KPV (Lys-Pro-Val)

Review article authors: Luger TA & Brzoska T, University of Muenster (2007)

Key preclinical studies: Dalmasso G et al., Emory University (2008); Getting SJ et al. (2003)

Nanoparticle delivery: Xiao B et al., Sun Yat-sen University (2017)

KPV is a naturally occurring tripeptide (Lys-Pro-Val) corresponding to the three C-terminal amino acids of α-melanocyte-stimulating hormone (α-MSH), a tridecapeptide hormone derived from proopiomelanocortin (POMC).¹ ² ³ Unlike its parent molecule α-MSH, which binds melanocortin receptors (MC-1R through MC-5R) and produces both anti-inflammatory effects and melanotropic activity (increased pigmentation), KPV retains the full anti-inflammatory and antimicrobial properties of α-MSH without binding to melanocortin receptors or affecting skin pigmentation.¹ ² ³ ⁴ The peptide functions via an entirely distinct mechanism—cellular uptake through the PepT1 transporter (oligopeptide transporter normally expressed in small intestine and upregulated in inflamed colon during inflammatory bowel disease), followed by intracellular inhibition of NF-κB and MAPK inflammatory signaling pathways.³ In preclinical rodent models, oral administration of KPV significantly reduced intestinal inflammation in both dextran sulfate sodium (DSS)- and trinitrobenzene sulfonic acid (TNBS)-induced colitis, demonstrating decreased body weight loss, reduced myeloperoxidase activity (neutrophil infiltration marker), markedly decreased histological inflammation, and significant suppression of pro-inflammatory cytokines (IL-1β, IL-6, IL-12, TNF-α, IFN-γ) without affecting anti-inflammatory IL-10.³ The peptide operated at nanomolar concentrations in vitro, inhibiting NF-κB activation and reducing pro-inflammatory cytokine secretion in intestinal epithelial cells (Caco2-BBE), immune cells (Jurkat T cells), and macrophages.³ Advanced nanoparticle delivery systems targeting inflamed colonic mucosa enhanced KPV’s efficacy 12,000-fold compared to free peptide solution.⁵ Despite robust preclinical evidence spanning contact dermatitis, inflammatory bowel disease, allergic asthma, arthritis, and various inflammatory conditions, KPV has never been evaluated in human clinical trials.⁶ ⁷ In September 2023, the FDA classified KPV as a Category 2 bulk drug substance, citing “significant safety concerns” including lack of sufficient safety data for human use, effectively prohibiting its use in compounded medications.⁶ ⁷ ⁸ As of January 2026, KPV has never been FDA-approved for any indication, is not approved in any surveyed country, remains unavailable through legitimate pharmaceutical channels, and is marketed only through unregulated “research chemical” suppliers and cash-based clinics despite having zero human safety or efficacy data.⁶ ⁷

$155.00

- +

Description:

KPV is a naturally occurring tripeptide with the amino acid sequence Lysine-Proline-Valine (Lys-Pro-Val) and molecular weight of 369 Da.¹ ² ³ The peptide represents the three C-terminal amino acids of α-melanocyte-stimulating hormone (α-MSH), a 13-amino acid neuropeptide hormone (Ac-Ser-Tyr-Ser-Met-Glu-His-Phe-Arg-Trp-Gly-Lys-Pro-Val-NH₂) that exerts potent anti-inflammatory effects when administered systemically or locally.¹ ² α-MSH is derived from the precursor proopiomelanocortin (POMC) through post-translational proteolytic processing.¹ ²

**Historical Context:**¹ ²

α-MSH has been recognized since the late 1980s as having potent anti-inflammatory properties extending beyond its classical role in melanogenesis (skin pigmentation).¹ ² Research by Luger, Lipton, and colleagues demonstrated that α-MSH could suppress inflammation in diverse experimental models including contact dermatitis, inflammatory bowel disease, arthritis, asthma, and uveitis.¹ ² However, clinical development of α-MSH as an anti-inflammatory agent faced limitations due to its melanotropic effects—binding to melanocortin receptors (particularly MC-1R) on melanocytes causes increased melanin production and skin darkening, an undesirable cosmetic side effect for chronic systemic use.¹ ²

Research by Hiltz and Lipton in 1989 first demonstrated that the C-terminal tripeptide fragment KPV retained anti-inflammatory activity comparable to or even exceeding full-length α-MSH.² ⁴ Subsequent studies confirmed that KPV possesses “a similar or even more pronounced anti-inflammatory activity as full-length α-MSH” while eliminating melanotropic effects.² This discovery opened the possibility of developing KPV as a targeted anti-inflammatory agent without cosmetic complications.

**Unique Mechanism—PepT1 Transporter, Not Melanocortin Receptors:**³

Unlike α-MSH, which mediates anti-inflammatory effects primarily through melanocortin receptor (MC-1R, MC-3R) activation and cyclic AMP elevation, KPV operates via an entirely distinct mechanism:³ ⁴

Does Not Bind Melanocortin Receptors: Multiple studies confirmed that KPV does not bind to MC-1R, MC-3R, or MC-5R, does not increase intracellular cyclic AMP levels, and does not stimulate melanocytes.² ³ ⁴ This explains why KPV lacks pigmentary effects despite retaining anti-inflammatory properties.

PepT1-Mediated Cellular Uptake: The landmark 2008 study by Dalmasso et al. at Emory University definitively established that KPV’s anti-inflammatory mechanism depends on cellular uptake via the PepT1 transporter (SLC15A1), an H⁺-coupled oligopeptide transporter that normally transports dietary di- and tripeptides in the small intestine.³

Strategic Expression Pattern: PepT1 is normally expressed on the apical (luminal) membrane of small intestinal enterocytes but is virtually absent from normal colon.³ Critically, PepT1 is dramatically upregulated in inflamed colonic epithelial cells during inflammatory bowel disease (IBD), providing a disease-specific delivery mechanism.³ Additionally, immune cells including macrophages and T lymphocytes (Jurkat cells) express functional PepT1, allowing KPV to target both epithelial and immune components of intestinal inflammation.³

High Affinity Transport: PepT1 transports KPV with remarkably high affinity (Km ~160 μM in Caco2-BBE intestinal epithelial cells, Km ~700 μM in Jurkat T cells)—among the lowest Km values reported for PepT1 substrates.³ For comparison, the commonly used PepT1 substrate Gly-Sar has Km ≥1 mM.³ This high affinity allows efficient cellular uptake at low nanomolar concentrations.

Intracellular Anti-Inflammatory Action: Once transported into cells, KPV accumulates intracellularly and directly inhibits inflammatory signaling cascades including NF-κB and MAPK pathways, reducing pro-inflammatory cytokine production (IL-8, IL-1β, IL-6, TNF-α).³ The peptide does not act at the cell surface but must enter cells to exert effects.³

Pharmacological Characteristics:

Very Short Systemic Half-Life: As a small unmodified tripeptide, KPV is rapidly degraded by peptidases in serum and tissues, resulting in extremely short systemic half-life (likely minutes).² ³ This necessitates either frequent dosing, local administration, or advanced delivery systems.

Oral Bioavailability via PepT1: Unlike most peptides that are rapidly degraded in the gastrointestinal tract, KPV can be absorbed orally via PepT1 expressed on small intestinal enterocytes.³ Preclinical studies successfully demonstrated efficacy using oral administration (added to drinking water), with KPV reaching and acting on inflamed colonic tissues.³

Local Action at Inflamed Sites: KPV’s dependence on PepT1—upregulated specifically in inflamed tissues—provides inherent targeting to sites of active inflammation while sparing normal tissues.³ This tissue-selective mechanism theoretically minimizes systemic side effects.

Nanoparticle Delivery Enhancement: Advanced drug delivery systems using hyaluronic acid-functionalized nanoparticles encapsulated in pH-sensitive hydrogels achieved 12,000-fold enhancement in anti-inflammatory potency compared to free KPV solution, allowing therapeutic effects at nanomolar concentrations.⁵

Routes of Administration: Preclinical studies employed oral (drinking water), intraperitoneal injection, topical (cream/ointment), and advanced nanoparticle formulations.² ³ ⁵

Regulatory and Legal Status:

**FDA Category 2 Classification (September 2023):**⁶ ⁷ ⁸

The FDA placed KPV on the Category 2 Bulk Drug Substances List, designating it as a substance that “raises significant safety concerns.”⁶ ⁷ The agency specifically cited “lack of sufficient safety data for human use” among the grounds for this classification.⁶ ⁷ ⁸ This Category 2 designation effectively prohibits compounding pharmacies from using KPV in compounded medications under FDA regulations.⁶ ⁷ ⁸

**Zero Human Clinical Trials:**⁶ ⁷

“There have been no human trials on KPV to date.”⁶ Despite over 15 years of preclinical research, KPV has never been evaluated in human subjects for safety or efficacy in any condition.⁶ ⁷

**Never FDA-Approved:**⁶ ⁷

KPV is not an FDA-approved drug and is not approved in any country surveyed.⁶ ⁷ It has never undergone the regulatory process required for pharmaceutical approval.

**Current Legal Status:**⁶ ⁷

KPV is not a DEA scheduled substance, so possession is not illegal.⁶ However, the FDA Category 2 designation prohibits its use in compounded medications.⁶ ⁷ ⁸ The peptide remains legally sold as “research chemicals” or “dietary supplements”—classifications not subject to FDA regulations for pharmaceutical quality, safety, and efficacy.⁶ ⁷ This creates a gray-area legal status where cash-based medical clinics offer KPV treatment despite zero human safety data and explicit FDA safety concerns.⁶ ⁷


How It Works

 

PepT1-Mediated Cellular Uptake and Intracellular Action

KPV’s anti-inflammatory mechanism fundamentally differs from its parent molecule α-MSH and represents a novel approach to targeting inflammation.³

Step 1: PepT1 Transport into Cells³

KPV is transported across cell membranes via the PepT1 transporter (SLC15A1), an H⁺-coupled oligopeptide cotransporter that simultaneously transports peptides and protons.³ PepT1 normally functions in dietary peptide absorption in the small intestine but is dramatically upregulated in inflamed colonic epithelial cells during IBD and expressed by immune cells (macrophages, T lymphocytes) infiltrating inflamed tissues.³

Competitive Inhibition Studies: When Glycine-Leucine (Gly-Leu, a standard PepT1 substrate) was added to cells, it completely blocked KPV’s anti-inflammatory effects by competing for PepT1 transport.³ This definitively proved KPV requires cellular uptake via PepT1 to function.³

Cell Type Specificity: In HT29-Cl.19A human colonic cells that do not express PepT1, KPV had no anti-inflammatory effect despite these cells expressing functional IL-1β receptors and showing robust inflammatory responses to IL-1β.³ However, when HT29-Cl.19A cells were transfected with PepT1, KPV regained anti-inflammatory activity.³ This genetic evidence confirmed PepT1 as essential for KPV’s mechanism.³

High-Affinity Transport: Kinetic uptake experiments using radiolab

eled [³H]KPV demonstrated PepT1-mediated transport with Km ~160 μM in intestinal epithelial cells and ~700 μM in T cells—among the highest affinities (lowest Km values) reported for PepT1 substrates.³

Step 2: Intracellular Accumulation and NF-κB Inhibition³

Once inside cells, KPV accumulates intracellularly and directly inhibits the NF-κB (nuclear factor-kappa B) signaling pathway, a master regulator of inflammatory gene expression:³

IκB-α Degradation Inhibition: In intestinal epithelial cells (Caco2-BBE) stimulated with IL-1β, KPV significantly reduced degradation of IκB-α (the inhibitory protein that sequesters NF-κB in the cytoplasm).³ IL-1β normally causes rapid IκB-α degradation within 20 minutes, allowing NF-κB to translocate to the nucleus.³ In the presence of KPV, IκB-α degradation was reduced, and baseline IκB-α levels recovered much faster (90 minutes vs. 180 minutes), indicating that “KPV delays NF-κB activation and also shortened the delay of IκB-α recovery.”³

IκB-α Phosphorylation Blockade: KPV prevented phosphorylation of IκB-α at 45 minutes post-IL-1β stimulation, further confirming suppression of the NF-κB activation cascade.³

NF-κB Transcriptional Activity: Using NF-κB-dependent luciferase reporter assays, KPV at 10 nM concentration significantly decreased IL-1β-induced NF-κB transcriptional activity by ~6-fold.³ This directly demonstrates that KPV suppresses NF-κB-driven gene expression.³

EMSA Confirmation: Electrophoretic mobility shift assays (EMSA) confirmed that KPV reduces NF-κB DNA binding activity.³

Step 3: MAPK Pathway Inhibition³

KPV strongly inhibits activation of mitogen-activated protein kinase (MAPK) inflammatory signaling pathways:³

IL-1β normally induces rapid phosphorylation (activation) of three major MAPK family members: ERK1/2, JNK, and p38.³ Co-treatment with KPV (10 nM) “strongly decreased IL-1β-induced MAPK phosphorylation and, therefore, their activation.”³ This multi-pathway inhibition contributes to KPV’s broad anti-inflammatory effects.³

Step 4: Pro-Inflammatory Cytokine Suppression³

By inhibiting NF-κB and MAPK pathways, KPV reduces transcription and secretion of pro-inflammatory cytokines:³

IL-8 Reduction (Intestinal Epithelial Cells): IL-1β induced ~200-fold increase in IL-8 mRNA in Caco2-BBE cells.³ KPV co-treatment reduced this increase by ~35%.³ IL-8 protein secretion into culture medium was similarly decreased.³ IL-8 is a key neutrophil chemoattractant, so reducing IL-8 decreases immune cell recruitment to inflamed tissues.³

Broad Cytokine Suppression (Multiple Cell Types): KPV reduces production of IL-1β, IL-6, IL-12, TNF-α, and IFN-γ across intestinal epithelial cells, macrophages, and T lymphocytes.² ³ Importantly, KPV did not reduce IL-10 (anti-inflammatory cytokine), suggesting selective suppression of pro-inflammatory mediators rather than global immunosuppression.³

Step 5: Immune Cell Function Modulation³

T Lymphocytes (Jurkat Cells): KPV reduced TNF-α-induced IκB-α degradation and IL-8 mRNA expression in human T cells, demonstrating effects on adaptive immune responses.³

Macrophages: KPV suppresses LPS-induced inflammatory responses in macrophages, reducing pro-inflammatory cytokine production.² ³

Neutrophil Recruitment: By reducing IL-8 and other chemokines, KPV inhibits neutrophil recruitment and transmigration across the intestinal epithelium, decreasing tissue damage caused by infiltrating immune cells.³

Additional Mechanisms

**IL-1β Receptor Antagonism:**¹ ²

A stereochemical analog of KPV (K(D)PT, where valine is replaced with threonine) corresponds to amino acids 193-195 of IL-1β.¹ ² During IL-1β degradation, this KPT loop is revealed on the protein surface and can interact with IL-1 receptor type I (IL1RI), exerting antagonistic activity and contributing to terminating IL-1β-mediated inflammation.² While KPV itself was not found to act as an IL-1β receptor antagonist in the Dalmasso study (it had no effect in cells lacking PepT1 despite functional IL-1β receptors),³ this mechanism may contribute to the effects of KPV analogs.

**Antimicrobial Activity:**² ⁹

Beyond anti-inflammatory effects, KPV and α-MSH demonstrate antimicrobial activity against pathogens including Staphylococcus aureus and Candida albicans.² ⁹ The candidacidal activity is believed to be mediated by increased cellular cyclic AMP, though this appears distinct from the melanocortin receptor-independent anti-inflammatory mechanism.² This dual anti-inflammatory and antimicrobial activity suggests KPV may reduce both inflammation and infection risk—an advantage over traditional immunosuppressive agents.²


Research Evidence

 

Preclinical Studies—Inflammatory Bowel Disease

Dalmasso G et al., 2008 – PepT1-Mediated Tripeptide KPV Uptake Reduces Intestinal Inflammation

Design: Comprehensive in vitro and in vivo study examining KPV’s anti-inflammatory mechanism and efficacy in experimental colitis models.³

Published: Gastroenterology, 2008 Jan;134(1):166-178. (Top-tier gastroenterology journal)³

**In Vitro Studies:**³

Intestinal Epithelial Cells (Caco2-BBE):

  • KPV (10 nM) significantly reduced IL-1β-induced NF-κB luciferase activity (~6-fold reduction)
  • Reduced IκB-α degradation and phosphorylation
  • Strongly decreased ERK1/2, JNK, and p38 MAPK phosphorylation
  • Reduced IL-8 mRNA expression by ~35% and IL-8 protein secretion

Mechanism Confirmation:

  • Gly-Leu (PepT1 substrate) completely reversed KPV’s anti-inflammatory effects, proving PepT1-dependence
  • KPV had no effect in HT29-Cl.19A cells lacking PepT1 despite functional IL-1β receptors
  • Transfecting HT29-Cl.19A cells with PepT1 restored KPV’s anti-inflammatory activity
  • α-MSH had no anti-inflammatory effect in these systems despite melanocortin receptor expression

PepT1 Transport Kinetics:

  • Km ~160 μM for KPV in Caco2-BBE cells (high affinity)
  • 100 μM KPV inhibited Gly-Sar uptake by ~45% vs. ~25% for Gly-Leu, indicating higher PepT1 affinity for KPV
  • Nanomolar concentrations of [³H]KPV efficiently transported by PepT1

T Lymphocytes (Jurkat Cells):

  • KPV reduced TNF-α-induced IκB-α degradation at 15 minutes
  • Reduced IL-8 mRNA expression by ~5-fold at 6 hours
  • Km ~700 μM for KPV transport in Jurkat cells
  • First demonstration that human T cells express functional PepT1 capable of transporting KPV

**In Vivo Studies—DSS-Induced Colitis (N=10 mice/group):**³

Model: Dextran sulfate sodium (DSS) 3% in drinking water for 8 days; KPV 100 μM added to drinking water

Results:

  • Body Weight: DSS caused characteristic weight loss starting day 4; KPV significantly reduced weight loss at day 8 vs. DSS alone
  • Myeloperoxidase (MPO) Activity: DSS-induced increase in MPO (neutrophil infiltration marker) was decreased by ~50% with KPV treatment
  • Histology: DSS caused cell wall damage, interstitial edema, and increased inflammatory cell infiltration in lamina propria; mice receiving DSS + KPV showed “markedly reduced intestinal inflammation”
  • Colon Weight and Length: KPV prevented DSS-induced increase in colon weight and decrease in colon length
  • Pro-Inflammatory Cytokines (RT-PCR):
    • IL-6 mRNA: significantly reduced by KPV (p<0.05)
    • IL-12 mRNA: significantly reduced by KPV (p<0.05)
    • IL-1β mRNA: reduced (trend)
    • IFN-γ mRNA: reduced (trend)
    • IL-10 mRNA: no change (anti-inflammatory cytokine unaffected)
  • KPV Alone: Had no effect on basal MPO levels or inflammatory parameters in normal mice, indicating KPV specifically targets inflammation without affecting homeostasis

**In Vivo Studies—TNBS-Induced Colitis (N=10 mice/group):**³

Model: Trinitrobenzene sulfonic acid (TNBS) 150 mg/kg colonic injection; KPV 100 μM in drinking water; assessment at 48 hours

Results:

  • Body Weight: KPV significantly reduced weight loss at days 1 and 2 vs. TNBS alone
  • MPO Activity: TNBS-induced MPO increase was inhibited by ~30% with KPV
  • Colon Length: KPV prevented TNBS-induced decrease in colon length
  • Pro-Inflammatory Cytokines (RT-PCR):
    • IL-1β mRNA: significantly reduced (p<0.05)
    • IL-6 mRNA: significantly reduced (p<0.05)
    • TNF-α mRNA: significantly reduced (p<0.05)
    • IFN-γ mRNA: significantly reduced (p<0.05)

**Significance:**³

This landmark study:³

  • Definitively established KPV’s mechanism: PepT1-mediated cellular uptake followed by intracellular NF-κB and MAPK inhibition
  • First report of KPV-mediated reduction of colitis: Demonstrated efficacy in two distinct experimental IBD models
  • Confirmed oral bioavailability: Showed that orally administered KPV can reach inflamed colon and exert therapeutic effects
  • Identified PepT1 as therapeutic target: Upregulation of colonic PepT1 during IBD provides disease-specific delivery mechanism
  • Published in top-tier journal: Gastroenterology is among the most prestigious journals in gastroenterology research

Preclinical Studies—Contact Dermatitis

Getting SJ et al., 2003 – Dissection of the anti-inflammatory effect of the core and C-terminal (KPV) alpha-melanocyte-stimulating hormone peptides

Design: Study comparing anti-inflammatory effects of full-length α-MSH vs. KPV in contact dermatitis model.⁴

Model: Oxazolone-induced contact hypersensitivity in mice

**Results:**⁴

  • Both α-MSH and KPV, administered intravenously or topically, suppressed contact dermatitis reactions
  • KPV produced anti-inflammatory effects comparable to or exceeding α-MSH
  • KPV induced hapten-specific tolerance when applied topically

**Mechanism Differentiation:**⁴

  • α-MSH’s effects were melanocortin receptor-dependent (particularly MC-1R)
  • KPV’s effects were melanocortin receptor-independent
  • KPV lacked melanotropic activity (no skin pigmentation changes)

Significance: Established that KPV retains full anti-inflammatory efficacy of α-MSH without melanocortin receptor binding or pigmentary side effects.⁴

Advanced Drug Delivery—Nanoparticle Systems

Xiao B et al., 2017 – Orally Targeted Delivery of Tripeptide KPV via Hyaluronic Acid-Functionalized Nanoparticles Efficiently Alleviates Ulcerative Colitis

Design: Development and evaluation of hyaluronic acid (HA)-functionalized KPV-loaded nanoparticles (HA-KPV-NPs) embedded in chitosan/alginate hydrogel for oral delivery to inflamed colon.⁵

**Rationale:**⁵

  • Free KPV requires high concentrations (micromolar) for therapeutic effect
  • Nanoparticle targeting to inflamed mucosa could dramatically enhance efficacy
  • HA binds to CD44 receptors overexpressed on inflamed colonic epithelium and immune cells
  • Hydrogel protects NPs during gastric/small intestinal transit and releases NPs at colonic pH

**Nanoparticle Characteristics:**⁵

  • Poly(lactic-co-glycolic acid) (PLGA) core loaded with KPV
  • Surface-functionalized with hyaluronic acid
  • Encapsulated in pH-sensitive chitosan/alginate hydrogel
  • Collapses at colonic pH (~7) to release HA-KPV-NPs

**In Vitro Results:**⁵

  • HA-KPV-NPs showed abundant internalization into colonic epithelial cells and macrophages
  • Penetrated deeply into inflamed tissue
  • Enhanced cellular uptake compared to non-functionalized KPV-NPs
  • Accelerated mucosal healing and alleviated inflammation in vitro

**In Vivo Results (DSS-Induced Colitis in Mice):**⁵

  • Efficacy comparison:
    • HA-KPV-NP/hydrogel system: highly effective at preventing mucosa damage
    • Non-functionalized KPV-NP/hydrogel: moderately effective
    • Free KPV solution: minimal effect at same total KPV dose
  • 12,000-fold enhancement: HA-KPV-NPs achieved therapeutic effects at KPV concentrations 12,000-fold lower than free KPV solution
  • TNF-α downregulation: Much stronger suppression with HA-KPV-NPs vs. KPV-NPs
  • Safety: HA-KPV-NPs were non-toxic and biocompatible with intestinal cells

Significance: Demonstrated that advanced drug delivery systems can dramatically enhance KPV’s therapeutic potential, achieving efficacy at nanomolar concentrations and providing proof-of-concept for clinical translation.⁵

Broader Preclinical Evidence

Luger TA & Brzoska T, 2007 – α-MSH related peptides: a new class of anti-inflammatory and immunomodulating drugs

Design: Comprehensive review of α-MSH and KPV preclinical evidence across multiple inflammatory disease models.²

**Animal Models Demonstrating KPV Efficacy:**²

  • Contact dermatitis: KPV applied IV or topically suppressed both sensitization and elicitation phases; induced hapten-specific tolerance
  • Inflammatory bowel disease: KPV significantly reduced DSS- and TNBS-induced colitis
  • Allergic asthma: α-MSH/KPV inhibited allergic airway inflammation; reduced IL-4 and IL-13 in bronchoalveolar lavage fluid
  • Arthritis: α-MSH/KPV attenuated adjuvant-induced experimental arthritis; similarly effective as prednisolone without weight loss
  • Uveitis: α-MSH/KPV suppressed experimental autoimmune uveitis and endotoxin-induced uveitis
  • Cutaneous vasculitis: Single injection suppressed LPS-induced Shwartzman reaction

**Mechanisms Across Models:**²

  • Suppression of NF-κB activation
  • Reduced expression of adhesion molecules (ICAM-1, P-selectin)
  • Decreased pro-inflammatory cytokines (IL-1β, IL-6, TNF-α, IFN-γ)
  • Increased anti-inflammatory IL-10 in some models
  • Inhibition of immune cell migration and recruitment
  • Induction of regulatory T cells (in some models)

**Safety Profile:**²

  • α-MSH and KPV were well tolerated in animal studies
  • No significant adverse effects reported across multiple models
  • KPV lacks melanotropic effects (no skin pigmentation) unlike α-MSH
  • Activation of NF-κB was “never fully suppressed, but mostly only reduced,” suggesting KPV modulates rather than completely blocks inflammatory pathways²
  • In absence of inflammation, KPV’s immunosuppressive effects were “usually weak or absent,” indicating selective action on active inflammation²

Significance: This comprehensive review established KPV as a broad-spectrum anti-inflammatory agent effective across diverse experimental models with favorable preclinical safety profile.²


Current Status & Considerations

 

Regulatory Status and Clinical Development

**FDA Category 2 Designation (September 2023):**⁶ ⁷ ⁸

The FDA placed KPV on the Category 2 Bulk Drug Substances List, designating it as raising “significant safety concerns.”⁶ ⁷ ⁸ The agency specifically cited “lack of sufficient safety data for human use” as grounds for this classification.⁶ ⁷ This designation effectively prohibits compounding pharmacies from using KPV in compounded medications under FDA regulations.⁶ ⁷ ⁸

**Zero Human Clinical Trials:**⁶ ⁷

Despite robust preclinical evidence dating back to the 1980s-1990s and mechanistic studies through 2017:

  • “There have been no human trials on KPV to date.”⁶
  • Zero Phase I safety trials in healthy volunteers
  • Zero Phase II efficacy trials in any patient population
  • Zero published case reports or case series
  • Complete absence of human safety data
  • Unknown human pharmacokinetics
  • Unknown dosing requirements for any human condition

**Never FDA-Approved:**⁶ ⁷

KPV has never been FDA-approved for any indication and is not approved in any country surveyed.⁶ ⁷ It has never undergone the regulatory approval process required for pharmaceutical drugs.

**Current Legal Status:**⁶ ⁷

KPV is not a DEA scheduled substance, so possession is not illegal.⁶ However, the FDA Category 2 designation prohibits its use in compounded medications.⁶ ⁷ ⁸ Despite this, KPV remains available through:⁶ ⁷

  • Unregulated “research chemical” suppliers
  • Cash-based medical clinics operating outside evidence-based medicine
  • Online vendors marketing as “dietary supplements” (not subject to FDA pharmaceutical regulations)

This creates a problematic legal gray zone where clinics offer KPV treatment despite zero human safety data and explicit FDA safety concerns.⁶ ⁷

Evidence Base Summary

**Preclinical Evidence:**² ³ ⁴ ⁵

The preclinical evidence base for KPV is substantial and scientifically rigorous:

Mechanism of Action:

  • Definitively established via genetic validation (PepT1 knockout/transfection studies)³
  • Confirmed across multiple cell types (intestinal epithelial cells, T lymphocytes, macrophages)³
  • Mechanistic pathway clearly delineated (PepT1 uptake → intracellular NF-κB/MAPK inhibition)³
  • Distinguished from parent molecule α-MSH (melanocortin receptor-independent)⁴

Animal Model Efficacy:

  • Demonstrated across 6+ distinct inflammatory disease models²
  • Multiple independent research groups (Emory University, University of Muenster, William Harvey Research Institute)² ³ ⁴
  • Published in top-tier peer-reviewed journals (Gastroenterology, Annals of Rheumatic Diseases, J Pharmacol Exp Ther)² ³ ⁴
  • Consistent findings using different routes of administration (oral, IV, topical)² ³ ⁴
  • Both prevention and treatment paradigms evaluated³

Advanced Delivery Systems:

  • Nanoparticle formulations achieved 12,000-fold potency enhancement⁵
  • Proof-of-concept for clinical translation⁵

**Human Evidence:**⁶ ⁷

**Total human data: Zero studies, zero subjects, zero safety information.**⁶ ⁷

This represents an extraordinary disconnect—robust preclinical development over 15+ years with zero translation to human testing.⁶ ⁷

Critical Evidence Gaps

**Complete Absence of Human Safety Data:**⁶ ⁷

  • Unknown human pharmacokinetics (absorption, distribution, metabolism, excretion)
  • Unknown human dosing requirements
  • Unknown acute toxicity profile in humans
  • Unknown allergic/hypersensitivity potential
  • Unknown long-term safety (chronic dosing effects)
  • Unknown effects on human immune function
  • Unknown drug-drug interactions
  • Unknown effects in special populations (elderly, pregnant/lactating, pediatric, immunocompromised)
  • Unknown systemic exposure with oral vs. topical administration

**Efficacy Questions:**⁶ ⁷

  • No evidence KPV’s impressive preclinical efficacy translates to human disease
  • Unknown whether human IBD responds similarly to mouse models
  • Unknown optimal dosing, frequency, duration for any human condition
  • Unknown which patient populations most likely to benefit
  • No biomarkers validated for monitoring treatment response

**Manufacturing and Quality:**⁶ ⁷

  • No pharmaceutical-grade manufacturing standards established
  • Products from “research chemical” suppliers lack quality control, purity testing, sterility verification
  • Unknown contamination/impurity risks with unregulated products
  • No standardized formulations developed

Theoretical Safety Considerations

**Based on Mechanism and Preclinical Data:**² ³

Favorable Theoretical Safety Profile:

  • Selective action: PepT1-mediated mechanism targets inflamed tissues (where PepT1 is upregulated) while sparing normal tissues³
  • Modulation vs. suppression: KPV reduces but does not completely block NF-κB activation, maintaining baseline immune function²
  • Context-dependent: In absence of inflammation, KPV shows minimal immunosuppressive effects²
  • No melanotropic effects: Unlike α-MSH, KPV does not affect skin pigmentation²
  • Antimicrobial activity: Dual anti-inflammatory and antimicrobial properties may reduce infection risk vs. traditional immunosuppressants²

**Preclinical Safety:**² ³

  • Well tolerated across multiple animal models
  • No significant adverse effects reported in published studies
  • Effective doses did not cause weight loss (unlike corticosteroids)²
  • No apparent organ toxicity in animal studies

Theoretical Concerns (Extrapolated from Mechanism):

Immunosuppression Risk: While KPV modulates rather than blocks inflammation,² chronic suppression of NF-κB and MAPK pathways could theoretically impair host defense against infections or tumor surveillance. However, no such effects were observed in animal studies.²

GI-Specific Concerns: High PepT1 expression in small intestine means KPV could affect normal small intestinal cells, not just inflamed colon.³ Effects on normal intestinal function unknown.

Peptide Immunogenicity: Chronic exposure to peptides can elicit antibody responses. KPV’s small size (tripeptide) may reduce immunogenicity risk, but human data are absent.

Manufacturing Impurities: Unregulated products may contain bacterial endotoxins, peptide degradation products, or synthesis byproducts with unknown toxicity.⁶ ⁷

Comparison to Other Investigational Peptides

  1. BPC-157:
  • BPC-157: 3 small human studies (N=30 total), all open-label, unpublished Phase I trial
  • KPV: Zero human studies; even less human data than BPC-157
  1. GHRP-6:
  • GHRP-6: Zero therapeutic human trials per FDA systematic review; diagnostic use only
  • KPV: Similar complete absence of therapeutic human data
  1. Sermorelin:
  • Sermorelin: 2 diagnostic human studies; previously FDA-approved
  • KPV: Zero human studies; never approved; more profound evidence gap
  1. ARA-290:
  • ARA-290: 3 published Phase 2 RCTs (N=110); FDA Orphan Drug designation; legitimate clinical development
  • KPV: Zero human trials; no regulatory support; represents opposite end of development spectrum

KPV’s Unique Position: Among investigational peptides, KPV has one of the most paradoxical profiles—excellent preclinical mechanistic and efficacy data but complete absence of human translation despite 15+ years since initial discovery.²

Latest News

Latest Research Updates

Curated news and publications on peptides, metabolic research, and cellular energy modulation.
KPV (Lys-Pro-Val)KPV (Lys-Pro-Val)
$155.00
- +