218949-48-5
Lipodystrophies
Tesamorelin is a synthetic peptide analogue of the endogenous hypothalamic peptide, Growth Hormone Releasing Hormone (GHRH), also known as Growth Hormone Releasing Factor (GRF).[1] It is classified pharmacologically as a GHRH analog and a somatotropin agonist.[1]
Tesamorelin is a polypeptide comprising the full 44 amino acid sequence of human GHRH.[2] Its structure is distinguished by a modification at the N-terminus, where a trans-3-hexenoic acid group is attached.[4] This chemical modification serves a crucial function: it enhances the peptide's stability and confers resistance to enzymatic degradation, particularly cleavage by dipeptidyl aminopeptidase (DPP-IV), compared to the native GHRH molecule.[2] This increased stability results in an improved pharmacokinetic profile, allowing for therapeutic efficacy with subcutaneous administration.[2] The Chemical Abstracts Service (CAS) Registry Number for Tesamorelin is 218949-48-5.[5] Its molecular formula is C221H366N72O67S, corresponding to a molecular weight of approximately 5135.8 g/mol.[5]
The primary, regulatory-approved indication for Tesamorelin is the reduction of excess abdominal fat, specifically targeting visceral adipose tissue (VAT), in adult patients infected with the Human Immunodeficiency Virus (HIV) who have developed lipodystrophy.[1] HIV-associated lipodystrophy is recognized as a complex metabolic syndrome characterized by abnormal body fat distribution, which includes the accumulation of VAT (central adiposity), often accompanied by peripheral fat loss (lipoatrophy), insulin resistance, and dyslipidemia (hypertriglyceridemia and altered cholesterol levels).[1] This condition is frequently associated with the use of certain antiretroviral therapies (ART), particularly older regimens, although the HIV virus itself may also contribute.[1]
Tesamorelin was developed by Theratechnologies Inc., a Canadian biopharmaceutical company.[7] It has been marketed under several brand names corresponding to different formulations: Egrifta® (the original F1 formulation), Egrifta SV® (F4 formulation), and most recently, Egrifta WR™ (F8 formulation).[1]
The U.S. Food and Drug Administration (FDA) prescribing information specifies important limitations for Tesamorelin's use.[9] Firstly, the long-term cardiovascular safety profile of Tesamorelin has not been established. This lack of long-term cardiovascular outcome data represents a significant knowledge gap and was a contributing factor to differing regulatory assessments between the US and other regions, such as Europe.[16] Secondly, Tesamorelin is explicitly not indicated for weight loss management; clinical studies have shown it generally has a weight-neutral effect, primarily redistributing fat away from the visceral compartment.[9] Thirdly, there is no evidence to support that Tesamorelin therapy improves patient compliance with their ART regimens.[9] These limitations clearly define the boundaries of the drug's approved application and highlight areas where evidence is either lacking or where the drug is not intended for use, guiding appropriate clinical practice.
Tesamorelin exerts its effects by acting as a specific agonist for the GHRH receptor (GHRH-R).[1] These receptors are primarily located on the surface of somatotroph cells within the anterior pituitary gland.[2]
Upon binding to the GHRH-R, Tesamorelin mimics the physiological action of endogenous GHRH.[17] This interaction triggers intracellular signaling cascades within the somatotrophs, leading to the synthesis and subsequent release of Growth Hormone (GH) into the systemic circulation.[1] Tesamorelin stimulates GH release in a pulsatile manner, similar to the natural pattern of GH secretion, rather than causing continuous release.[17] Due to its N-terminal modification, Tesamorelin is more stable and potentially more potent in stimulating GH release compared to native GHRH.[4]
The increased levels of circulating GH act primarily on the liver, but also on other peripheral tissues, to stimulate the production and secretion of Insulin-like Growth Factor 1 (IGF-1).[1] Consequently, Tesamorelin administration leads to dose-dependent increases in serum concentrations of both GH and IGF-1.[1] Levels of Insulin-like Growth Factor Binding Protein 3 (IGFBP-3), the main carrier protein for IGF-1 in circulation, also increase following Tesamorelin administration.[1]
The physiological effects of Tesamorelin are mediated largely through the actions of GH and IGF-1.
The pharmacokinetic profile of Tesamorelin describes its absorption, distribution, metabolism, and excretion (ADME) following administration.
Tesamorelin is administered via subcutaneous (SC) injection, typically into the abdomen.[2] Following SC injection, its absolute bioavailability is low, estimated to be less than 4% in healthy adult subjects receiving a 2 mg dose.[1] Despite low bioavailability, absorption is rapid, with peak plasma concentrations (Tmax) achieved quickly, approximately 9 to 10 minutes (0.15-0.16 hours) post-dose in both healthy volunteers and HIV-infected patients.[8]
Tesamorelin exhibits a relatively large apparent volume of distribution (Vd). Initial reports indicated a Vd of 9.4 ± 3.1 L/kg in healthy subjects and 10.5 ± 6.1 L/kg in HIV-infected patients.[1] A subsequent population pharmacokinetic analysis, potentially offering a more integrated estimate across studies, calculated the Vd to be approximately 200 L, with an interindividual variability (coefficient of variation, CV) of 17.7%.[19] While the absolute values differ between sources, both suggest extensive distribution beyond the plasma compartment.
As Tesamorelin is a peptide, its primary route of metabolism is expected to involve proteolytic degradation into smaller peptides and constituent amino acids.[2] This breakdown likely occurs both systemically via circulating peptidases and potentially locally at or near receptor sites.[2] Formal metabolism studies specifically evaluating the metabolic pathways and resulting fragments in humans have not been conducted or reported.[1]
Following proteolytic breakdown, the resulting smaller peptides and amino acids are expected to be eliminated primarily via renal clearance.[4] Detailed studies characterizing the specific routes and extent of excretion for Tesamorelin or its metabolites are limited in the available literature.
Tesamorelin has a short elimination half-life (t1/2). Reported values are approximately 26 minutes in healthy subjects and slightly longer, around 38 minutes, in HIV-infected patients.[1] This rapid elimination necessitates daily dosing to maintain sustained pharmacodynamic effects (i.e., stimulation of GH/IGF-1).
A population PK analysis provided further characterization of Tesamorelin's behavior.[19] The data were best described by an open one-compartment model incorporating both first-order and zero-order absorption processes, along with first-order (linear) elimination. Plasma clearance (CL) was estimated to be 1,060 L/h, with substantial interindividual variability (CV 33.6%). An interesting finding was that the fraction of the dose absorbed via the first-order process appeared to increase by approximately 13.1% after 14 days of daily dosing compared to the first day, suggesting a potential time-dependent change in absorption kinetics, although the clinical significance of this is unclear. Importantly, the analysis evaluated the influence of several covariates, including age, body size measures (e.g., weight, BMI), race, and health status (HIV-infected vs. healthy), on the pharmacokinetic parameters (CL and Vd). None of these covariates were found to be statistically significant predictors of Tesamorelin pharmacokinetics within the range studied.[19] The lack of influence from common factors like body weight or BMI on drug exposure simplifies the dosing regimen, supporting the use of a fixed daily dose (e.g., 2 mg for Egrifta SV® or 1.28 mg for Egrifta WR™) across the indicated patient population without the need for weight-based adjustments. Model evaluation procedures, including predictive checks and non-parametric bootstrapping, confirmed the model's appropriateness for describing Tesamorelin concentrations over time in both study populations.[19]
Table 1: Key Pharmacokinetic Parameters of Tesamorelin
Parameter | Value (Mean ± SD or Range) | Population | Source Snippet(s) |
---|---|---|---|
Administration Route | Subcutaneous (SC) Injection | N/A | 2 |
Absolute Bioavailability | < 4% (after 2 mg SC dose) | Healthy Adults | 1 |
Time to Peak (Tmax) | 0.15 - 0.16 hours (~9-10 min) | Healthy & HIV+ | 8 |
Volume of Distribution (Vd) | 9.4 ± 3.1 L/kg | Healthy | 1 |
10.5 ± 6.1 L/kg | HIV+ | 1 | |
~200 L (17.7% CV) - PopPK Estimate | Healthy & HIV+ | 19 | |
Metabolism | Proteolytic degradation; no formal human studies | N/A | 1 |
Elimination Route | Presumed renal clearance after proteolysis | N/A | 4 |
Elimination Half-Life (t1/2) | ~26 minutes | Healthy | 1 |
~38 minutes | HIV+ | 1 | |
Plasma Clearance (CL) | 1,060 L/h (33.6% CV) - PopPK Estimate | Healthy & HIV+ | 19 |
Note: PopPK = Population Pharmacokinetics; CV = Coefficient of Variation. Discrepancy noted in Vd values between sources.
The clinical efficacy of Tesamorelin for reducing excess abdominal fat in HIV-infected individuals with lipodystrophy was established primarily through two large, multicenter, Phase III pivotal trials.[10]
These trials (often referred to as LIPO-010 and CTR-1011, or Trial 10 and Trial 11) shared a similar design.[10] They were randomized, double-blind, placebo-controlled studies conducted in North America and Europe, enrolling a total of 816 HIV-infected adults (aged 18-65) with clinical signs of lipodystrophy (defined by waist circumference and waist-to-hip ratio criteria) and confirmed excess VAT (>130 cm² by CT scan) who were on stable ART for at least 8 weeks.[8] Patients with medically treated diabetes mellitus were excluded.[10] Participants were randomized in a 2:1 ratio to receive either Tesamorelin 2 mg or placebo, self-administered via subcutaneous injection once daily for an initial 26-week main treatment phase.[8] This was followed by a 26-week extension phase. In the extension, participants initially assigned to placebo were switched to Tesamorelin (P-T group), while those initially on Tesamorelin were re-randomized to either continue Tesamorelin (T-T group) or switch to placebo (T-P group).[13]
The primary efficacy endpoint in both trials was the percentage change from baseline in VAT area, measured by a single-slice abdominal computed tomography (CT) scan at the L4-L5 vertebral level, after 26 weeks of treatment.[10]
Both trials met their primary endpoint, demonstrating a statistically significant and clinically meaningful reduction in VAT with Tesamorelin compared to placebo:
Further analysis indicated that the majority of the VAT reduction occurred within the first 13 weeks of treatment, with continued, albeit smaller, reductions observed between weeks 13 and 26.[22]
Based on regulatory consultation and consensus, a reduction in VAT area of ≥8% from baseline was predefined as a clinically significant response.[10] Pooled analyses of the Phase III trials showed that significantly more patients achieved this response threshold with Tesamorelin compared to placebo. Approximately 69% of participants receiving Tesamorelin were classified as VAT responders at 26 weeks, compared to only 33% of those receiving placebo (P < 0.001).[23]
Tesamorelin treatment also resulted in improvements in several secondary endpoints:
Data from the 26-week extension phase provided crucial insights into the durability of the effect. Patients who continued Tesamorelin for the full 52 weeks (T-T group) maintained or showed further modest reductions in VAT.[10] In contrast, patients who were switched from Tesamorelin to placebo at week 26 (T-P group) experienced a rapid re-accumulation of VAT, returning towards their baseline levels by week 52.[10] This finding clearly demonstrates that continuous treatment with Tesamorelin is necessary to sustain the reduction in visceral adiposity.
A key finding from analyses of the trial data is the strong association between the magnitude of VAT reduction and improvements in metabolic parameters.[19] When patients treated with Tesamorelin were categorized as VAT responders (≥8% reduction) or non-responders (<8% reduction), the responders exhibited significantly better metabolic outcomes over 52 weeks. Compared to non-responders, responders experienced significantly greater reductions in triglyceride levels and showed attenuated adverse changes (or even slight improvements) in parameters of glucose homeostasis, including fasting glucose and HbA1c.[19] Responders also had significantly greater increases in adiponectin levels.[19] These associations strongly suggest that the improvements in lipid profiles and the relative preservation of glucose control observed with Tesamorelin are primarily mediated by the reduction in the metabolically active VAT depot itself, rather than being solely direct effects of the increased GH or IGF-1 levels. This provides a compelling mechanistic rationale for targeting VAT reduction as a means to improve the metabolic health of individuals with HIV-associated lipodystrophy.
Table 2: Key Efficacy Outcomes from Pivotal Phase III Trials (26 Weeks, Tesamorelin 2 mg vs. Placebo)
Outcome Measure | Trial | Tesamorelin Group (Mean Change) | Placebo Group (Mean Change) | LS Mean Treatment Difference (95% CI) | P-value | Source Snippet(s) |
---|---|---|---|---|---|---|
VAT (% Change from Baseline) | LIPO-010 | ~ -15% to -18% | ~ -0.6% to +2% | -19.6% (-23.7% to -15.3%) | < 0.001 | 8 |
CTR-1011 | ~ -14% | ~ -2% | -11.7% (-16.2% to -7.1%) | < 0.001 | 10 | |
Waist Circumference (cm Change) | LIPO-010 | Data not specified | Data not specified | -1.8 cm (-2.8 cm to -0.9 cm) | < 0.001 | 13 |
CTR-1011 | Data not specified | Data not specified | -1.3 cm (-2.4 cm to -0.2 cm) | < 0.05 | 13 | |
Triglycerides (% Change) | Pooled/Rep. | Significant Decrease | Minimal Change/Increase | Favored Tesamorelin | Significant | 8 |
HDL Cholesterol (% Change) | Pooled/Rep. | Increase | Minimal Change | Favored Tesamorelin | Significant | 8 |
Total Chol/HDL Ratio (% Change) | Pooled/Rep. | Decrease | Minimal Change | Favored Tesamorelin | Significant | 8 |
Adiponectin (µg/mL Change) | Pooled/Rep. | Increase (e.g., +0.5 ± 2.7) | Decrease (e.g., -0.1 ± 1.3) | Favored Tesamorelin | Significant | 21 |
Note: VAT = Visceral Adipose Tissue; LS = Least Squares; CI = Confidence Interval. Specific mean change values for lipids/adiponectin varied across analyses; significance is consistently reported. Pooled/Rep. = Pooled or Representative data from trials.
Tesamorelin was generally considered well-tolerated in the Phase III clinical trials involving over 800 patients.[8] While a high proportion of participants in both treatment and placebo groups experienced at least one treatment-emergent adverse event (AE), the overall rates were often comparable or only slightly higher in the Tesamorelin groups compared to placebo groups during the main 26-week phase.[13]
The most frequently reported AEs associated with Tesamorelin use, often occurring at rates higher than placebo, include:
Several significant risks and potential SAEs are associated with Tesamorelin therapy, primarily related to its mechanism of action involving GH stimulation:
Tesamorelin is strictly contraindicated under the following conditions:
Key precautions include monitoring for hypersensitivity reactions, assessing glucose status before and during therapy, monitoring IGF-1 levels, being vigilant for signs of fluid retention, carefully considering use in patients with a history of malignancy, and being aware of the potential for increased mortality risk in acute critical illness.
Breastfeeding is not recommended for mothers using Tesamorelin. This is primarily due to the recommendation by the Centers for Disease Control and Prevention (CDC) that HIV-infected mothers in the United States should not breastfeed to avoid the risk of postnatal HIV transmission. Additionally, the potential for serious adverse reactions in nursing infants from Tesamorelin itself is unknown.[9]
The safety profile of Tesamorelin is largely predictable based on its mechanism of action, involving the stimulation of the GH/IGF-1 axis. This necessitates careful patient selection to exclude those with contraindications (especially active malignancy or pituitary disruption) and diligent monitoring during therapy for potential metabolic complications (glucose intolerance, persistently high IGF-1), fluid retention, and hypersensitivity reactions.
Table 3: Summary of Key Safety Information for Tesamorelin
Category | Specific Event/Condition | Key Details/Management | Source Snippet(s) |
---|---|---|---|
Common AEs (>5%) | Injection Site Reactions (Erythema, Pruritus, Pain, Swelling, etc.) | Rotate injection sites within abdomen. | 2 |
Arthralgia (Joint Pain) | Often related to fluid retention. Monitor. | 7 | |
Peripheral Edema (Swelling of extremities) | Often related to fluid retention. Monitor. | 7 | |
Pain in Extremity | Often related to fluid retention/musculoskeletal effects. Monitor. | 7 | |
Myalgia (Muscle Pain) | Often related to fluid retention/musculoskeletal effects. Monitor. | 7 | |
Serious Risks/Warnings | Hypersensitivity Reactions | Can range from rash/urticaria to serious systemic reactions. Seek immediate medical attention if severe symptoms occur; discontinue treatment. | 2 |
Glucose Intolerance / Diabetes Mellitus | Increased risk due to GH effects. Evaluate glucose prior to and during therapy. Monitor diabetics closely. Consider discontinuation if develops without efficacy. | 1 | |
Increased Risk of Neoplasms | Contraindicated in active malignancy. Caution with history of malignancy/neoplasms; assess benefit-risk. Discontinue if recurrence. | 2 | |
Elevated IGF-1 Levels | Monitor IGF-1 during therapy. Long-term effects unknown. Consider discontinuation for persistent elevation (>3 SDS), especially if efficacy is poor. | 2 | |
Fluid Retention (Edema, Arthralgia, Carpal Tunnel Syndrome) | Monitor for symptoms. May be transient or require discontinuation. | 9 | |
Increased Mortality in Acute Critical Illness | Consider discontinuing therapy in patients who become acutely critically ill (based on GH studies). | 9 | |
Contraindications | Disruption of Hypothalamic-Pituitary Axis (tumor, surgery, trauma, hypopituitarism etc.) | Do not use. | 9 |
Active Malignancy | Do not use. | 9 | |
Known Hypersensitivity to Tesamorelin or Mannitol | Do not use. | 9 | |
Pregnancy (Category X) | Do not use. Potential fetal harm. Use effective contraception. | 1 |
Tesamorelin has several clinically relevant drug interactions, primarily stemming from its effects on the GH/IGF-1 axis and related enzyme systems.
A significant interaction exists with glucocorticoids such as cortisone and prednisone.[1] Tesamorelin inhibits the activity of 11β-hydroxysteroid dehydrogenase type 1 (11βHSD-1), a microsomal enzyme essential for converting the inactive pro-drugs cortisone and prednisone into their active metabolites, cortisol and prednisolone, respectively.[9] By inhibiting this conversion, Tesamorelin can reduce the intracellular concentrations of active glucocorticoids, potentially leading to decreased efficacy.[1] This interaction is particularly important for patients receiving glucocorticoid replacement therapy (e.g., for adrenal insufficiency or hypopituitarism). These patients may require an increase in their maintenance or stress doses of cortisone acetate or prednisone upon initiation of Tesamorelin therapy to compensate for the reduced activation.[9] Close monitoring for signs of adrenal insufficiency or reduced glucocorticoid effect is warranted.
Growth hormone, the secretion of which is stimulated by Tesamorelin, has been shown to modulate the activity of hepatic Cytochrome P450 (CYP450) enzymes.[8] This creates the potential for Tesamorelin to alter the clearance of various drugs that are substrates for CYP450 enzymes. Examples of drug classes potentially affected include corticosteroids (beyond the 11βHSD-1 interaction), sex steroids (e.g., estrogens, androgens), anticonvulsants (e.g., carbamazepine, phenytoin), and immunosuppressants like cyclosporine.[9] While the specific CYP isozymes affected and the magnitude of the interaction may vary, clinical vigilance and potentially therapeutic drug monitoring (e.g., for anticonvulsants like carbamazepine and phenytoin) are recommended when Tesamorelin is co-administered with sensitive CYP450 substrates.[29] Specific interactions noted for monitoring include those with conjugated estrogens, estradiol combinations, levonorgestrel, medroxyprogesterone, norethindrone (potentially affecting contraceptive efficacy), theophylline, and irinotecan liposomal.[29]
Macimorelin is a diagnostic agent used to assess growth hormone deficiency by stimulating GH release. Co-administration with Tesamorelin (or other drugs affecting GH secretion) can interfere with the diagnostic accuracy of the macimorelin test by blunting the expected GH response.[29] Therefore, Tesamorelin therapy should be discontinued at least one week prior to administering a macimorelin diagnostic test.[29]
Concomitant use of Tesamorelin and ulipristal should be avoided. Tesamorelin may decrease the serum concentration or effect of ulipristal, potentially through altered metabolism, reducing its efficacy.[29]
Given Tesamorelin's potential to affect glucose tolerance, caution and monitoring are advised when it is used concurrently with other drugs that impact blood glucose levels, such as diazoxide or incretin mimetics like exenatide.[29]
No specific interactions between Tesamorelin and food have been identified or reported.[1]
The interaction profile highlights the need for careful review of concomitant medications before initiating Tesamorelin. The glucocorticoid interaction is mechanistically well-defined and requires proactive management in patients on replacement therapy. The potential for broader CYP450 interactions necessitates awareness and monitoring for altered efficacy or toxicity of co-administered drugs metabolized through these pathways.
Tesamorelin has been available in different formulations over time, with recent changes aimed at improving patient convenience.
The development pathway from daily to weekly reconstitution with Egrifta WR™ addresses a significant practical challenge associated with chronic daily injectable therapies. By simplifying the preparation process and reducing the injection volume, the newer formulation aims to reduce patient burden and potentially improve long-term adherence, which is critical for maintaining the therapeutic benefit of VAT reduction.[12]
Theratechnologies has indicated that it is developing a multi-dose pen injector intended for use with the Egrifta WR™ (F8) formulation, which could further enhance administration convenience.[14]
The regulatory status of Tesamorelin varies significantly across major global regions.
Tesamorelin has received regulatory approval from the U.S. Food and Drug Administration (FDA).
Tesamorelin is not approved for use in the European Union. Theratechnologies formally withdrew its Marketing Authorisation Application (MAA) submitted to the European Medicines Agency (EMA) in June 2012.[16] The primary reasons cited by the EMA's Committee for Medicinal Products for Human Use (CHMP) for a likely negative opinion, leading to the withdrawal, were insufficient long-term safety data (beyond 48 weeks) regarding the effects of sustained elevation of IGF-1 levels, and the lack of long-term cardiovascular outcome data to adequately assess the overall risk-benefit ratio in the target population.[16] Available EMA documents confirm the authorization of other drugs but not Tesamorelin.[34]
The regulatory status in Canada appears to be not approved. Health Canada's Therapeutic Products Directorate issued a Notice of Non-compliance-Withdrawal (NON/w) for the Tesamorelin New Drug Submission (NDS) in March 2013.[35] Although Health Canada agreed to resume its review of the submission in November 2013 following discussions with the sponsor [36], there is no indication in the provided materials or readily accessible public databases that Tesamorelin subsequently received approval for marketing in Canada.[1] News reports regarding the 2025 FDA approval of Egrifta WR™ do not mention concurrent Canadian approval.[12]
This divergence in regulatory outcomes between the US and other major agencies like the EMA and Health Canada highlights differing interpretations of the available clinical data package or variations in regulatory requirements and risk tolerance. Specifically, the concerns regarding the long-term implications of elevated IGF-1 levels and the absence of dedicated cardiovascular safety outcome data appear to have been pivotal factors preventing approval outside the United States.[16]
While Tesamorelin's primary approved indication is for HIV-associated lipodystrophy, research has explored its potential utility in related metabolic conditions, particularly non-alcoholic fatty liver disease (NAFLD).
Non-alcoholic fatty liver disease (NAFLD) and its more severe form, non-alcoholic steatohepatitis (NASH), are increasingly recognized comorbidities in people living with HIV, potentially affecting up to 25% of this population in developed countries.[32] NAFLD is characterized by excess fat accumulation in the liver (hepatic steatosis) and can progress to inflammation, fibrosis (scarring), cirrhosis, and liver cancer.[32] Lipodystrophy, particularly the visceral fat accumulation targeted by Tesamorelin, is considered a risk factor for NAFLD development in HIV patients.[32]
Clinical trial evidence has emerged supporting a role for Tesamorelin in managing NAFLD in this specific population:
These findings suggest that Tesamorelin offers a potential therapeutic benefit for managing NAFLD in patients with HIV, particularly those who also exhibit central adiposity. By reducing liver fat content and preventing fibrosis progression, it addresses a significant comorbidity beyond the cosmetic improvement of abdominal fat accumulation.[32] The lack of effect on established fibrosis, however, underscores the potential importance of early diagnosis and intervention in NAFLD to achieve the greatest clinical benefit from such therapies.[33]
Given the positive results in the HIV/NAFLD population, investigations into the efficacy of Tesamorelin for reducing liver fat in non-HIV individuals with NAFLD were reported to be ongoing or planned.[32] However, results from such studies were not available in the reviewed materials.
Tesamorelin has also been evaluated, at least preliminarily, as a potential therapy for conditions characterized by insulin resistance and obesity, leveraging its effects on GH/IGF-1 and lipolysis.[2] Further details on these investigations were limited in the provided sources.
Tesamorelin is a synthetic GHRH analog specifically developed and approved in the United States for the reduction of excess visceral adipose tissue (VAT) in adult patients with HIV-associated lipodystrophy. Its mechanism involves stimulating the pituitary gland to release endogenous growth hormone, which in turn increases IGF-1 levels and promotes lipolysis, preferentially targeting VAT.
Clinical efficacy has been robustly demonstrated in Phase III trials, showing statistically significant reductions in VAT (approximately 15% over 26 weeks) compared to placebo. This VAT reduction is associated with improvements in waist circumference, lipid profiles (notably triglycerides and HDL cholesterol), and adiponectin levels. Importantly, analyses suggest these metabolic benefits are strongly linked to the degree of VAT reduction achieved, highlighting the therapeutic value of targeting this specific fat depot. Continuous therapy is required to maintain the reduction in VAT.
The safety profile of Tesamorelin is primarily characterized by adverse events related to increased GH/IGF-1 activity and subcutaneous administration. Common side effects include injection site reactions, arthralgia, peripheral edema, myalgia, and other musculoskeletal complaints often related to fluid retention. Significant risks requiring monitoring and caution include the potential for glucose intolerance or diabetes mellitus, hypersensitivity reactions, sustained elevations in IGF-1 levels (with unknown long-term consequences), and a theoretical increased risk of neoplasm progression (contraindicated in active malignancy).
Drug interactions of note include reduced efficacy of cortisone and prednisone due to inhibition of 11βHSD-1, and potential alterations in the metabolism of CYP450 substrates.
The development of the Egrifta WR™ formulation, allowing for weekly reconstitution while maintaining daily injection and reducing administration volume, represents a significant improvement in convenience that may enhance patient adherence to this chronic therapy.
Tesamorelin occupies a specific niche in managing a complication of HIV and its treatment. Its use requires careful patient selection, exclusion of contraindications, ongoing monitoring for metabolic and other side effects, and patient education regarding the need for continuous treatment. The divergence in regulatory approval between the US and other regions underscores unresolved questions regarding long-term safety, particularly cardiovascular outcomes and the effects of prolonged IGF-1 elevation. Emerging evidence suggests a promising role for Tesamorelin in managing NAFLD and preventing fibrosis progression in the HIV population, potentially expanding its therapeutic utility in addressing comorbidities associated with HIV and lipodystrophy.
Published at: May 12, 2025
This report is continuously updated as new research emerges.