REPORT ID: RECON-2024-MECH-I01

INTEL REPORT: Comparative Mechanisms of Action in Therapeutic Peptides

Classification: CONFIDENTIAL
Updated: 2024-10-08
INTEL REPORT: Peptide Mechanism Analysis - RECON-2024-MECH-I01

REPORT ID: RECON-2024-MECH-I01

CLASSIFICATION: SECRET

TOPIC: Cross-Cutting Peptide Mechanism Analysis

DATE: 2024

INTEL REPORT: Comparative Mechanisms of Action in Therapeutic Peptides

I. EXECUTIVE INTELLIGENCE SUMMARY

This tactical intelligence assessment examines the mechanistic landscape of advanced therapeutic peptides currently under evaluation in performance enhancement, metabolic optimization, and regenerative medicine applications. Through systematic analysis of receptor binding profiles, downstream signaling cascades, and cellular-level physiological effects, this report establishes a comparative framework for understanding how structurally diverse peptides achieve their documented biological outcomes.

The peptide agents analyzed in this assessment represent distinct mechanistic classes: growth hormone secretagogues (GHRPs and GHRH analogs), melanocortin receptor agonists, thymosin derivatives, copper-binding peptides, antimicrobial defense modulators, and mitochondrial-targeted compounds. Despite their structural heterogeneity, these agents share common tactical features including receptor-mediated signal transduction, pulsatile or sustained hormone modulation, and tissue-selective activity patterns that minimize systemic adverse effects.

Intelligence gathered from peer-reviewed clinical and preclinical studies reveals that therapeutic efficacy correlates strongly with receptor subtype selectivity, pharmacokinetic profiles, and the integration of peptide signaling with endogenous regulatory networks. Understanding these mechanistic principles is essential for tactical deployment, cycle architecture, and risk mitigation in operational contexts.

II. RECEPTOR BINDING PROFILES AND SELECTIVITY PATTERNS

The initial tactical consideration in peptide mechanism analysis is the receptor binding profile, which determines both primary biological activity and potential off-target effects. Therapeutic peptides demonstrate remarkable specificity for their cognate receptors, a characteristic that differentiates them from small-molecule pharmaceuticals with broader binding patterns.

Growth Hormone Secretagogue Receptors

The growth hormone secretagogue receptor type 1a (GHS-R1a), commonly termed the ghrelin receptor, serves as the primary target for peptides including Ipamorelin, GHRP-2, GHRP-6, and hexarelin. These compounds function as ghrelin mimetics, binding to the GHS-R1a with high affinity and inducing conformational changes that activate intracellular G-protein coupled signaling cascades. Ipamorelin demonstrates particularly high selectivity, with minimal activity at receptors for cortisol, prolactin, or thyroid hormones, resulting in a clean growth hormone release profile without the appetite stimulation or cortisol elevation observed with earlier generation GHRPs [Source: Raun et al., 1998].

Growth hormone-releasing hormone (GHRH) analogs such as CJC-1295 and Sermorelin operate through a distinct receptor mechanism, binding to the GHRH receptor (GHRHR) expressed on somatotroph cells within the anterior pituitary. The modified structure of CJC-1295, which incorporates Drug Affinity Complex (DAC) technology, extends receptor occupancy through albumin binding, prolonging the duration of GHRHR activation and enabling sustained growth hormone secretion over multiple days rather than hours [Source: Jetté et al., 2005].

Melanocortin Receptor Systems

Melanocortin receptor agonists represent another mechanistic class with significant tactical applications. Melanotan II demonstrates broad-spectrum activity across melanocortin receptor subtypes MC1R, MC3R, MC4R, and MC5R, each mediating distinct physiological responses. MC1R activation in melanocytes drives eumelanin synthesis and photoprotection; MC4R stimulation in hypothalamic centers modulates appetite, energy expenditure, and sexual function; MC3R and MC5R contribute to immune modulation and exocrine gland function. This multi-receptor activity profile produces the compound's characteristic effects on pigmentation, body composition, and libido, but also necessitates tactical consideration of dose-dependent side effects including nausea and spontaneous erections [Source: Wessells et al., 2000].

Pattern Recognition Receptors and Immunomodulation

Thymosin peptides including Thymosin Alpha-1 and Thymosin Beta-4 operate through more complex, less receptor-specific mechanisms involving modulation of innate immune pattern recognition receptors and direct nuclear transcription factor regulation. TB-500, the synthetic analog of Thymosin Beta-4, functions through G-actin sequestration and modulation of actin polymerization dynamics, influencing cell migration, angiogenesis, and wound healing through cytoskeletal reorganization rather than classical receptor-ligand binding [Source: Goldstein et al., 2012].

Table 1: Comparative Receptor Binding Profiles
Peptide Class Primary Receptor Target Receptor Type Binding Affinity (Ki/EC50) Selectivity Profile
Ipamorelin GHS-R1a GPCR EC50: 2.1 nM High (GH-selective)
CJC-1295 GHRHR GPCR EC50: 0.8 nM Very High
Melanotan II MC1R/MC3R/MC4R/MC5R GPCR EC50: 1-10 nM (varies by subtype) Broad (multi-receptor)
BPC-157 Multiple (angiogenic factors) Growth factor pathways Not defined (multi-target) Tissue-selective
TB-500 G-actin Cytoskeletal protein Kd: ~0.5 μM Actin-specific
LL-37 Bacterial membranes / TLRs Pattern recognition Variable (charge-dependent) Broad antimicrobial

III. SIGNAL TRANSDUCTION CASCADES AND CELLULAR MESSAGING

Following receptor engagement, therapeutic peptides initiate complex intracellular signaling cascades that amplify the initial binding event into coordinated cellular responses. The nature of these pathways determines both the speed and duration of peptide effects, as well as their tissue-specific activity patterns.

GPCR-Mediated cAMP/PKA Signaling

Growth hormone secretagogues and GHRH analogs activate classical G-protein coupled receptor pathways. Upon peptide binding, the GPCR undergoes conformational changes that activate heterotrimeric G-proteins (primarily Gq and Gs subtypes). The alpha subunit of Gs stimulates adenylyl cyclase, catalyzing the conversion of ATP to cyclic AMP (cAMP), a critical second messenger. Elevated cAMP levels activate protein kinase A (PKA), which phosphorylates transcription factors including CREB (cAMP response element-binding protein). In somatotroph cells, this cascade culminates in the transcription and secretion of growth hormone, with effects observable within 20-30 minutes of peptide administration and peak GH levels occurring at 40-60 minutes post-injection.

The temporal dynamics of GPCR signaling explain the pulsatile nature of growth hormone release following GHRP administration. Unlike exogenous GH, which provides sustained supraphysiological levels, GHRPs generate physiological pulses that more closely mimic endogenous secretion patterns, preserving feedback regulation and receptor sensitivity.

MAPK/ERK Pathway Activation

Multiple peptides including BPC-157 and TB-500 activate mitogen-activated protein kinase (MAPK) pathways, particularly the extracellular signal-regulated kinase (ERK) cascade. This pathway transduces signals from surface receptors to nuclear transcription factors, regulating cell proliferation, differentiation, and survival. BPC-157 has been demonstrated to enhance ERK1/2 phosphorylation in multiple tissue types, contributing to its documented effects on angiogenesis, fibroblast migration, and collagen synthesis in wound healing contexts. The MAPK pathway also cross-talks with growth factor signaling networks including VEGF (vascular endothelial growth factor) and FGF (fibroblast growth factor), amplifying regenerative responses through multiple parallel mechanisms [Source: Sikiric et al., 2013].

PI3K/AKT Pathway and Metabolic Regulation

The phosphoinositide 3-kinase (PI3K) and protein kinase B (AKT) pathway represents a critical hub for metabolic regulation, cell survival, and protein synthesis. Growth hormone, released in response to secretagogue administration, activates this pathway in target tissues including skeletal muscle, adipose, and liver. AKT phosphorylation promotes glucose uptake through GLUT4 translocation, activates mTOR (mechanistic target of rapamycin) to drive protein synthesis, and inhibits FoxO transcription factors that would otherwise promote protein degradation. This explains the anabolic effects of GH secretagogues on muscle tissue and their ability to improve body composition even in caloric restriction contexts.

Table 2: Primary Signaling Pathways by Peptide Class
Peptide Primary Pathway Secondary Pathways Key Effector Molecules Activation Timeline
Ipamorelin Gs/cAMP/PKA Ca2+ mobilization CREB, GH 20-60 minutes
CJC-1295 Gs/cAMP/PKA IP3/DAG CREB, GH, IGF-1 Sustained (days)
BPC-157 MAPK/ERK FAK, VEGF, eNOS ERK1/2, VEGFR Hours to days
TB-500 Actin dynamics MAPK, Notch G-actin, integrin Hours to weeks
Melanotan II Gs/cAMP/PKA MITF, POMC Tyrosinase, α-MSH Hours (pigmentation: days)
GHK-Cu TGF-β pathway Metalloproteinase modulation Collagen I/III, MMP-2 Days to weeks

IV. TISSUE-LEVEL PHYSIOLOGICAL EFFECTS AND ORGAN SYSTEM INTEGRATION

The ultimate tactical value of peptide therapeutics derives from their tissue-level effects and integration with complex physiological systems. Unlike receptor binding and signal transduction, which occur on millisecond to minute timescales, tissue-level remodeling and systemic adaptations occur over days to weeks and represent the measurable outcomes of peptide interventions.

Anabolic Effects in Skeletal Muscle

Growth hormone secretagogues exert powerful anabolic effects in skeletal muscle tissue through multiple convergent mechanisms. The GH released by secretagogue administration stimulates hepatic and local IGF-1 (insulin-like growth factor-1) production. IGF-1 binds to IGF-1 receptors on myocytes, activating the PI3K/AKT/mTOR pathway and driving protein synthesis while simultaneously inhibiting protein degradation through FoxO suppression. Clinical studies demonstrate that sustained GH secretagogue administration increases lean body mass, with the combination of a GHRP and GHRH analog (such as Ipamorelin with CJC-1295) producing additive effects through complementary mechanisms of GH pulse generation and pulse amplitude enhancement.

Adipose Tissue Mobilization and Metabolic Effects

Growth hormone demonstrates potent lipolytic activity in adipose tissue, particularly in visceral fat deposits. GH activates hormone-sensitive lipase (HSL) through PKA-mediated phosphorylation, catalyzing the hydrolysis of triglycerides into free fatty acids and glycerol. These liberated fatty acids become available for oxidation in skeletal muscle and other tissues, improving body composition through preferential fat loss while preserving or building lean mass. This metabolic reprogramming explains the documented effects of GH secretagogues in improving body composition metrics even during caloric maintenance or moderate restriction.

Cardiovascular and Angiogenic Responses

Several peptides including BPC-157, TB-500, and GHK-Cu demonstrate significant cardiovascular and angiogenic effects. BPC-157 promotes angiogenesis through upregulation of VEGF expression and activation of the VEGF receptor signaling pathway, leading to endothelial cell proliferation, migration, and tube formation. This angiogenic activity has been documented in multiple injury models, where enhanced microvascular density accelerates tissue repair and functional recovery. TB-500 similarly promotes angiogenesis through both VEGF-dependent and VEGF-independent mechanisms, including direct effects on endothelial cell migration and differentiation [Source: Smart et al., 2007].

Dermal Remodeling and Extracellular Matrix Synthesis

The copper peptide GHK-Cu operates through distinct mechanisms involving extracellular matrix remodeling. Copper ions delivered by the peptide serve as essential cofactors for lysyl oxidase, the enzyme responsible for cross-linking collagen and elastin fibers. GHK-Cu also modulates matrix metalloproteinase (MMP) expression, enhancing MMP-2 while suppressing MMP-1 and MMP-9, resulting in balanced matrix turnover that favors collagen synthesis over degradation. These effects translate to improved dermal thickness, elasticity, and tensile strength in aging skin, with documented improvements in wound healing and scar remodeling.

Immune System Modulation

Thymosin Alpha-1 and antimicrobial peptides like LL-37 exert significant immunomodulatory effects through distinct mechanisms. Thymosin Alpha-1 enhances T-cell maturation and function, increases expression of TLR (Toll-like receptor) signaling components, and promotes dendritic cell maturation, collectively strengthening both innate and adaptive immune responses. LL-37 demonstrates direct antimicrobial activity through membrane disruption of bacterial pathogens while also functioning as an immune signaling molecule, recruiting and activating immune cells to sites of infection. These dual functions position LL-37 as both a direct anti-infective agent and an immune system amplifier.

Table 3: Tissue-Specific Effects and Therapeutic Applications
Peptide Primary Target Tissues Cellular Effects Measurable Outcomes Timeline to Effect
Ipamorelin Skeletal muscle, adipose, bone ↑ Protein synthesis, ↑ lipolysis Lean mass gain, fat loss 4-8 weeks
CJC-1295 Systemic (via GH/IGF-1) ↑ IGF-1, ↑ collagen synthesis Body composition, recovery 4-12 weeks
BPC-157 GI tract, tendons, vasculature ↑ Angiogenesis, ↑ fibroblast migration Accelerated healing, reduced inflammation 1-4 weeks
TB-500 Muscle, tendon, cardiac ↑ Cell migration, ↑ differentiation Tissue repair, flexibility 2-6 weeks
Melanotan II Melanocytes, hypothalamus, adipose ↑ Eumelanin, ↑ MC4R activation Tanning, appetite suppression, libido Pigmentation: 1-3 weeks
GHK-Cu Skin, wound sites ↑ Collagen, ↑ angiogenesis, ↓ inflammation Skin quality, wound closure 4-8 weeks
LL-37 Mucosa, skin, immune cells Membrane disruption, immune recruitment Antimicrobial defense, wound healing Hours to days

V. PHARMACOKINETIC PROFILES AND TACTICAL ADMINISTRATION CONSIDERATIONS

The pharmacokinetic properties of therapeutic peptides—including absorption, distribution, metabolism, and elimination—critically determine dosing protocols, administration frequency, and the temporal relationship between administration and measurable effects. Unlike small-molecule drugs with oral bioavailability, most therapeutic peptides require parenteral administration due to rapid enzymatic degradation in the gastrointestinal tract.

Absorption and Bioavailability

Subcutaneous injection represents the standard route of administration for most therapeutic peptides, providing reliable absorption with bioavailability typically ranging from 60-90%. The subcutaneous depot allows gradual peptide release into systemic circulation, producing sustained receptor activation compared to intravenous bolus injection. Absorption half-lives vary from 15 minutes for unmodified peptides to several hours for peptides with fatty acid modifications or other half-life extension technologies.

CJC-1295 with DAC exemplifies advanced peptide engineering for pharmacokinetic optimization. The Drug Affinity Complex technology involves attachment of a reactive chemical group that forms covalent bonds with albumin following injection. This albumin binding dramatically extends the elimination half-life from approximately 30 minutes (for unmodified GHRH) to 6-8 days, enabling sustained GH elevation with dosing frequencies of once or twice weekly rather than multiple daily injections.

Metabolism and Elimination

Peptide metabolism occurs primarily through enzymatic cleavage by proteases in plasma, tissues, and renal tubular cells. The kidney represents the major elimination route for most small peptides, with glomerular filtration and tubular secretion contributing to clearance. Larger peptides may undergo reticuloendothelial system uptake and lysosomal degradation. The metabolic vulnerability of peptides to proteolytic degradation has driven the development of modified peptides with D-amino acids or other non-natural residues that confer protease resistance while preserving receptor binding activity.

Temporal Dynamics of Peptide Effects

Understanding the temporal relationship between peptide administration and physiological effects is essential for tactical deployment. Acute effects such as GH release following Ipamorelin administration occur within 20-60 minutes and persist for 2-3 hours. Intermediate effects including IGF-1 elevation become apparent within 4-8 hours and may persist for 24-48 hours. Long-term adaptive effects including changes in body composition, tissue remodeling, or immune system enhancement require sustained administration over weeks to months.

This temporal layering of peptide effects necessitates strategic cycle design. Growth hormone secretagogues demonstrate optimal efficacy with dosing frequencies of 1-3 times daily, timed to avoid interference with endogenous GH pulses. Regenerative peptides like BPC-157 and TB-500 may be administered once or twice daily with sustained tissue-level effects accumulating over weeks. The long half-life of CJC-1295 with DAC enables simple dosing schedules of 1-2 injections per week while maintaining elevated GH secretion throughout the dosing interval.

VI. MECHANISTIC SYNERGY AND COMBINATION PROTOCOLS

A sophisticated understanding of peptide mechanisms enables rational combination protocols that leverage complementary or synergistic effects. The concept of mechanistic synergy—where combined agents produce effects greater than the sum of individual contributions—represents an advanced tactical approach to peptide deployment.

GHRP/GHRH Synergy

The combination of a growth hormone releasing peptide (such as Ipamorelin) with a GHRH analog (such as CJC-1295) represents the canonical example of mechanistic synergy. These agents operate through distinct receptor systems and signaling mechanisms but converge on the same target cell population (somatotrophs) to produce amplified GH release. GHRP administration generates a strong GH secretory pulse, while GHRH analog co-administration increases the amplitude of that pulse through complementary signaling pathway activation. Clinical data demonstrate that combined GHRP/GHRH administration produces GH levels 2-3 times higher than either agent alone at equivalent doses, with additive rather than merely additive effects on body composition and metabolic outcomes [Source: Laron et al., 1995].

Regenerative Peptide Stacking

The combination of BPC-157 and TB-500 for injury recovery represents another mechanistically rational approach. BPC-157 primarily enhances angiogenesis through VEGF pathway activation, while TB-500 promotes cell migration and differentiation through actin cytoskeleton modulation. These non-overlapping mechanisms address different rate-limiting steps in tissue repair, with BPC-157 ensuring adequate microvascular supply to healing tissues and TB-500 facilitating the migration of progenitor cells and fibroblasts into the injury site. The addition of GHK-Cu provides further mechanistic complementarity through its effects on collagen cross-linking and matrix remodeling.

GH Secretagogue and Anabolic Peptide Combinations

Advanced protocols may combine GH secretagogues with direct tissue-targeted peptides. For example, the use of a GHRP/GHRH combination to elevate systemic GH and IGF-1 levels creates an anabolic hormonal environment, while concurrent BPC-157 administration targets specific injury sites with localized regenerative effects. This approach leverages both systemic metabolic optimization and targeted tissue repair, potentially accelerating recovery while simultaneously improving body composition.

Table 4: Rational Peptide Combinations and Mechanistic Rationale
Combination Mechanism of Agent 1 Mechanism of Agent 2 Synergistic Rationale Tactical Application
Ipamorelin + CJC-1295 GHS-R1a activation (pulse generation) GHRHR activation (pulse amplitude) Complementary GH secretion pathways Body composition, recovery, anti-aging
BPC-157 + TB-500 VEGF-mediated angiogenesis Actin dynamics, cell migration Non-overlapping repair mechanisms Injury recovery, tissue repair
BPC-157 + GHK-Cu Growth factor upregulation Collagen synthesis, MMP modulation Angiogenesis + matrix remodeling Wound healing, skin quality
Melanotan II + Mod GRF 1-29 MC4R appetite suppression GH-mediated lipolysis Complementary fat loss mechanisms Body recomposition, cutting phases
Thymosin Alpha-1 + LL-37 Adaptive immune enhancement Innate immune defense, direct antimicrobial Comprehensive immune support Infection prevention/recovery, immune optimization

VII. MECHANISTIC BASIS OF ADVERSE EFFECTS AND RISK MITIGATION

Understanding the mechanisms underlying potential adverse effects is essential for risk assessment and mitigation. Unlike the therapeutic effects which result from intended receptor interactions, adverse effects typically arise from off-target receptor binding, excessive pathway activation, or disruption of homeostatic feedback systems.

Receptor-Mediated Side Effects

The broad melanocortin receptor activity of Melanotan II illustrates how off-target receptor binding produces predictable side effects. While MC1R activation produces desired tanning effects and MC4R stimulation contributes to appetite suppression and body composition benefits, these same receptor activations also produce less desirable effects. MC4R activation in neural centers controlling sexual function can produce spontaneous erections and increased libido, effects that may be desired by some users but problematic for others. Nausea and flushing result from MC4R and MC5R activation in hypothalamic and brainstem centers. These effects are dose-dependent and typically attenuate with continued use as receptor desensitization occurs.

Feedback System Disruption

Chronic administration of supraphysiological doses of any GH-elevating peptide carries theoretical risks of disrupting the hypothalamic-pituitary-somatotroph axis. Unlike exogenous GH administration which suppresses endogenous production through negative feedback, GH secretagogues preserve pulsatile secretion patterns and maintain feedback sensitivity. However, chronic excessive stimulation could theoretically lead to receptor desensitization or altered sensitivity of feedback mechanisms. Tactical protocols typically incorporate periodic discontinuation or cycling to preserve long-term responsiveness.

Injection Site Reactions and Antibody Formation

As foreign proteins, peptides carry inherent immunogenic potential. Repeated injection of the same peptide can theoretically induce antibody formation, potentially reducing efficacy or producing allergic reactions. However, clinical experience with therapeutic peptides suggests that most commonly used compounds have low immunogenicity, with injection site reactions (redness, swelling, itching) representing localized inflammatory responses rather than systemic immune reactions. Proper injection technique, site rotation, and use of pharmaceutical-grade peptides minimize these risks.

Risk Mitigation Through Mechanistic Understanding

Tactical deployment of peptides should incorporate mechanistic knowledge to minimize adverse effect potential. This includes appropriate dose titration (starting with lower doses and gradually increasing), timing administration to align with circadian rhythms and endogenous hormone patterns, incorporating wash-out periods or cycling protocols, and avoiding concurrent use of peptides with overlapping mechanisms that could produce excessive pathway activation. For growth hormone secretagogues, avoiding bedtime dosing too close to natural nocturnal GH pulses preserves physiological pulsatility. For melanocortin agonists, slow dose escalation allows receptor tolerance to develop, reducing acute side effects.

VIII. TACTICAL CONCLUSIONS AND STRATEGIC IMPLICATIONS

This mechanistic intelligence assessment establishes several key conclusions for tactical peptide deployment:

First, receptor selectivity profiles directly determine both efficacy and side effect profiles. Peptides with high selectivity for their primary target receptors (such as Ipamorelin for GHS-R1a) demonstrate cleaner effect profiles with fewer off-target complications compared to promiscuous receptor binders.

Second, understanding signal transduction cascades enables prediction of temporal dynamics and tissue-specific effects. GPCR-mediated peptides produce relatively rapid effects through second messenger amplification, while peptides operating through gene transcription or cytoskeletal remodeling require longer timeframes for measurable outcomes.

Third, pharmacokinetic optimization through structural modifications (such as DAC technology in CJC-1295) significantly enhances tactical utility by reducing injection frequency while maintaining therapeutic efficacy.

Fourth, mechanistically rational combinations leveraging complementary pathways can produce synergistic effects exceeding those of individual agents, but require sophisticated understanding of receptor systems and signaling networks to implement safely and effectively.

Fifth, adverse effects are mechanistically predictable based on receptor binding profiles and pathway activation patterns, enabling proactive risk mitigation through dose optimization, administration timing, and cycling protocols.

The therapeutic peptide landscape continues to expand as new compounds enter research pipelines and clinical development. However, the fundamental mechanistic principles elucidated in this assessment—receptor-mediated signaling, pathway convergence, temporal dynamics, and tissue-selective effects—provide a stable framework for evaluating both established and emerging peptide therapeutics. Operators equipped with this mechanistic intelligence are positioned to make informed tactical decisions regarding peptide selection, dosing, combination strategies, and risk management in pursuit of performance enhancement, metabolic optimization, and regenerative medicine objectives.

The strategic value of peptide therapeutics lies not merely in their immediate biological effects, but in their ability to modulate endogenous regulatory systems in physiologically appropriate ways. Unlike blunt pharmacological interventions that override normal physiology, well-designed peptide protocols work within existing homeostatic frameworks, amplifying beneficial processes while preserving feedback mechanisms that maintain long-term stability and safety.

As research continues to unravel additional mechanistic details and clinical data accumulates from both formal studies and field experience, the sophistication of peptide deployment protocols will continue to advance. This assessment provides the mechanistic foundation necessary for interpreting emerging data and integrating new tactical intelligence into operational frameworks. The future of performance optimization and regenerative medicine will increasingly rely on peptide-based interventions, guided by deep mechanistic understanding and systematic intelligence analysis of the type presented in this classified assessment.

INTELLIGENCE SOURCES

  1. Raun K, Hansen BS, Johansen NL, et al. Ipamorelin, the first selective growth hormone secretagogue. Eur J Endocrinol. 1998;139(5):552-561. [PubMed: 9849822]
  2. Jetté L, Léger R, Thibaudeau K, et al. Human growth hormone-releasing factor (hGRF)1-29-albumin bioconjugates activate the GRF receptor on the anterior pituitary in rats: identification of CJC-1295 as a long-lasting GRF analog. Endocrinology. 2005;146(7):3179-3186. [PubMed: 16352683]
  3. Wessells H, Fuciarelli K, Hansen J, et al. Synthetic melanotropic peptide initiates erections in men with psychogenic erectile dysfunction: double-blind, placebo controlled crossover study. J Urol. 2000;163(4):1290-1293. [PubMed: 11416836]
  4. Goldstein AL, Goldstein AL. From lab to bedside: emerging clinical applications of thymosin alpha 1. Expert Opin Biol Ther. 2012;12(3):307-319. [PubMed: 17879319]
  5. Sikiric P, Seiwerth S, Rucman R, et al. Stable gastric pentadecapeptide BPC 157: novel therapy in gastrointestinal tract. Curr Pharm Des. 2013;19(1):126-132. [PubMed: 24080448]
  6. Smart N, Risebro CA, Melville AA, et al. Thymosin beta4 induces adult epicardial progenitor mobilization and neovascularization. Nature. 2007;445(7124):177-182. [PubMed: 15358113]
  7. Laron Z, Frenkel J, Gil-Ad I, et al. Growth hormone releasing activity of growth hormone releasing peptide-6 (GHRP-6) in patients with growth hormone deficiency. Clin Endocrinol (Oxf). 1995;43(5):635-640. [PubMed: 9467542]
  8. Khatib MN, Gaidhane A, Gaidhane S, Quazi ZS. Ghrelin as a promising therapeutic option for cancer cachexia. Cell Physiol Biochem. 2018;48(5):2172-2188. [PubMed: 30134245]