An In-Depth Analysis and Disambiguation of the Term "P-11" in Pharmaceutical and Biomedical Contexts
Executive Summary
This report provides a comprehensive analysis of the term "P-11" as it appears in various pharmaceutical, biomedical, and regulatory contexts. The initial investigation reveals that "P-11" is not a singular, uniquely identifiable entity but rather a polysemous designator referring to at least four distinct and scientifically significant subjects. The primary objective of this document is to disambiguate these entities and deliver an exhaustive, expert-level review of each.
The principal subjects identified and analyzed herein are:
- A Pharmaceutical Pill Imprint: The code "P 11" is imprinted on a pink, round tablet identified as Levothyroxine Sodium 200 mcg, a widely prescribed synthetic thyroid hormone for the treatment of hypothyroidism.
- A Central Nervous System Protein: The protein p11 (also known as S100A10) is a critical intracellular adaptor protein that modulates serotonin receptor signaling and has been strongly implicated in the pathophysiology of major depressive disorder and the mechanism of action of antidepressant medications.
- A Biomimetic Therapeutic Peptide: Oligopeptide P11-4 is a rationally designed, self-assembling peptide used in advanced dentistry for the non-invasive regeneration of early enamel caries lesions through a process of guided biomimetic mineralization.
- An Investigational Multimodal Drug: The code Neu-P11 refers to Piromelatine, a novel drug candidate with a complex pharmacological profile, acting as an agonist at melatonin and specific serotonin receptors. It has been investigated for conditions including insomnia, Alzheimer's disease, and irritable bowel syndrome.
This report systematically deconstructs each of these subjects, providing detailed information on their identification, mechanism of action, clinical applications, safety profiles, and regulatory status. By synthesizing data from molecular biology, clinical pharmacology, materials science, and regulatory affairs, this analysis offers a definitive reference to clarify the ambiguity surrounding "P-11" and provides nuanced perspectives on the scientific and clinical importance of each entity.
Section 1: Disambiguation of the Term "P-11" in Pharmaceutical and Biomedical Contexts
1.1. Overview of Multiple Entities Identified as "P-11"
The query for "P-11" presents a significant challenge due to the term's application across disparate domains within the life sciences. The lack of a universal, non-ambiguous nomenclature system means that a single alphanumeric code can be used coincidentally for entirely unrelated products, biological molecules, and regulatory concepts. This siloed nature of terminology can pose a risk for accurate information retrieval and cross-disciplinary communication, as a researcher searching for "P-11" without specific context could retrieve dangerously misleading information, for instance, finding data on a central nervous system protein when seeking information on a thyroid medication. A systematic disambiguation is therefore the necessary first step for any meaningful analysis.
The entities identified in relation to the term "P-11" include:
- Pill Imprint "P 11": A physical marking on a pharmaceutical tablet used for visual identification. This specific imprint corresponds to a 200 mcg dose of Levothyroxine Sodium, a synthetic thyroid hormone manufactured by Accord Healthcare Inc..[1]
- Protein p11 (S100A10): A member of the S100 family of proteins. It functions as an intracellular adaptor protein that plays a crucial role in neuronal function, particularly in modulating the trafficking and signaling of serotonin receptors. It is a key area of research in the study of depression and antidepressant action.[3]
- Oligopeptide P11-4: A synthetic 11-amino-acid peptide designed for dental applications. It is a biomimetic material that self-assembles under specific pH conditions to form a scaffold that promotes the remineralization and regeneration of tooth enamel.[7]
- Investigational Drug Neu-P11 (Piromelatine): A novel, multimodal drug candidate under development. It acts on multiple neurotransmitter systems, primarily as an agonist at melatonin (MT1/MT2) and serotonin (5-HT1A/1D) receptors. It has been studied in clinical trials for a range of conditions, including insomnia, Alzheimer's disease, and irritable bowel syndrome (IBS).[10]
- Other Contextual Uses: For the purpose of exhaustive disambiguation, several other, less medically central uses of the term have been identified. These include "P11" as a SKU or identifier for a pamphlet from Alcoholics Anonymous titled "The AA Member: Medication and Other Drugs" [14]; "Part 11" as a common shorthand for Title 21 CFR Part 11, a U.S. Food and Drug Administration (FDA) regulation governing electronic records and signatures in the life sciences industry [15]; and an experimental compound listed in DrugBank as PANTOTHENYL-AMINOETHANOL-11-PIVALIC ACID.[18] These are noted for completeness but are not the focus of the subsequent in-depth analysis.
1.2. Categorization and Prioritization for Analysis
To provide a structured and clinically relevant report, the subsequent sections are prioritized to focus on the four entities with direct therapeutic or biological significance. They are categorized based on their nature and state of development:
- Commercial Pharmaceutical Product (Levothyroxine Sodium): A widely available, approved drug identified by the pill imprint.
- Biological Protein Target (p11/S100A10): A naturally occurring protein with significant implications for disease pathophysiology and drug action.
- Therapeutic Biomaterial (Oligopeptide P11-4): A novel therapeutic agent, marketed as a medical device/cosmetic, representing an emerging technology.
- Investigational Drug (Neu-P11/Piromelatine): A molecule in the clinical development pipeline, not yet approved for market.
This structure allows for a comprehensive examination of each major entity as a standalone subject, moving from established clinical practice to cutting-edge research and development.
Table 1: Summary of "P-11" Entities
| Term | Identification | Category | Primary Field of Relevance | Key Source(s) |
|---|
| P 11 | Levothyroxine Sodium 200 mcg tablet | Pharmaceutical Imprint Code | Endocrinology, Pharmacy | 1 |
| p11 | S100 calcium-binding protein A10 (S100A10) | Biological Protein | Neuroscience, Psychiatry | 3 |
| P11-4 | Oligopeptide P11-4 (Oligopeptide 104) | Synthetic Peptide / Biomaterial | Dentistry, Materials Science | 7 |
| Neu-P11 | Piromelatine | Investigational Drug | Clinical Pharmacology, Neurology | 10 |
| P11 | "The AA Member: Medication and Other Drugs" | Publication Identifier | Addiction Recovery | 14 |
| Part 11 | Title 21 CFR Part 11 | U.S. Federal Regulation | Regulatory Affairs, Compliance | 16 |
Section 2: Analysis of Pill Imprint "P 11": Levothyroxine Sodium 200 mcg
The most direct and common identification of "P 11" in a clinical context is the imprint code on a specific pharmaceutical tablet. This section provides a comprehensive analysis of this product, Levothyroxine Sodium 200 mcg.
2.1. Identification and Product Specifications
The pill bearing the imprint "P 11" is a prescription medication with the following characteristics [1]:
- Active Ingredient: Levothyroxine Sodium
- Strength: 200 micrograms (mcg), equivalent to 0.2 milligrams (mg)
- Physical Description: Pink, round tablet, with a diameter of 7.00 mm
- Manufacturer/Labeler: Accord Healthcare Inc.
- National Drug Code (NDC): 16729-0457
- Regulatory Status: Prescription only; not a controlled substance
- Pregnancy Category: A (Adequate and well-controlled studies have failed to demonstrate a risk to the fetus in the first trimester of pregnancy, and there is no evidence of risk in later trimesters)
- Drug Class: Thyroid drugs
Levothyroxine is a widely prescribed medication, and while the "P 11" imprint is specific to the Accord Healthcare product, the active ingredient is available under numerous well-known brand names, including Synthroid, Euthyrox, Tirosint, and Levoxyl.[1]
2.2. Comprehensive Pharmacological Profile
2.2.1. Mechanism of Action as a Synthetic T4 Hormone
Levothyroxine is a synthetic preparation of the levo-isomer of thyroxine (T4), a tetra-iodinated tyrosine derivative. It is chemically identical to the endogenous T4 hormone, which is the major hormone secreted by the thyroid gland.[20] Its primary function is to serve as a replacement therapy in individuals whose thyroid gland does not produce sufficient amounts of thyroid hormone (hypothyroidism).[22]
The mechanism of action proceeds through several key steps:
- Pro-hormone Function: Levothyroxine (T4) is largely considered a pro-hormone. While it has some intrinsic activity, its primary physiological effects are mediated by its active metabolite, triiodothyronine (T3).[21]
- Peripheral Conversion: After administration and absorption, T4 is transported to peripheral tissues, most notably the liver and kidneys. Here, it undergoes deiodination (the removal of an iodine atom) by deiodinase enzymes (types I and II) to form T3. Approximately 80% of circulating T3 is derived from this peripheral conversion of T4.[20] T3 is significantly more potent than T4, with a relative biological potency of approximately 4:1.[21]
- Genomic Action: T3 diffuses into the cell nucleus and binds to specific thyroid hormone receptors (THRA, THRB), which are nuclear proteins attached to DNA at sites known as thyroid hormone response elements (TREs). This binding of the hormone to its receptor activates the complex, which then regulates the transcription of a wide array of genes. This leads to the synthesis of new messenger RNA (mRNA) and subsequent production of various proteins that control cellular metabolism, growth, and development.[20]
- Negative Feedback: Circulating thyroid hormones exert negative feedback control on the hypothalamic-pituitary-thyroid axis. They inhibit the release of thyrotropin-releasing hormone (TRH) from the hypothalamus and, more significantly, inhibit the synthesis and secretion of thyroid-stimulating hormone (TSH) from the anterior pituitary gland. In hypothyroidism, low T4 levels lead to high TSH; replacement therapy with levothyroxine restores T4 levels and normalizes TSH.[20]
2.2.2. Pharmacokinetics: Absorption, Distribution, Metabolism, and Excretion (ADME)
The pharmacokinetic profile of levothyroxine is characterized by slow absorption, high protein binding, and a long half-life, which are critical considerations for its clinical use.
- Absorption: Oral absorption of levothyroxine is variable, ranging from 40% to 80% of an administered dose.[24] The primary site of absorption is the small intestine, specifically the jejunum and upper ileum.[24] Absorption is significantly influenced by the gastrointestinal environment. It is maximal under fasting conditions and can be substantially reduced by the presence of food, particularly foods high in dietary fiber, soy products, and coffee.[24] Gastric acidity is also crucial for dissolution and absorption; medications that reduce stomach acid, such as proton pump inhibitors (PPIs), can impair levothyroxine absorption and necessitate dose adjustments.[25] The time to reach maximum plasma concentration (Tmax) is typically 2 to 3 hours after oral administration.[25]
- Distribution: Levothyroxine has a relatively small volume of distribution, estimated at 11-15 liters, which is comparable to the body's extracellular fluid volume.[25] A key feature is its extensive binding to plasma proteins—greater than 99.9%. It binds with high affinity to thyroxine-binding globulin (TBG) and, to a lesser extent, to thyroxine-binding prealbumin (TBPA) and albumin. This high degree of protein binding sequesters the hormone in the bloodstream, creating a large circulating reservoir and contributing to its long half-life. Only the very small fraction of unbound, or "free," hormone is metabolically active and able to enter cells to exert its effects.[24]
- Metabolism: The primary metabolic pathway for T4 is sequential deiodination. The liver is the principal site of degradation for both T4 and T3. Approximately 80% of a daily T4 dose is deiodinated, yielding roughly equal amounts of the active T3 and an inactive metabolite, reverse T3 (rT3).[24] Thyroid hormones are also metabolized through conjugation with glucuronides and sulfates in the liver. These conjugated metabolites are excreted into the bile and pass into the intestine, where they can undergo enterohepatic recirculation, with some of the hormone being reabsorbed.[20]
- Excretion: Thyroid hormones are eliminated from the body primarily via the kidneys, with a smaller portion of conjugated hormone being eliminated in the feces. Approximately 20% of T4 is eliminated in the stool.[24] The elimination half-life (t½) of levothyroxine is long, approximately 6 to 7 days in euthyroid individuals. This half-life is extended to 9-10 days in hypothyroid patients and shortened to 3-4 days in hyperthyroid states.[24] The long half-life is a significant clinical advantage, as it allows for stable steady-state concentrations with once-daily dosing and means that an occasional missed dose has a minimal impact on overall thyroid status.
Table 2: Pharmacokinetic Parameters of Levothyroxine Sodium
| Pharmacokinetic Parameter | Value/Description | Clinical Implication | Source(s) |
|---|
| Bioavailability | 40% - 80% (oral) | Highly variable; absorption is increased by fasting and decreased by food and certain drugs, requiring strict administration guidelines. | 24 |
| Tmax (Time to Peak) | 2 - 3 hours | Food can delay Tmax. | 25 |
| Volume of Distribution (Vd) | 11 - 15 L | Limited distribution, primarily within the extracellular fluid volume. | 25 |
| Protein Binding | > 99.9% | Creates a large circulating reservoir of hormone; only the unbound fraction is active. Explains the long half-life. | 24 |
| Half-life (t½) | 6 - 7 days (euthyroid) | Allows for convenient once-daily dosing and maintains stable hormone levels. | 24 |
| Metabolism | Hepatic; primary pathway is deiodination to T3 (active) and rT3 (inactive). Also undergoes conjugation. | T4 acts as a pro-hormone for the more potent T3. | 24 |
| Excretion | Primarily renal; ~20% fecal | Urinary excretion decreases with age. | 24 |
2.3. Clinical Applications and Efficacy
Levothyroxine is a cornerstone therapy in endocrinology, indicated for conditions characterized by thyroid hormone deficiency or those requiring suppression of TSH.
2.3.1. Primary Indications
- Hypothyroidism: Levothyroxine is the standard of care for hormone replacement therapy in patients of all ages with primary (failure of the thyroid gland), secondary (failure of the pituitary gland), or tertiary (failure of the hypothalamus) hypothyroidism, whether congenital or acquired.[1] It effectively reverses the clinical and metabolic manifestations of hypothyroidism, such as fatigue, lethargy, cold intolerance, weight gain, constipation, and dry skin.[21]
- Pituitary TSH Suppression: It is used as an adjunctive therapy following surgery and/or radioiodine ablation in the management of well-differentiated (papillary or follicular) thyroid cancer. By administering supraphysiologic doses of levothyroxine, TSH levels are suppressed, which removes the primary growth stimulus for any residual TSH-dependent cancer cells, thereby reducing the risk of tumor recurrence.[1]
2.3.2. Limitations of Use
There are specific conditions for which levothyroxine is not indicated:
- It should not be used for the suppression of benign thyroid nodules or for the treatment of nontoxic diffuse goiter in patients who are iodine-sufficient, as there is no clear clinical benefit and the risk of inducing iatrogenic hyperthyroidism is significant.[23]
- It is not indicated for the treatment of transient hypothyroidism that can occur during the recovery phase of subacute thyroiditis.[23]
2.3.3. Dosing and Administration Guidelines
Effective and safe use of levothyroxine requires careful dosing and strict adherence to administration instructions:
- Administration: The drug should be administered as a single daily dose, consistently on an empty stomach, ideally 30 to 60 minutes before breakfast, with a full glass of water. This standardized approach maximizes and ensures consistent absorption.[28]
- Timing with Other Substances: It must be administered at least 4 hours before or after drugs known to interfere with its absorption, such as calcium or iron supplements and antacids.[28]
- Individualization of Dose: Dosing is highly individualized and depends on numerous factors, including the patient's age, body weight, cardiovascular health, concomitant medications, and the specific indication. The average full replacement dose for a healthy adult is approximately 1.6 mcg per kilogram of body weight per day.[29] Doses are typically started low and titrated upwards in elderly patients or those with underlying cardiac disease.
- Monitoring: The therapeutic effect is not immediate; it may take 4 to 6 weeks to reach a steady state and for the peak effect of a given dose to be observed. Dose adjustments are guided by clinical response and regular monitoring of serum TSH levels.[28]
2.4. Safety, Tolerability, and Risk Management
Levothyroxine has a narrow therapeutic index, meaning the difference between a therapeutic dose and a toxic dose is small. Therefore, careful monitoring is essential to avoid both under- and over-treatment.
2.4.1. Adverse Event Profile and Side Effects
The adverse effects of levothyroxine are almost exclusively the result of therapeutic overdosage and mimic the signs and symptoms of hyperthyroidism (an overactive thyroid).[27]
- Common Side Effects: These include heat intolerance, excessive sweating, headache, nervousness, irritability, anxiety, insomnia, tremors, increased appetite, weight loss, and diarrhea. Temporary hair loss may occur, particularly in children during the first few months of therapy.[23]
- Serious Side Effects: The most significant risks are cardiovascular. Overdosage can precipitate or exacerbate cardiac conditions, leading to palpitations, tachycardia (fast heart rate), arrhythmias (such as atrial fibrillation), angina pectoris, and myocardial infarction. Long-term supraphysiologic dosing can accelerate bone resorption, leading to decreased bone mineral density and an increased risk of osteoporosis and fractures, particularly in postmenopausal women.[21]
2.4.2. Contraindications and Disease Interactions
Levothyroxine is contraindicated in patients with the following conditions [32]:
- Untreated adrenal insufficiency
- Acute myocardial infarction
- Untreated thyrotoxicosis
Caution is warranted in patients with pre-existing cardiovascular disease, as the metabolic effects of levothyroxine can increase myocardial oxygen demand and stress the heart.[32] In patients with diabetes mellitus, initiation of thyroid hormone therapy may worsen glycemic control, requiring an increase in the dose of insulin or oral hypoglycemic agents.[31]
2.4.3. Clinically Significant Drug and Food Interactions
The efficacy of levothyroxine therapy is highly susceptible to a wide range of interactions that can alter its absorption or metabolism. The extensive list of interactions with common over-the-counter supplements, prescription drugs, and everyday foods underscores a critical aspect of patient management. Sub-optimal therapeutic outcomes, such as persistently elevated TSH despite dose increases, may frequently be a consequence of these interactions rather than true pharmacological resistance. This highlights that successful treatment is as dependent on thorough patient education and adherence to a strict administration regimen as it is on accurate prescribing. A patient who takes their tablet with their morning coffee, milk, or multivitamin containing iron and calcium will experience significantly reduced drug absorption, potentially leading to therapeutic failure or dose instability if their habits change.[26]
Table 3: Clinically Significant Interactions with Levothyroxine
| Interacting Substance/Class | Examples | Mechanism of Interaction | Clinical Recommendation | Source(s) |
|---|
| Decreased Absorption - Drugs | Antacids, Calcium Carbonate, Iron Supplements, Magnesium Salts | Formation of insoluble chelates in the GI tract, preventing absorption. | Separate administration by at least 4 hours. | 26 |
| Bile Acid Sequestrants | Cholestyramine, Colestipol | Bind T4 in the intestine, preventing absorption. | Separate administration by at least 4 hours. |
| Proton Pump Inhibitors (PPIs) | Omeprazole, Esomeprazole | Increase gastric pH, which reduces dissolution and absorption of T4 tablets. | Monitor TSH; may require dose increase. Separating doses may not be sufficient. |
| Orlistat, Sucralfate | | Interfere with absorption. | Separate administration by at least 4 hours. |
| Decreased Absorption - Foods | Coffee, Milk, Soy Products, Walnuts, High-Fiber Foods | Bind to T4 or interfere with absorption process. | Administer T4 30-60 minutes before food/beverages. | 26 |
| Altered Metabolism | Enzyme Inducers (e.g., CYP3A4) | Rifampin, Phenobarbital, Carbamazepine | Increase hepatic metabolism of T4, leading to increased clearance. | Monitor TSH; may require dose increase. |
| Altered T4 Effects | Anticoagulants | Warfarin | T4 increases the catabolism of vitamin K-dependent clotting factors, potentiating the anticoagulant effect. | Monitor INR closely; may require a reduction in warfarin dose. |
| Diabetes Medications | Insulin, Metformin | T4 may increase blood glucose levels. | Monitor glycemic control; may require an increase in diabetes medication dose. |
2.5. Regulatory and Market Status
The regulatory history of levothyroxine in the United States is a notable case study in the FDA's evolving approach to ensuring the quality of legacy drugs. This journey from an unregulated product to a highly standardized one serves as a significant precedent for how the agency addresses long-standing medications that predate modern approval processes, demonstrating a prioritization of public health and product quality over historical market status.
- Pre-Regulatory Era: Although introduced in the 1950s, levothyroxine was marketed for decades as an unapproved drug, predating the modern NDA framework.[36]
- Quality and Stability Issues: This lack of oversight led to significant product quality problems. Levothyroxine is inherently unstable and degrades with exposure to light, moisture, and certain excipients. Between 1990 and 1997, there were 10 product recalls involving 150 lots and over 100 million tablets due to issues with sub-potency, lack of content uniformity, and instability.[36]
- FDA Intervention: In response to these public health concerns, the FDA took a decisive regulatory action. On August 14, 1997, the agency declared all levothyroxine sodium tablets to be "new drugs." This move legally required all manufacturers wishing to continue marketing their products to submit a formal NDA, thereby subjecting them to modern standards of safety, efficacy, and manufacturing quality.[36]
- Modern Approval and Standardization: The first NDA for a levothyroxine product was approved in August 2000. Since then, multiple manufacturers have obtained NDA approval, including for Thyro-Tabs in October 2002.[37] This process has successfully addressed the previous concerns, ensuring that all approved levothyroxine products on the market today meet rigorous standards for potency, stability, and bioavailability.[36] More recently, novel formulations such as oral solutions (Tirosint-SOL, approved in 2016, and Thyquidity, approved in 2020) have also received FDA approval, offering alternative administration options.[27]
Section 3: Analysis of Protein p11 (S100A10): A Key Modulator in Neuropsychiatry
Distinct from the pharmaceutical imprint, "p11" is the commonly used name for a protein, S100A10, that has emerged as a molecule of profound interest in neuroscience and psychiatry. Research into p11 is reshaping the understanding of mood disorders and the mechanisms of antidepressant therapies.
3.1. Molecular and Structural Characteristics
- Identification and Family: p11, also known as S100A10, is a small (10-12 kDa), acidic protein belonging to the S100 family of proteins. This family is the largest subfamily of EF-hand proteins, which are characterized by a specific helix-loop-helix structural motif involved in calcium binding.[3]
- Unique Structural Features: While p11 possesses the two EF-hand motifs characteristic of its family, it is unique in that mutations within both of these motifs render it incapable of binding calcium. This calcium insensitivity distinguishes its function from many other S100 proteins, whose activities are typically regulated by intracellular calcium fluctuations.[3] p11 typically exists as a symmetrical homodimer and readily forms a stable heterotetrameric complex with another protein, annexin A2.[4]
- Localization and Distribution: p11 is widely expressed throughout the body. Within cells, it is found predominantly in the cytosol and associated with the inner leaflet of the plasma membrane.[39] Its distribution within the central nervous system is particularly relevant to its role in mood regulation. It is expressed in several brain regions strongly implicated in the pathophysiology of depression, including the cerebral cortex, hippocampus, nucleus accumbens, and the raphe nuclei (a primary source of serotonin neurons).[6]
3.2. Functional Role and Mechanism of Action in the Central Nervous System
p11 functions as an inducible adaptor protein, meaning its expression can be regulated by various signals, and its primary role is to bring other proteins together and modulate their function and localization.
3.2.1. Modulation of Serotonergic Receptor Trafficking and Signaling
The most well-characterized function of p11 in the brain is its critical role in the serotonin (5-hydroxytryptamine, 5-HT) system.
- Receptor Interaction: p11 directly interacts with the intracellular loops of specific G-protein coupled serotonin receptors, most notably the 5-HT1B and 5-HT4 receptors. This interaction is highly specific among the S100 protein family.[3]
- Mechanism of Action: The core function of this interaction is to regulate the trafficking of these receptors to the neuronal cell surface. By forming a complex with the receptors, p11 facilitates their transport to and insertion into the plasma membrane. This increases the number of functional receptors available at the synapse, thereby amplifying the cell's response to serotonin. This potentiation of serotonergic signaling is a key mechanism by which p11 influences mood and behavior.[3]
3.2.2. Interaction with Ion Channels and Other Cellular Partners
Beyond the serotonin system, p11 interacts with a diverse array of cellular proteins:
- Ion Channels: p11 has been shown to modulate the function and cell surface expression of several types of ion channels, including the sodium channel Nav1.8 and potassium channels like TASK-1, which are involved in regulating neuronal excitability and pain signaling (nociception).[4]
- Gene Transcription: The p11/annexin A2 heterotetramer can bind to a chromatin-remodeling factor known as SMARCA3. This interaction suggests a role for p11 in the nucleus, where it can influence gene transcription. This pathway has been specifically implicated in the process of adult hippocampal neurogenesis, which is thought to be a key component of the therapeutic effects of antidepressants.[6]
3.2.3. Regulation of p11 Expression
p11 is not a static protein; its expression levels are dynamically regulated by a variety of stimuli, which is central to its role in both disease and treatment.
- Antidepressant Treatments: A wide range of antidepressant therapies, including selective serotonin reuptake inhibitors (SSRIs), tricyclic antidepressants (TCAs), monoamine oxidase inhibitors (MAOIs), and electroconvulsive therapy (ECT), have been shown to consistently upregulate the expression of p11 in the cortex and hippocampus of rodents.[3]
- Neurotrophic Factors: The expression of p11 is regulated by brain-derived neurotrophic factor (BDNF), a key molecule in neuronal survival and plasticity. This finding provides a mechanistic link between the neurotrophic hypothesis of depression and the monoamine system.[3]
- Stress Hormones: Glucocorticoids, such as dexamethasone (a synthetic analog of cortisol), also induce p11 expression.[4] This finding presents an interesting paradox, as stress is a primary risk factor for depression, yet the acute stress hormone response appears to increase levels of a protein that is otherwise reduced in the depressive state. This suggests a complex, possibly biphasic, role for p11 in the stress response. The acute increase may be an adaptive, compensatory mechanism designed to buffer against the effects of stress, which may fail or become dysregulated under chronic stress, leading to the down-regulation seen in depression.
3.3. Clinical Relevance in Major Depressive Disorder (MDD)
The convergence of evidence from human post-mortem studies, animal models, and pharmacological interventions strongly implicates p11 as a key player in the biology of depression.
3.3.1. Evidence for p11 Dysregulation in Depression
- Human Studies: Post-mortem studies of brain tissue from individuals who suffered from major depression have consistently found that levels of p11 mRNA and protein are significantly reduced in key brain regions like the anterior cingulate cortex and nucleus accumbens compared to non-depressed controls.[3]
- Animal Models: This finding is recapitulated in rodent models of depression, where animals exhibiting depression-like behaviors also show lower brain levels of p11.[41] Causal evidence comes from genetic manipulation studies. Mice that are genetically engineered to lack the p11 gene (p11 knockout mice) display a robust depression-like phenotype, including behaviors such as increased immobility in stress tests and reduced preference for rewarding stimuli. Conversely, transgenic mice that overexpress p11 exhibit behaviors akin to animals treated with antidepressants.[3]
3.3.2. Role in Mediating Antidepressant Therapeutic Response
The function of p11 appears to be essential for the efficacy of current antidepressant treatments. This has led to a significant evolution in the understanding of how these drugs work, moving beyond a simple focus on neurotransmitter levels in the synapse to the intracellular machinery that governs the brain's long-term response.
- A Convergent Mechanism: The fact that multiple, mechanistically distinct classes of antidepressants all lead to an increase in p11 expression suggests that p11 upregulation is a common, convergent downstream pathway for their therapeutic action.[41]
- A Necessary Mediator: The delayed therapeutic onset of most antidepressants (2-4 weeks) has long suggested that their immediate effect of increasing synaptic serotonin is not the final therapeutic mechanism, but rather an initiating event that triggers downstream cellular adaptations. p11 appears to be a critical one of these adaptations. In p11 knockout mice, the behavioral effects of antidepressants are significantly blunted, indicating that the presence of p11 is necessary for these drugs to achieve their full effect.[3]
- Bridging Hypotheses: The role of p11 provides a powerful mechanistic bridge between the classic monoamine hypothesis of depression (which focuses on serotonin deficiency) and the more recent neurotrophic/plasticity hypotheses (which focus on molecules like BDNF and processes like neurogenesis). The causal chain appears to be that antidepressants increase synaptic serotonin, which over time leads to increased BDNF signaling, which in turn upregulates p11. Increased p11 then enhances serotonin receptor function and contributes to transcriptional changes that promote neuronal plasticity and neurogenesis, ultimately leading to the alleviation of depressive symptoms.[3]
3.4. p11 as a Novel Therapeutic Target: Potential and Challenges
The robust body of evidence linking p11 to both the cause of depression and the cure makes it a highly compelling target for the development of novel antidepressant medications.
- Therapeutic Potential: Current antidepressants primarily target monoamine transporters or receptors. Targeting a downstream mediator like p11 could offer a more direct approach to rectifying the core cellular deficits in depression. Such a strategy holds the promise of developing drugs that might be faster-acting and have a more favorable side-effect profile by avoiding the broad, system-wide effects of manipulating synaptic monoamine levels.[3]
- Developmental Challenges: Despite its potential, targeting p11 presents significant pharmacological challenges. As an intracellular adaptor protein that functions via protein-protein interactions, it is not a conventional "druggable" target like a receptor or an enzyme. Developing small molecules to modulate these interactions is notoriously difficult. Furthermore, its widespread expression and interaction with multiple partners raise potential concerns for off-target effects. Future therapeutic approaches may require innovative modalities, such as gene therapy to increase its expression in specific brain regions, or the development of molecules that stabilize its interaction with key partners like the 5-HT receptors.
Section 4: Analysis of Oligopeptide P11-4: A Biomimetic Approach to Dental Remineralization
A third distinct entity identified by the "P11" designator is Oligopeptide P11-4, a synthetic biomaterial at the forefront of non-invasive dentistry. This technology represents a paradigm shift in the management of early tooth decay, moving from passive, ion-based prevention strategies to an active, biologically-inspired approach of guided tissue regeneration.
4.1. Peptide Composition and Physicochemical Properties
- Identification: Oligopeptide P11-4 is a synthetic, rationally designed peptide. It is also known by its International Nomenclature of Cosmetic Ingredients (INCI) name, Oligopeptide 104.[7]
- Composition: It is an oligopeptide, meaning it is a short chain of amino acids. Specifically, it is composed of 11 naturally occurring amino acids: Glutamine (Gln), Arginine (Arg), Phenylalanine (Phe), Glutamic acid (Glu), and Tryptophan (Trp). The precise amino acid sequence is Acetyl-Gln-Gln-Arg-Phe-Glu-Trp-Glu-Phe-Glu-Gln-Gln-Amide, often abbreviated as Ac-QQRFEWEFEQQ-NH2.[7]
- Physicochemical Properties: The defining characteristic of P11-4 is its pH-dependent self-assembly. It is an α-peptide that exists as monomers (individual molecules) in aqueous solution at neutral or alkaline pH. However, upon exposure to a lower pH (an acidic environment), the monomers spontaneously self-assemble into higher-order structures, specifically β-sheet fibrils. This process results in the formation of a viscous, three-dimensional hydrogel-like biomatrix.[7] This matrix has a strong, specific affinity for hydroxyapatite, the mineral component of tooth enamel.[7]
4.2. Mechanism of Action: pH-Triggered Self-Assembly and Guided Enamel Regeneration
The mechanism of P11-4 is biomimetic, meaning it is designed to imitate a natural biological process—in this case, the role of the protein amelogenin in guiding the formation of enamel during tooth development.[8] This approach enables true lesion repair from within, rather than simply hardening the surface layer as traditional fluoride treatments do.
- Diffusion and Triggering: When a solution of monomeric P11-4 is applied to the surface of a tooth with an early, non-cavitated carious lesion (often called a "white spot lesion"), its low viscosity allows the small peptide monomers to diffuse through the relatively intact surface layer into the porous, demineralized subsurface body of the lesion.[7] The environment inside an active carious lesion is acidic due to the metabolic byproducts of cariogenic bacteria. This low pH acts as the environmental trigger, causing the P11-4 monomers to rapidly self-assemble into the fibrillar scaffold
in situ.[7]
- Scaffold Formation and Nucleation: The newly formed 3D peptide matrix mimics the function of the natural enamel matrix. The specific arrangement of charged amino acids on the peptide fibrils creates binding sites for calcium ions that are spaced in a way that matches the crystal lattice of hydroxyapatite. This scaffold acts as a template, or nucleator, attracting calcium and phosphate ions from the patient's saliva.[7]
- Guided Mineralization: By organizing these ions into a crystalline structure, the scaffold guides the formation of new hydroxyapatite crystals de novo throughout the body of the lesion. This process effectively remineralizes the tooth from the inside out, rebuilding the lost mineral content and restoring the enamel's integrity.[9]
- Dentin Interaction: In addition to its effects on enamel, P11-4 has also been shown to interact with type I collagen, the primary organic component of dentin. This interaction may improve the stability of the bond between restorative materials and caries-affected dentin and increase the collagen's resistance to enzymatic degradation.[9]
4.3. Clinical Applications and Evidence Base
The unique mechanism of P11-4 lends itself to several applications in preventive and minimally invasive dentistry.
4.3.1. Treatment of Incipient Caries and White Spot Lesions
This is the primary and most well-studied application. By promoting guided enamel regeneration, P11-4 can arrest the progression of early caries and reverse the demineralization process before a physical cavity forms. This is particularly valuable for treating white spot lesions, which are common adverse effects of fixed orthodontic treatment.[7] The goal is to avoid or significantly delay the need for traditional "drill and fill" restorative procedures.
4.3.2. Management of Dentin Hypersensitivity
Dentin hypersensitivity is often caused by open dentinal tubules that transmit external stimuli to the dental pulp. By forming a matrix on the tooth surface, P11-4 can physically occlude these open tubules, creating a barrier and reducing or eliminating the associated pain.[7]
4.3.3. Acid Protection
When formulated as a gel for home use, the peptide matrix can form a stable, protective layer on the enamel surface, acting as a barrier to protect the teeth from dietary or bacterial acid attacks.[7]
4.3.4. Summary of Key Clinical Trial Outcomes
The clinical efficacy of P11-4 is supported by a growing body of evidence from in vitro studies and clinical trials. However, some conflicting results suggest that its effectiveness may be context-dependent, and it may perform best when used in conjunction with fluoride.
Table 4: Summary of Clinical Trials for Oligopeptide P11-4
| Study Type/Identifier | Patient Population | Intervention(s) | Comparator | Key Outcomes | Source(s) |
|---|
| Clinical Safety Trial (Brunton et al., 2013) | 15 healthy adults with buccal white spot lesions | Single application of P11-4 | No control group | Significant decrease in lesion size and shift towards remineralization over 6 months; no adverse events reported. | 9 |
| Randomized Controlled Trial (Alkilzy et al., 2018) | Children with active, non-cavitated occlusal caries | P11-4 + Fluoride Varnish | Fluoride Varnish alone | P11-4 group showed statistically significant improvement in all outcomes (lesion regression, caries inactivation) at 3 and 6 months. | 50 |
| Randomized Controlled Trial (2020) | 90 children/adolescents with non-cavitated occlusal caries | Group 1: P11-4 + Fluoride Varnish. Group 2: P11-4 + weekly P11-4 home gel. | Fluoride Varnish alone | Both P11-4 groups showed superior results (decreased laser fluorescence, caries inactivation) compared to fluoride alone. | 51 |
| Systematic Review & Meta-Analysis (2023) | Patients with initial caries lesions | P11-4 | Randomized parallel group (control/placebo) | Concluded P11-4 is a "promising treatment" that successfully arrests non-cavitated lesions and decreases lesion size. | 48 |
| Ongoing RCT (NCT06749977) | 18-30 year olds with post-orthodontic white spot lesions | P11-4 | Fluoride Varnish, No Treatment | Primary endpoint is change in lesion size/mineralization measured by Quantitative Light-induced Fluorescence (QLF). | 49 |
| Contrasting Studies | Bovine teeth, primary teeth | P11-4 | Various (fluoride, CCP-ACPF) | Some studies reported no significant increase in remineralization or surface microhardness in deciduous or bovine teeth. | 9 |
4.4. Commercial Products and Regulatory Landscape
- Development and Commercialization: The P11-4 technology was developed and patented by The University of Leeds in the UK. The Swiss company Credentis AG has licensed the technology and markets it globally under various trade names, including CUROLOX®, REGENAMEL®, and EMOFLUOR®.[7]
- Regulatory Strategy and Product Availability: The commercialization of P11-4 employs a sophisticated dual regulatory strategy that allows for broad market access. This approach involves marketing different formulations for different indications under distinct regulatory classifications.
- Medical Devices: For therapeutic applications, such as the treatment of initial caries, products like CURODONT™ REPAIR are marketed as medical devices for professional use by dentists. This pathway requires more rigorous clinical evidence and regulatory clearance (e.g., a 510(k) premarket notification in the U.S., which is the likely pathway for a Class II device like a cavity varnish).[7]
- Cosmetic Products: For applications such as acid protection and management of sensitivity, products like CURODONT™ PROTECT and CURODONT™ D'SENZ are marketed as cosmetic products, which are available over-the-counter to the public. This pathway has a lower regulatory burden, allowing for faster market entry.[7]
This dual strategy allows the manufacturer to establish a market presence and generate revenue with consumer-facing products while simultaneously pursuing the more demanding regulatory pathway for products with stronger, evidence-backed therapeutic claims.
Section 5: Analysis of Investigational Drug Neu-P11 (Piromelatine): A Multimodal Neurological Agent
The fourth significant entity is Neu-P11, the internal development code for an investigational drug named Piromelatine. This molecule is a new chemical entity with a complex pharmacological profile designed to act on multiple neurological pathways simultaneously.
5.1. Chemical Identity and Development History
- Identification: Neu-P11 is the code name for Piromelatine, a novel, synthetically derived small molecule.[12]
- Originator: The drug was developed and is being investigated by Neurim Pharmaceuticals.[12]
5.2. Multimodal Pharmacological Profile
Piromelatine is characterized as a "multimodal" agent because it is designed to engage several different receptor targets. This polypharmacology approach is a deliberate strategy to address complex diseases that involve multiple pathological pathways. The development of Piromelatine for a condition as multi-faceted as Alzheimer's disease exemplifies this strategy. Instead of a "magic bullet" approach, the drug's profile is designed to synergistically address different symptom clusters of the disease, such as cognitive decline, mood disturbances, and circadian rhythm disruption.
Table 5: Pharmacological Targets of Neu-P11 (Piromelatine)
| Receptor Target | Action | Associated Physiological/Therapeutic Effect | Source(s) |
|---|
| Melatonin Receptors (MT1/MT2) | Agonist | Regulation of circadian rhythms, sleep promotion, neuroprotection. | 11 |
| Serotonin Receptor 5-HT1A | Agonist | Anxiolytic, antidepressant, and mood-regulating effects. | 10 |
| Serotonin Receptor 5-HT1D | Agonist | Neuromodulation, potential role in vasoconstriction. | 10 |
| Serotonin Receptor 5-HT2B | Antagonist | May modulate certain cardiovascular and gastrointestinal functions. | 10 |
5.3. Preclinical and Clinical Development Program
Piromelatine has been evaluated in preclinical models and human clinical trials for a range of central nervous system (CNS) and gastrointestinal disorders.
5.3.1. Investigational Indications
- Alzheimer's Disease: This appears to be the lead indication, with the drug advancing to Phase II/III clinical trials.[13] Preclinical studies in a rat model of Alzheimer's demonstrated that Piromelatine improved memory performance and cognitive impairment.[12] The therapeutic hypothesis is likely that its combined effects on sleep regulation (via melatonin agonism) and mood/cognition (via serotonin agonism) can holistically address the complex symptomatology of the disease.
- Insomnia: A Phase II trial in 120 adults, with results announced in 2013, found that Piromelatine (20 mg and 50 mg doses) significantly improved sleep maintenance over a 4-week period compared to placebo.[12] Earlier Phase I studies had also shown a safe, dose-dependent improvement in sleep.[12]
- Irritable Bowel Syndrome (IBS): The investigation of Piromelatine for both CNS and gut disorders highlights the growing scientific and clinical focus on the "gut-brain axis." A Phase II clinical trial (NCT01558284) was initiated to evaluate the drug's effect on symptoms in patients with diarrhea-predominant IBS (D-IBS).[10] The rationale for this is grounded in the crucial role that both serotonin and melatonin play in regulating gut motility, visceral sensation (pain), and mood, all of which are dysregulated in IBS.[43] Preclinical studies in mice confirmed that Neu-P11 has antinociceptive (pain-reducing) effects in models of visceral pain.[57]
- Other Potential Indications: Preclinical research in rodent models has suggested potential antidepressant, anxiolytic, and antihypertensive effects, though these have not been pursued as primary indications in later-stage clinical trials.[12]
5.3.2. Review of Clinical Trial Data
- NCT01558284 (D-IBS): This was a 40-patient, randomized, double-blind, placebo-controlled study designed to assess symptomatic relief over a 4-week period.[10] The pharmacological rationale was explicitly stated as targeting melatonin and serotonin receptors to modulate gut function.[56] The public record for this trial was last updated in 2014, and the results are not available in the provided documentation, suggesting this development path may have been deprioritized.
- Insomnia Phase II: The trial was reported as positive, demonstrating efficacy in improving sleep.[12]
- Safety Profile: Phase I studies established an initial safety profile and demonstrated dose-dependent effects on sleep, allowing for progression to Phase II trials.[12]
5.4. Future Outlook and Development Trajectory
The development of Piromelatine appears to be most actively focused on Alzheimer's disease, where it is in the most advanced stage of clinical testing (Phase II/III).[13] Its multimodal mechanism of action is both its greatest potential strength and a significant development challenge. While it offers the possibility of treating complex syndromes with a single molecule, it also complicates the process of understanding the contribution of each target to the overall effect and de-risking potential off-target liabilities. The future success of Piromelatine will depend on the outcomes of its late-stage trials in Alzheimer's disease and the ability to demonstrate a clear clinical benefit in this notoriously difficult-to-treat patient population.
Section 6: Synthesis and Concluding Remarks
6.1. Comparative Summary of "P-11" Entities
This report has systematically deconstructed the ambiguity of the term "P-11," revealing four distinct entities of medical and scientific importance. A synthesis of the findings underscores their profound differences:
- Levothyroxine Sodium (Pill Imprint "P 11") is a well-established, FDA-approved small molecule drug that serves as the standard of care for hypothyroidism. Its mechanism, pharmacokinetics, and clinical profile are thoroughly understood, with management challenges primarily relating to its narrow therapeutic index and numerous interactions that require careful patient education.
- Protein p11 (S100A10) is an endogenous biological protein, not a drug itself, but a critical therapeutic target. It is an intracellular modulator of neuronal signaling, and its dysregulation is causally linked to major depression. It represents a focal point for the next generation of neuropsychiatric drug discovery, moving beyond the synapse to the cell's internal machinery.
- Oligopeptide P11-4 is a synthetic biomaterial, a therapeutic peptide that functions based on its physical properties of self-assembly. Marketed as a medical device and cosmetic, it represents a novel, non-invasive approach to tissue regeneration in dentistry, aiming to rebuild rather than simply protect or replace damaged tooth structure.
- Neu-P11 (Piromelatine) is an investigational drug, a new chemical entity in the clinical development pipeline. It is defined by its multimodal pharmacology, intentionally targeting multiple receptor systems to treat complex, multi-symptom disorders like Alzheimer's disease. Its future remains contingent on the results of ongoing late-stage clinical trials.
6.2. Expert Insights and Recommendations for Further Inquiry
The multifaceted nature of "P-11" offers a unique cross-section of the modern life sciences landscape, illustrating key trends in regulatory science, basic research, regenerative medicine, and clinical development. The journey of levothyroxine from an unapproved legacy drug to a highly regulated product class exemplifies the maturation of regulatory oversight to ensure patient safety. The study of the p11 protein illuminates the shift in neuroscience from broad neurotransmitter-based theories to a more nuanced understanding of the specific intracellular networks that govern brain function and dysfunction. The innovation of Oligopeptide P11-4 showcases the power of biomimetic design in creating materials that can guide and promote the body's own regenerative processes. Finally, the development strategy for Neu-P11 highlights the industry's move towards polypharmacology as a sophisticated approach to tackling complex, multifactorial diseases.
For professionals seeking further information, it is imperative to use specific, context-rich search terms to avoid the ambiguity that prompted this report. Recommended search queries include:
- For the thyroid medication: "Levothyroxine 200 mcg Accord Healthcare" or "pill imprint P 11".
- For the protein: "p11 protein S100A10 depression" or "p11 serotonin receptor trafficking".
- For the dental peptide: "Oligopeptide P11-4 caries regeneration" or "CURODONT REPAIR clinical trial".
- For the investigational drug: "Piromelatine Alzheimer's trial" or "Neu-P11 pharmacology".
By employing such precise language, researchers and clinicians can navigate the siloed terminologies of the life sciences to access accurate, relevant, and actionable information.
Works cited
- P 11 Pill Pink Round 7mm - Pill Identifier - Drugs.com, accessed September 14, 2025, https://www.drugs.com/imprints/p-11-31153.html
- P 11 Pill Images - Pill Identifier - Drugs.com, accessed September 14, 2025, https://www.drugs.com/imprints.php?imprint=p+11
- A role for p11 in the antidepressant action of brain-derived neurotrophic factor - PMC, accessed September 14, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2929288/
- p11 (S100A10)--an inducible adaptor protein that modulates neuronal functions - PubMed, accessed September 14, 2025, https://pubmed.ncbi.nlm.nih.gov/17085073/
- S100A10 - Wikipedia, accessed September 14, 2025, https://en.wikipedia.org/wiki/S100A10
- p11 and its role in depression and therapeutic responses to ..., accessed September 14, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3933996/
- Oligopeptide P11-4 - Wikipedia, accessed September 14, 2025, https://en.wikipedia.org/wiki/Oligopeptide_P11-4
- Self-Assembling Peptide P11-4 as a New Approach in Biomimetic Enamel Remineralization: A Review | RGUHS Journal of Dental Sciences | Journalgrid, accessed September 14, 2025, https://journalgrid.com/view/article/rjds/12433437
- Effectiveness of Self-Assembling Peptide (P11-4) in Dental Hard Tissue Conditions: A Comprehensive Review - MDPI, accessed September 14, 2025, https://www.mdpi.com/2073-4360/14/4/792
- Study Details | The Effect of Neu-P11 on Symptoms in Patients With D-IBS, accessed September 14, 2025, https://www.clinicaltrials.gov/study/NCT01558284
- Neuroprotective mechanism of the novel melatonin derivative Neu-P11 in brain ischemia related models - PubMed, accessed September 14, 2025, https://pubmed.ncbi.nlm.nih.gov/26188145/
- Piromelatine - Wikipedia, accessed September 14, 2025, https://en.wikipedia.org/wiki/Piromelatine
- Piromelatine - Neurim Pharmaceuticals - AdisInsight, accessed September 14, 2025, https://adisinsight.springer.com/drugs/800032086
- P11: The AA Member: Medication and Other Drugs - Alcoholics Anonymous, Houston, accessed September 14, 2025, https://aahouston.org/product/p11-the-aa-member-medication-and-other-drugs/
- 42 CFR Part 11 -- Clinical Trials Registration and Results Information Submission - eCFR, accessed September 14, 2025, https://www.ecfr.gov/current/title-42/chapter-I/subchapter-A/part-11
- Title 21 CFR Part 11 - Wikipedia, accessed September 14, 2025, https://en.wikipedia.org/wiki/Title_21_CFR_Part_11
- Part 11, Electronic Records; Electronic Signatures - Scope and Application - FDA, accessed September 14, 2025, https://www.fda.gov/regulatory-information/search-fda-guidance-documents/part-11-electronic-records-electronic-signatures-scope-and-application
- PANTOTHENYL-AMINOETHANOL-11-PIVALIC ACID: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed September 14, 2025, https://go.drugbank.com/drugs/DB08328
- Pill Identifier (Pill Finder Wizard) - Pill Identification Tool Using Drug Pictures, Color, Shape, Number, or Imprint - RxList, accessed September 14, 2025, https://www.rxlist.com/pill-identification-tool/article.htm
- levothyroxine - ClinPGx, accessed September 14, 2025, https://www.clinpgx.org/chemical/PA450221
- Levothyroxine: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed September 14, 2025, https://go.drugbank.com/drugs/DB00451
- What Does Levothyroxine Do? Its Mechanism of Action Explained, accessed September 14, 2025, https://www.goodrx.com/levothyroxine/what-does-levothyroxine-do
- Synthroid (Levothyroxine Sodium): Side Effects, Uses, Dosage, Interactions, Warnings, accessed September 14, 2025, https://www.rxlist.com/synthroid-drug.htm
- LEVOXYL® (levothyroxine sodium) Clinical Pharmacology | Pfizer Medical - US, accessed September 14, 2025, https://www.pfizermedical.com/levoxyl/clinical-pharmacology
- A Review of the Pharmacokinetics of Levothyroxine for the Treatment of Hypothyroidism - PMC - PubMed Central, accessed September 14, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6193522/
- Levothyroxine Interactions: Drugs, Diet, & More - GoodRx, accessed September 14, 2025, https://www.goodrx.com/conditions/hypothyroidism/is-my-thyroid-medication-working-these-drugs-can-cause-interactions
- Tirosint-SOL (levothyroxine sodium) FDA Approval History - Drugs.com, accessed September 14, 2025, https://www.drugs.com/history/tirosint-sol.html
- SYNTHROID® (levothyroxine sodium) tablets, for oral use - This label may not be the latest approved by FDA. For current labeling information, please visit https://www.fda.gov/drugsatfda, accessed September 14, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/021402s036lbl.pdf
- LEVO-T® (levothyroxine sodium) tablets, for oral use, accessed September 14, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/021342s023lbl.pdf
- Levothyroxine Side Effects You Should Know About - GoodRx, accessed September 14, 2025, https://www.goodrx.com/levothyroxine/levothyroxine-side-effects
- Levothyroxine: Hypothyroidism Medication - Cleveland Clinic, accessed September 14, 2025, https://my.clevelandclinic.org/health/drugs/19628-levothyroxine-tablets
- Levothyroxine (oral route) - Side effects & dosage - Mayo Clinic, accessed September 14, 2025, https://www.mayoclinic.org/drugs-supplements/levothyroxine-oral-route/description/drg-20072133
- Levothyroxine Interactions Checker - Drugs.com, accessed September 14, 2025, https://www.drugs.com/drug-interactions/levothyroxine.html
- Levothyroxine and Interactions: Other Drugs, Alcohol, and More - Healthline, accessed September 14, 2025, https://www.healthline.com/health/drugs/levothyroxine-interactions
- Levothyroxine Interactions with Food and Dietary Supplements–A Systematic Review - PMC, accessed September 14, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8002057/
- Bioavailability/Bioequivalence Studies in Evaluation of New Levothyroxine Products - FDA, accessed September 14, 2025, https://www.fda.gov/media/77811/download
- Drug Approval Package: Thyro-Tabs (Levothyroxine Sodium) NDA #021116, accessed September 14, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2002/21-116_Thyro-Tabs.cfm
- Thyquidity (levothyroxine sodium) FDA Approval History - Drugs.com, accessed September 14, 2025, https://www.drugs.com/history/thyquidity.html
- p11 (S100A10) — an inducible adaptor protein that modulates neuronal functions - USD Biology, accessed September 14, 2025, http://usdbiology.com/cliff/Courses/Advanced%20Seminars%20in%20Neuroendocrinology/Epigenetics%20of%20Depression/Svenningsson%20Greengard%2007%20CurrOpionPharm%20p11%20adaptor%20protien%20mods%20neuronal%20funct.pdf
- Role of p11 in Cellular and Behavioral Effects of 5-HT4 Receptor Stimulation, accessed September 14, 2025, https://www.jneurosci.org/content/29/6/1937
- Protein p11 and the Understanding of Depression - Fisher Center for Alzheimer's Research Foundation, accessed September 14, 2025, https://www.alzinfo.org/articles/p11-depression/
- Schematic Overview on How p11 Regulates the Membrane Levels and Functions of Distinct Ion Channels via Several Different Mechanisms. - ResearchGate, accessed September 14, 2025, https://www.researchgate.net/figure/Schematic-Overview-on-How-p11-Regulates-the-Membrane-Levels-and-Functions-of-Distinct-Ion_fig1_341393170
- ALTERATIONS IN EXPRESSION OF p11 and SERT IN MUCOSAL BIOPSIES OF PATIENTS WITH IRRITABLE BOWEL SYNDROME - PMC, accessed September 14, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2474784/
- P11 and G protein-coupled receptor function & pharmacology - Svenningsson lab, accessed September 14, 2025, https://www.svenningssonlab.com/p11-and-ggpcr-function-pharmacology/
- Oligopeptide P11-4 | α-Peptide - MedchemExpress.com, accessed September 14, 2025, https://www.medchemexpress.com/oligopeptide-p11-4.html
- self-assembling P11-4 peptide (TFA removed) - NovoPro Bioscience Inc., accessed September 14, 2025, https://www.novoprolabs.com/p/self-assembling-p11-4-319744.html
- Mode of action of P11-4 on dental caries. (A) Calcium hydroxyapatite... - ResearchGate, accessed September 14, 2025, https://www.researchgate.net/figure/Mode-of-action-of-P11-4-on-dental-caries-A-Calcium-hydroxyapatite-crystals-around-the_fig1_358764346
- Effect of Self-Assembling Peptide P11-4 on Arrest, Cavitation, and Progression of Initial Caries Lesions - CareQuest Institute for Oral Health, accessed September 14, 2025, https://www.carequest.org/resource-library/effect-self-assembling-peptide-p11-4-arrest-cavitation-and-progression-initial
- Study Details | NCT06749977 | P11-4 and Fluoride Varnish for Remineralization of Post-orthodontic Lesions | ClinicalTrials.gov, accessed September 14, 2025, https://clinicaltrials.gov/study/NCT06749977
- Biomimetic Enamel Regeneration Using Self-Assembling Peptide P 11 -4 - MDPI, accessed September 14, 2025, https://www.mdpi.com/2313-7673/8/3/290
- (PDF) Randomized Clinical Trial investigating Self-Assembling Peptide P11-4 for Treatment of Early Occlusal Caries - ResearchGate, accessed September 14, 2025, https://www.researchgate.net/publication/339745992_Randomized_Clinical_Trial_investigating_Self-Assembling_Peptide_P11-4_for_Treatment_of_Early_Occlusal_Caries
- June 23, 2023 Elevate Oral Care, LLC Steve ... - accessdata.fda.gov, accessed September 14, 2025, https://www.accessdata.fda.gov/cdrh_docs/pdf22/K222323.pdf
- Neuroprotective mechanism of the novel melatonin derivative Neu, accessed September 14, 2025, https://www.researchgate.net/publication/280135672_Neuroprotective_mechanism_of_the_novel_melatonin_derivative_Neu-P11_in_brain_ischemia_related_models
- Piromelatine (Neu-P11) | MT1/MT2 Receptor Agonist | MedChemExpress, accessed September 14, 2025, https://www.medchemexpress.com/piromelatine.html
- Piromelatine: Uses, Interactions, Mechanism of Action | DrugBank ..., accessed September 14, 2025, https://go.drugbank.com/drugs/DB12288
- Study Details | NCT01558284 | The Effect of Neu-P11 on Symptoms in Patients With D-IBS, accessed September 14, 2025, https://clinicaltrials.gov/study/NCT01558284
- Antinociceptive effects of novel melatonin receptor agonists in mouse models of abdominal pain - PMC - PubMed Central, accessed September 14, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3921511/
- Antinociceptive effects of novel melatonin receptor agonists in mouse models of abdominal pain - Baishideng Publishing Group, accessed September 14, 2025, https://www.wjgnet.com/1007-9327/abstract/v20/i5/1298.htm