Linette Chilton
Linette Chilton

Linette Chilton

      |      

Subscribers

   About

Metandienone Wikipedia

Introduction



The compound under discussion has been extensively examined in both pre‑clinical and clinical settings over the past two decades. Its primary pharmacological action is as an inhibitor of the cysteine protease cathepsin L (and to a lesser extent, other lysosomal enzymes such as cathepsin B). The drug’s therapeutic potential spans several disease domains—including viral infections (notably SARS‑CoV‑2), various cancers, and neurodegenerative disorders—owing largely to its ability to modulate proteolytic pathways that are dysregulated in these conditions. Importantly, the compound has reached phase III trials for COVID‑19 and is available as a prescription medication in several countries under brand names such as "Kerapax" and "Serelex".



---




1. Key Preclinical Findings



Disease area Model & species Major outcome Dose regimen Relevance to human disease


SARS‑CoV‑2 In vitro Vero E6, A549 cells; in vivo hACE2 transgenic mice 50–100 µM inhibited viral replication by >90% at 24 h; improved survival in mice (70 % vs. 10 %) 20 mg/kg IP q12h Supports antiviral activity and therapeutic window


Alzheimer’s disease APP/PS1 transgenic mice, aged 6–8 mo 15 mg/kg orally daily for 4 wk → 30 % reduction in Aβ plaque burden; improved Y-maze performance 15 mg/kg PO q24h Suggests potential disease-modifying effect


Cancer (lung) H1299 NSCLC xenografts in nude mice 10 mg/kg IP thrice weekly → tumor volume reduced by 60 % vs. vehicle; no weight loss observed 10 mg/kg IP 3×/wk Indicates antitumor activity with tolerable toxicity


---




4. Toxicological Observations & Limitations



Study Dose & Duration Key Findings


Acute oral toxicity (rat) 2000 mg/kg PO single dose No mortality; slight transient GI irritation


Sub‑chronic (90 days, rat) 50–200 mg/kg PO daily Minor hepatotoxicity at >150 mg/kg (elevated ALT/AST); no renal toxicity


Reproductive toxicity (mice) 100 mg/kg PO twice weekly for 8 weeks No significant effects on fertility or litter size


Genotoxicity (Ames test) Up to 5 g/L in bacterial strains Negative – no mutagenic activity


These data indicate that the compound is relatively safe up to ~150 mg/kg/day orally, but higher doses may affect liver function.



---




3. Pharmacokinetic Profile (Based on In‑Vivo Studies)



Parameter Value (Rat; Oral)


Absorption Rapid; peak plasma concentration (Tmax) ~0.5 h


Bioavailability (F) ~45 % (due to first‑pass metabolism)


Half‑life (t½) 3–4 h (steady‑state achieved after 2–3 doses)


Clearance (CL) 0.9 L/h/kg


Volume of Distribution (Vd) 1.5 L/kg


Metabolism Hepatic; major metabolites are glucuronide conjugates


Excretion Primarily renal (~30 % unchanged)


---




3.2 Pharmacodynamic Profile



Parameter Value / Observation


Dose‑Response Curve Sigmoidal (Emax ≈ 80 % inhibition of target activity at high doses).


IC50 for Target Enzyme 0.35 µM in vitro.


Half‑Maximal Effective Concentration (EC50) ~1.2 µM in cell-based assay.


Therapeutic Index Approximately 15–20 based on preclinical toxicity data.


Mechanism of Action Competitive inhibitor binding to the active site; blocks substrate access.



Pharmacodynamic Summary






The drug’s potency is high (low IC50/EC50), indicating effective target engagement at low micromolar concentrations.


A relatively narrow therapeutic index requires careful dose titration and monitoring in clinical settings.


No evidence of significant off‑target activity was observed, but the potential for unforeseen interactions remains.







5. Comparative Assessment of the Two Drug Candidates



Feature Candidate A (Drug X) Candidate B (Drug Y)


Chemical Nature Small organic molecule (MW ≈ 350 Da). Peptidic/hybrid with cyclic constraints.


Physicochemical Properties Lipophilic, moderate aqueous solubility. Polar, limited membrane permeability.


Metabolic Stability Good in vitro; metabolic half‑life > 2 h. Susceptible to proteolytic enzymes; rapid clearance unless protected by cyclic structure.


Pharmacokinetics Oral bioavailability ≈ 50–60 %; plasma protein binding moderate. Requires parenteral administration or delivery strategies (e.g., liposomes).


Toxicity Profile No acute toxicity at therapeutic doses; mild hepatotoxicity in high‑dose chronic studies. Low systemic toxicity but potential local irritation if administered intramuscularly.


Regulatory Status In preclinical stage; no IND submitted yet. Preclinical data available, but formulation challenges need to be addressed before IND filing.


---




5. Conclusion & Recommendations




Therapeutic Potential


- Both compounds exhibit promising antiviral activities in vitro and favorable safety margins.

- The choice between them depends on the intended route of administration, pharmacokinetic requirements, and formulation feasibility.





Next Steps for Compound A


- Conduct PK/PD modeling to confirm dosing strategy.

- Explore formulation options (e.g., nanoparticle encapsulation) if oral bioavailability is suboptimal.

- Initiate toxicology studies in non‑rodent species (dog or rabbit) for cross‑species safety profiling.





Next Steps for Compound B


- Optimize the synthetic route to reduce cost and improve yield.

- Perform human PBPK simulations to predict plasma concentrations under various dosing regimens.

- Validate efficacy in a larger animal model (e.g., ferret or non‑human primate) that better mimics human disease.





Regulatory Pathway


- For both compounds, compile an Investigational New Drug (IND) dossier incorporating pharmacology, toxicology, chemistry, manufacturing, and controls (CMC).

- Engage with the regulatory agency early via pre‑IND meetings to confirm data requirements for Phase I trials.





Risk Mitigation


- Establish contingency plans for potential safety signals (e.g., hepatotoxicity).

- Implement pharmacovigilance protocols from the outset, including real‑time monitoring of adverse events during clinical studies.



---




Final Recommendations




Prioritize compound B for accelerated development based on its superior potency and favorable ADME profile.


Initiate a parallel in‑house synthesis program for both compounds to ensure rapid scale‑up.


Allocate resources toward generating the necessary preclinical safety data (GLP toxicology, pharmacokinetics).


Engage with regulatory agencies early to discuss the clinical trial design and potential expedited review pathways.



These actions will position our portfolio for a timely entry into Phase I clinical trials, capitalizing on the therapeutic opportunity presented by this novel target.

Gender: Female