- Human Chorionic Gonadotropin and Related Peptides: Candidate Anti-Inflammatory Therapy in Early Stages of Sepsis - Frontiers, accessed June 4, 2025, https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2021.714177/full
- Human Chorionic Gonadotropin Stimulation Test in Prepubertal Children with Micropenis Can Accurately Predict Leydig Cell Function in Pubertal or Postpubertal Adolescents - ResearchGate, accessed June 4, 2025, https://www.researchgate.net/publication/281634082_Human_Chorionic_Gonadotropin_Stimulation_Test_in_Prepubertal_Children_with_Micropenis_Can_Accurately_Predict_Leydig_Cell_Function_in_Pubertal_or_Postpubertal_Adolescents
- Blood Sciences hCG STIMULATION TEST – PAEDIATRIC PROTOCOL PF-BSM-CP-12, accessed June 4, 2025, https://qpulse.ipp-uk.com/QPulseDocumentService/Documents.svc/documents/active/attachment?number=PF-BSM-CP-12
- 3 day HCG Stimulation Test - Manchester University NHS Foundation Trust, accessed June 4, 2025, https://mft.nhs.uk/app/uploads/2024/09/3-day-HCG-Stimulation-Test-v8.pdf
- accessed January 1, 1970, https://www.sciencedirect.com/topics/medicine-and-dentistry/hcg-stimulation-test
- Human chorionic gonadotropin in pregnancy diagnostics | Request PDF - ResearchGate, accessed June 4, 2025, https://www.researchgate.net/publication/51186334_Human_chorionic_gonadotropin_in_pregnancy_diagnostics
- Ovitrelle | European Medicines Agency (EMA), accessed June 4, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/ovitrelle
- Novarel Side Effects, Uses, Interactions, Pictures, Warnings & Dosing - ScriptSave WellRx, accessed June 4, 2025, https://www.wellrx.com/novarel/drug-information/
- Pregnyl Disease Interactions - Drugs.com, accessed June 4, 2025, https://www.drugs.com/disease-interactions/chorionic-gonadotropin-hcg,pregnyl.html
- Pregnyl Disease Interactions - Drugs.com, accessed June 4, 2025, https://www.drugs.com/disease-interactions/chorionic-gonadotropin-hcg,pregnyl.html?professional=1
- Ganirelix and Pregnyl Interactions Checker - Drugs.com, accessed June 4, 2025, https://www.drugs.com/drug-interactions/ganirelix-with-pregnyl-1159-0-656-321.html
- Chorionic gonadotropin (hcg) Interactions - Drugs.com, accessed June 4, 2025, https://www.drugs.com/drug-interactions/chorionic-gonadotropin-hcg.html
- Ngenla | European Medicines Agency (EMA), accessed June 4, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/ngenla
- Medicines | European Medicines Agency (EMA), accessed June 4, 2025, https://www.ema.europa.eu/en/medicines
- Chorionic Gonadotropin (Human) Completed Phase 4 Trials for Ovarian Response Treatment | DrugBank Online, accessed June 4, 2025, https://go.drugbank.com/drugs/DB09126/clinical_trials?conditions=DBCOND0040629&phase=4&purpose=treatment&status=completed
- Chorionic Gonadotropin (Human) Completed Phase 4 Trials for Female Infertility Health Services Research - DrugBank, accessed June 4, 2025, https://go.drugbank.com/drugs/DB09126/clinical_trials?conditions=DBCOND0031380&phase=4&purpose=health_services_research&status=completed
- Gonadotropin Therapy for Mini-Puberty Induction in Male Infants With Hypogonadotropic Hypogonadism | The Journal of Clinical Endocrinology & Metabolism | Oxford Academic, accessed June 4, 2025, https://academic.oup.com/jcem/article/110/4/e921/7924152
- (PDF) Gonadotropins for pubertal induction in males with hypogonadotropic hypogonadism: systematic review and meta-analysis - ResearchGate, accessed June 4, 2025, https://www.researchgate.net/publication/377119196_Gonadotropins_for_pubertal_induction_in_males_with_hypogonadotropic_hypogonadism_systematic_review_and_meta-analysis
- A Window of Opportunity: The Diagnosis of Gonadotropin Deficiency in the Male Infant 1, accessed June 4, 2025, https://www.researchgate.net/publication/8005847_A_Window_of_Opportunity_The_Diagnosis_of_Gonadotropin_Deficiency_in_the_Male_Infant_1
- Gonadotropin Therapy for Mini-Puberty Induction in Male Infants ..., accessed June 4, 2025, https://academic.oup.com/jcem/article-abstract/110/4/e921/7924152
- Growth and descent of the testes in infants with hypogonadotropic hypogonadism receiving subcutaneous gonadotropin infusion - ResearchGate, accessed June 4, 2025, https://www.researchgate.net/publication/304849394_Growth_and_descent_of_the_testes_in_infants_with_hypogonadotropic_hypogonadism_receiving_subcutaneous_gonadotropin_infusion
- Book 1.indb - Scholarly Publications Leiden University, accessed June 4, 2025, https://scholarlypublications.universiteitleiden.nl/access/item%3A3464238/view
- Full article: G protein-coupled receptor (GPCR) gene variants and human genetic disease, accessed June 4, 2025, https://www.tandfonline.com/doi/full/10.1080/10408363.2023.2286606
- The influence of the pharmaceutical industry on the development of gonadotrophins and ovarian stimulation protocols in assisted reproductive technologies - PMC, accessed June 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC12006903/
- Alloimmune Causes of Recurrent Pregnancy Loss: Cellular Mechanisms and Overview of Therapeutic Approaches - PMC - PubMed Central, accessed June 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11596804/