Biotech
182212-66-4
Avotermin, also known by the investigational brand name Juvista, is a recombinant human Transforming Growth Factor-beta 3 (TGF−β3) developed as a first-in-class, investigational biologic agent for the prophylactic improvement of cutaneous scarring.[1] It represented a pioneering therapeutic approach aimed at addressing a significant unmet medical need in dermatology, plastic surgery, and general surgery, fields where existing scar treatments often lack robust, evidence-based support from rigorously controlled clinical trials.[3] The development of Avotermin was founded on an elegant scientific rationale derived from the observation of scar-free wound healing in early-gestation fetuses, a phenomenon attributed to a local cytokine environment dominated by
TGF−β3.[5] The therapeutic strategy was to pharmacologically replicate this regenerative environment in adult wounds by administering exogenous
TGF−β3 at the time of surgical incision.
This rationale was strongly supported by an extensive and successful program of Phase I and II clinical trials. These studies, involving over 1,500 human subjects, consistently demonstrated that intradermal injections of Avotermin were well-tolerated and produced statistically significant improvements in scar appearance compared to placebo across a range of surgical settings.[7] These promising results generated considerable optimism and led to a major licensing agreement with Shire plc for development and commercialization outside of Europe.[8]
Despite this strong early-phase performance, the development of Avotermin was abruptly terminated following the failure of its pivotal Phase III REVISE trial in patients undergoing scar revision surgery. In February 2011, the developer, Renovo, announced that the trial did not meet its primary or secondary endpoints.[10] A subsequent post-hoc analysis found no flaws in the trial's design or execution but instead attributed the failure to considerable and unpredictable biological variation in the scarring process within the broader, more heterogeneous patient population.[11] This outcome led to the termination of the licensing deal with Shire and the eventual restructuring and dissolution of Renovo.[9] The trajectory of Avotermin serves as a critical and cautionary case study in pharmaceutical research and development. It starkly illustrates the formidable challenges of translating efficacy from the highly controlled environments of early-phase trials to the complexities of late-stage clinical studies, particularly for conditions characterized by high biological variability and subjective assessment endpoints.
Avotermin is a protein-based biologic classified as a cytokine and a member of the Transforming Growth Factor-beta (TGF−β) superfamily.[13] This large family of signaling proteins plays a central and multifaceted role in numerous cellular processes, including proliferation, differentiation, apoptosis, and extracellular matrix (ECM) production.[14] Within the context of wound healing and tissue repair, the key isoforms—
TGF−β1, TGF−β2, and TGF−β3—exert distinct and often opposing effects that are fundamental to understanding Avotermin's scientific premise.
TGF−β1 and TGF−β2 are widely recognized as potent pro-fibrotic mediators. In adult cutaneous wounds, their expression is significantly elevated, where they drive the inflammatory response, stimulate fibroblast proliferation, and promote the excessive deposition of ECM components, primarily collagen.[2] This rapid but imperfect repair process results in the formation of scar tissue, which is characterized by dense, parallel bundles of collagen fibers that are functionally and aesthetically inferior to normal skin.[6]
In stark contrast, TGF−β3 is associated with an anti-fibrotic and regenerative phenotype. Its role is most clearly demonstrated in the unique biology of fetal wound healing, establishing a functional dichotomy within the TGF−β family that forms the core rationale for Avotermin's development.[6]
The conceptual foundation for Avotermin originated from a key observation in developmental biology: skin wounds created in early-gestation mammalian fetuses heal perfectly, without any trace of scarring.[5] Extensive research into this phenomenon revealed that the local wound environment in the fetus is markedly different from that in an adult. Specifically, fetal wounds are characterized by a high ratio of
TGF−β3 relative to the pro-fibrotic isoforms TGF−β1 and TGF−β2.[6] This unique cytokine milieu is believed to orchestrate a more regenerative, less inflammatory healing response that restores the normal architecture of the skin.[5]
Conversely, the adult wound healing process, which has evolved to prioritize rapid wound closure and barrier function over perfect restoration, is dominated by high levels of TGF−β1 and TGF−β2.[6] This leads to a robust inflammatory phase, rapid fibroblast proliferation, and the deposition of a disorganized, collagen-rich ECM that ultimately manifests as a scar.[16] Experimental evidence strongly supports this hypothesis. For instance, the addition of exogenous
TGF−β1 to a fetal wound induces scar formation, while neutralizing TGF−β1 and TGF−β2 or adding exogenous TGF−β3 to adult wounds can reduce scarring.[6]
Avotermin was therefore developed as a direct therapeutic application of this hypothesis. The strategy was to prophylactically administer recombinant human TGF−β3 directly into the wound margins at the time of injury, with the goal of shifting the local cytokine balance to mimic the fetal environment and thereby promote a more regenerative, scar-free healing outcome.[1]
Avotermin is a biotech therapeutic identified as recombinant human TGF−β3.[1] Biochemically, it is a non-glycosylated, disulfide-linked homodimer.[15] The mature, active protein consists of two identical polypeptide chains, each comprising 112-113 amino acids, giving the homodimer a total molecular mass of approximately 25-26 kDa.[15]
In its natural biological context, human TGF−β3 is synthesized from a 412-amino acid precursor preproprotein encoded by the TGFB3 gene.[19] This precursor undergoes proteolytic processing by a furin-like convertase, which cleaves it into two main components: a 220-amino acid N-terminal fragment known as the Latency-Associated Peptide (LAP) and the 112-amino acid C-terminal mature
TGF−β3 peptide.[21] The mature peptide remains non-covalently associated with LAP, forming a latent complex that is secreted from the cell. This latent complex must be activated in the extracellular space—a process mediated by enzymes like plasmin or matrix metalloproteinases—to release the biologically active
TGF−β3 homodimer.[19] The Avotermin drug product consists solely of the mature, active form of the protein, corresponding to amino acids 301-412 of the full precursor.[23] The amino acid sequence of the recombinant monomer is as follows: MALDTNYCFRN LEENCCVRPL YIDFRQDLGW KWVHEPKGYY ANFCSGPCPY LRSADTTHST VLGLYNTLNP EASASPCCVP QDLEPLTILY YVGRTPKVEQ LSNMVVKSCK CS.[20]
The proposed pharmacodynamic effect of Avotermin is to fundamentally alter the natural course of adult wound healing by intervening at its earliest stages. The adult wound repair process typically unfolds in three overlapping phases: inflammation, proliferation, and remodeling. Avotermin, when administered prophylactically, is believed to reduce both the magnitude and the duration of each of these phases.[6]
By introducing a high local concentration of anti-fibrotic TGF−β3 at the moment of injury, Avotermin is thought to attenuate the initial pro-inflammatory signaling cascade, which is normally driven by TGF−β1 and TGF−β2. This modulation of the inflammatory phase subsequently dampens the proliferative phase, leading to less aggressive fibroblast activation and reduced deposition of extracellular matrix. Finally, by influencing the quality and quantity of the matrix laid down, it facilitates a more efficient and effective remodeling phase. This multi-phase modulation represents a truly regenerative approach, aiming not just to suppress a single pathological step but to guide the entire healing process toward a more favorable outcome.
The ultimate mechanism of action of Avotermin is the promotion of a healed tissue structure that more closely resembles normal, uninjured skin rather than scar tissue.[24] This is achieved primarily by influencing the organization of collagen fibers within the dermis. Adult scar tissue is characterized by fine collagen fibers that are densely packed and aligned in parallel bundles, resulting in tissue that is stiff, weak, and visually distinct from the surrounding skin.[6]
In contrast, preclinical and early clinical studies demonstrated that wounds treated with Avotermin healed with a dermal architecture where the collagen was arranged in a more random, flexible "basket weave" orientation, which is the hallmark of normal skin.[6] This histological improvement was the structural underpinning of the macroscopic benefits observed in clinical trials, such as scars that were narrower, flatter, and less discolored.[7] This ability to guide the restitution of normal dermal architecture, rather than simply reducing the size of a scar, positioned Avotermin as a potentially transformative agent in regenerative medicine. The mechanism's reliance on intervention at the very beginning of the healing process underscores its classification as a prophylactic therapy. The precise timing of administration—at the moment of wounding and again 24 hours later—was found to be critical for achieving this effect, highlighting a potential logistical dependency that may have implications for its application in less controlled clinical settings.[3]
Avotermin's progression to late-stage development was propelled by an extensive and remarkably successful Phase I and II clinical program. This program, which included data from over 1,500 subjects, robustly established the drug's safety profile and provided strong, consistent evidence of its efficacy in improving scar appearance.[7] The trials were conducted across a diverse range of surgical indications, demonstrating the broad potential applicability of the therapy. Key areas of investigation included scar revision surgery, keloid treatment, varicose vein removal, mammaplasty, and studies in healthy volunteers to establish dose-response relationships.[13]
These studies systematically explored a wide dose range, from 5 ng to 500 ng per 100 µL per linear cm of wound margin, as well as different dosing schedules.[16] A key finding from this early work was that a twice-dosing regimen—administered immediately after wound closure and again 24 hours later—yielded greater and more consistent efficacy than a single dose.[26]
Across multiple double-blind, placebo-controlled trials, Avotermin demonstrated statistically significant improvements in scar appearance. Efficacy was assessed using a multi-modal approach that combined subjective and objective endpoints. The primary endpoint in many of these trials was a Visual Analogue Scale (VAS) score, where scars were evaluated by independent lay panels, investigators, and the patients themselves.[26] These subjective assessments were corroborated by objective, quantitative measurements such as profilometry, which measures scar surface area and topography, and histological analysis of scar biopsies, which confirmed the underlying improvement in collagen architecture.[17] For example, in a Phase II trial in scar revision surgery (NCT00432211), Avotermin-treated scars showed significantly improved VAS scores and a greater reduction in scar surface area compared to placebo.[33] Similarly, a Phase II study in patients undergoing varicose vein removal (NCT00430326) found that a single 500 ng dose of Avotermin significantly improved scar appearance as assessed by a lay panel.[30] This consistent pattern of positive results across numerous well-designed trials created a powerful foundation of evidence and high expectations for success in Phase III.
Building on the momentum of the successful Phase II program, Renovo initiated the pivotal REVISE trial (NCT00742443) to confirm the efficacy of Avotermin and support applications for regulatory approval in Europe and the US.[34] The trial was a large-scale, multicenter, double-blind, randomized study designed to enroll approximately 350 patients undergoing surgical revision of existing disfiguring scars.[35]
The trial employed a sophisticated within-patient, placebo-controlled design. After the existing scar was surgically excised, the resulting wound was divided into two equal segments. One segment was randomly assigned to receive intradermal injections of Avotermin (250 ng/100 µL per linear cm), while the other segment received a matching placebo injection.[34] This design is particularly powerful for scar studies as it uses each patient as their own control, thereby minimizing the confounding effects of inter-individual variability in healing, a factor known to be influenced by genetics, age, and anatomical location.[35]
The primary objective of the trial was to confirm the effectiveness of Avotermin in improving the appearance of the revised scar.[34] The primary endpoint, which had been agreed upon with the European Medicines Agency (EMEA), was the visual assessment of the scar at 12 months post-surgery. This assessment was to be performed by an independent panel of experts evaluating standardized photographs of the scars using a validated visual scale.[35] Secondary endpoints likely included assessments by investigators and patients, as well as objective measurements of scar characteristics. The trial was designed to be the definitive test of Avotermin's clinical utility in a broad and clinically relevant patient population.
Table 1: Summary of Key Avotermin Clinical Trials
| NCT Identifier | Phase | Condition / Indication | No. of Patients | Dosing Regimen | Primary Endpoint | Reported Outcome | Source(s) |
|---|---|---|---|---|---|---|---|
| NCT00432211 | 2 | Scar Revision Surgery | 60 | 200 ng, twice (at surgery & 24h later) | Total Scar Score (VAS) by lay panel (Months 1-7) | Statistically significant improvement vs. placebo (p=0.04) | 33 |
| NCT00430326 | 2 | Varicose Vein Removal | 156 | 5, 50, 200, or 500 ng, once | Total Scar Score (VAS) by lay panel (Weeks 6-Month 7) | 500 ng dose showed statistically significant improvement vs. placebo (p=0.036) | 30 |
| NCT00710333 | 1/2 | Keloid Excision | N/A | N/A | Safety | Completed; Investigated safety post-excision | 27 |
| NCT00836147 | 1/2 | Keloid Excision | N/A | 250 ng | Efficacy and Safety | Completed; Exploratory study | 27 |
| NCT00469235 | 1/2 | Keloid Excision | N/A | N/A | N/A | Completed; Investigated use following keloid removal | 27 |
| NCT00978302 | 1 | Promotion of Wound Healing / Scar Prevention | N/A | Escalating doses | Safety | Completed; Safety study of repeated doses | 37 |
| NCT00742443 (REVISE) | 3 | Scar Revision Surgery | ~350 | 250 ng, twice (at surgery & 24h later) | Visual assessment of scar at 12 months | Failed to meet primary and secondary endpoints | 9 |
In a significant setback for the field of scar therapeutics, Renovo announced in February 2011 that the pivotal Phase III REVISE trial of Juvista (Avotermin) had failed. The company stated in a terse release that the trial "did not meet its primary or secondary endpoints".[9] This outcome was met with extreme surprise and disappointment by the company, its partner Shire, and the broader investment and medical communities.[10] The failure was particularly jarring because it directly contradicted the large body of positive and statistically significant efficacy data generated throughout the extensive Phase I and II programs. This disconnect between promising early-phase results and late-stage failure represents a classic and challenging scenario in pharmaceutical development.
Following the announcement, Renovo conducted a thorough exploratory analysis of the trial data to understand the reasons for the unexpected failure. The company's investigation concluded that there were no identifiable flaws in the trial's design, technical execution, or statistical analysis.[11] The methodology, including the within-patient control design and the validated endpoints, was considered robust.
Instead, the analysis pointed to an insurmountable biological challenge: "considerable unpredictable variation was found in the appearance of the two halves of the revised scar in some individuals".[11] This finding suggests that in the broader and more heterogeneous patient population of the Phase III trial, the inherent biological variability of the human scarring response was so great that it masked or overwhelmed the therapeutic effect of Avotermin. Even with the sophisticated within-patient control design intended to minimize such variability, the "noise" of the biological system was greater than the "signal" from the drug.
Furthermore, the post-hoc analysis was unable to identify any meaningful subset of patients who did respond to the treatment, which eliminated the possibility of pursuing a more targeted indication.[11] Some external analyses have suggested that potential inconsistencies between the Phase II and Phase III trial designs—such as subtle differences in patient populations, surgical techniques across multiple centers, or the timing and measurement of outcomes—may have contributed to the different results.[17] Ultimately, the company concluded that the efficacy of Juvista was insufficient to demonstrate a significant benefit in a broad population of scar revision patients.[11] This outcome exemplifies the "valley of death" in drug development, where a promising candidate fails to translate its efficacy from the highly controlled and often more homogenous populations of early trials to the real-world complexity of a large-scale, confirmatory study.
The clinical failure of the REVISE trial had immediate and devastating commercial and corporate consequences. Upon the announcement, Renovo's stock price plummeted by approximately 75%, reflecting a complete loss of investor confidence in the company's lead asset.[9]
The most significant blow came shortly after, when Shire plc, Renovo's commercialization partner, terminated the $825-million licensing agreement and returned the development and commercialization rights for Juvista in the USA, Canada, and Mexico to Renovo.[9] This decision effectively eliminated the primary path to market for Avotermin in key territories and removed a critical source of future milestone payments and revenue.
Faced with the failure of its lead candidate and the loss of its major partner, the Board of Renovo concluded that no further development of Juvista would be initiated.[11] The company immediately began a major restructuring, which included significant reductions in expenditure and workforce, and halting recruitment for other pipeline programs.[11] Ultimately, the failure of Avotermin led to the company's decision to sell off its remaining assets and cease operations, serving as a stark reminder of the high-stakes nature of late-stage clinical development.[38]
Across its extensive clinical development program, Avotermin consistently demonstrated a favorable safety and tolerability profile.[2] Importantly, the discontinuation of the program was based solely on the lack of efficacy in the Phase III trial and was not associated with any safety concerns.[17]
Data from numerous studies indicated that adverse events (AEs) were generally mild, transient, and comparable between the Avotermin and placebo groups. The most commonly reported AEs were localized to the injection site, such as erythema (redness) and edema (swelling).[32] These events were typically considered consistent with the normal inflammatory response of wound healing and were equally frequent in placebo-treated wounds.[39] Systemic AEs, such as headaches, were reported in some patients but did not emerge as a significant safety signal.[39] There were no deaths attributed to the drug, and rates of study withdrawal due to adverse events were negligible and similar to placebo.[39] This benign safety profile was a significant strength of the drug candidate, particularly for a product intended for prophylactic use in elective surgical procedures. The fact that a drug with such a clean safety record failed underscores that efficacy remains the highest hurdle in pharmaceutical development.
Detailed pharmacokinetic data for Avotermin in humans is notably absent from public databases and literature. Major resources such as DrugBank explicitly list parameters for absorption, distribution, metabolism, and excretion (ADME) as "Not Available".[13] This lack of information is common for investigational drugs that are discontinued before reaching the market.
The only available quantitative PK data comes from a preclinical study conducted in a clinically relevant minipig model. In this study, intradermal administration of Avotermin resulted in transient plasma levels of TGF−β3 with a very low systemic bioavailability of approximately 0.1%.[40] This finding strongly suggests that Avotermin was designed to act locally within the microenvironment of the wound, with minimal systemic exposure. This local mechanism of action is consistent with the drug's excellent safety profile, as it would limit the potential for off-target systemic effects. However, it also creates a compelling paradox in light of the drug's failure: a locally acting therapeutic was ultimately defeated by systemic, patient-specific biological variability that it could not overcome at the site of action. This suggests that future anti-scarring therapies may need to be exceptionally potent locally or be combined with strategies that can modulate the systemic drivers of fibrosis.
The development program for Avotermin stands in stark contrast to the landscape of conventional scar management therapies. The field has long been characterized by a variety of treatments—including silicone sheeting, pressure garments, onion extract-based topicals, and intralesional corticosteroid injections—whose efficacy is supported by a mix of empirical evidence, small studies, and inconsistent results.[16] Robust, large-scale, prospective, randomized controlled trials (RCTs) to definitively support the use of many of these common therapies are notably lacking, making it difficult to establish a universally accepted standard of care.[4]
In this context, Avotermin was a trailblazer. Its development was anchored in a strong, mechanistic rationale derived from fundamental biology and was pursued through one of the most rigorous clinical trial programs ever conducted in the field of scar therapy.[2] The extensive series of double-blind, placebo-controlled Phase I and II studies set a new and higher standard for evidence generation in this therapeutic area. While the program ultimately failed to deliver a marketable product, its methodical, evidence-based approach represented a significant maturation for the field and provided a wealth of high-quality data that continues to inform research into wound healing and fibrosis.
Table 2: Comparative Overview of Scar Management Modalities
| Modality | Proposed Mechanism of Action | Typical Use | Level of Clinical Evidence | Source(s) |
|---|---|---|---|---|
| Avotermin (TGF-β3) | Modulates healing phases; Promotes regenerative collagen architecture | Prophylactic | Extensive Phase I/II RCTs; Failed Phase III RCT | 6 |
| Silicone Gel/Sheeting | Hydration, occlusion, pressure; May modulate cytokine expression | Prophylactic & Treatment | Widely used; Considered standard of care, but evidence from large RCTs is mixed | 41 |
| Pressure Garments | Induces hypoxia, reducing collagen synthesis and fibroblast proliferation | Prophylactic & Treatment | Standard for burn scars; Efficacy debated and compliance is low | 45 |
| Intralesional Corticosteroids | Anti-inflammatory; Reduces collagen synthesis and fibroblast proliferation | Treatment (esp. for keloids/hypertrophic scars) | Effective but with common side effects (atrophy, hypopigmentation) | 41 |
| Onion Extract Topicals | Anti-inflammatory, antioxidant; May modulate collagen breakdown | Prophylactic & Treatment | Evidence is limited and often conflicting in clinical studies | 47 |
| Laser Therapy (e.g., PDL) | Targets vasculature, reducing erythema; Modulates collagen remodeling | Treatment | Effective for improving color and texture; Often used in combination | 46 |
The story of Avotermin is a powerful lesson in the distinction between efficacy (a drug's performance under the idealized conditions of a clinical trial) and effectiveness (its performance in a real-world setting). The drug demonstrated clear efficacy in the highly controlled, relatively homogenous populations of its Phase II studies. However, it failed to demonstrate effectiveness when tested in the larger, more heterogeneous population of the Phase III REVISE trial, where the inherent biological variability of the scarring process proved too great to overcome.[11]
This failure highlights the profound challenges posed by patient heterogeneity in clinical development. Scar formation is a complex biological process influenced by a multitude of factors, including genetics, age, ethnicity, anatomical location of the wound, and surgical technique.[17] Avotermin's inability to show a benefit in this complex environment suggests that targeting a single growth factor, however logical, may be an insufficient strategy to reliably modulate such a multifactorial process. The failure also underscores the difficulty of developing drugs for conditions with subjective endpoints, where patient and observer perceptions of "improvement" can be highly variable and difficult to standardize across a large trial.
While Avotermin did not reach the market, its journey provides invaluable guidance for the future of anti-fibrotic and regenerative medicine. The failure of this single-agent approach suggests that the next generation of successful scar therapies may require more sophisticated strategies. These could include combination therapies that target multiple pathways in the fibrotic cascade simultaneously, or the use of advanced drug delivery systems, such as hydrogels or nanoparticles, to provide sustained, localized release of growth factors and better control the wound microenvironment.[17]
Furthermore, the issue of patient heterogeneity that thwarted Avotermin points toward the need for a more personalized approach to treatment. Future clinical trials may benefit from incorporating biomarker strategies or genetic screening to stratify patients and identify those most likely to respond to a specific therapy. Despite its ultimate failure, the Avotermin program successfully validated the TGF−β pathway as a central and druggable target in fibrosis and generated a rich, high-quality clinical dataset. It pushed the boundaries of clinical science in dermatology and left a legacy of rigorous, evidence-based development that will continue to inform and inspire the quest for a truly effective anti-scarring therapy.
Published at: September 15, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.