Anabolic Steroids: Uses, Side Effects, And Alternatives

Kommentare · 29 Ansichten

Anabolic Steroids: https://www.soundofrecovery.

Anabolic Steroids: Uses, Side Effects, And Alternatives


# Estrogen Receptor‑Positive Breast Cancer: A Comprehensive Overview

Breast cancer remains the most common malignancy among women worldwide. Roughly **70 % of all breast cancers** express estrogen receptors (ER), making them **estrogen‑receptor‑positive (ER+)** or hormone‑dependent tumors. These tumours rely on the growth‑promoting actions of estrogens and are therefore uniquely susceptible to therapies that alter estrogen signalling.

Below is a self‑contained review covering:

1. How ER+ breast cancers develop
2. The key cellular pathways that drive tumour growth
3. The spectrum of approved therapeutic agents (and their mechanisms)
4. Emerging treatments that may reshape the future of hormone therapy

---

## 1. Pathogenesis: From normal breast epithelium to ER‑positive cancer

| Step | What Happens | Key Mutations / Alterations |
|------|--------------|-----------------------------|
| **Normal** | Breast ductal cells express estrogen receptor α (ERα) and proliferate in response to circulating estrogens. | — |
| **Initiation** | DNA damage from oxidative stress, hormonal cycles, or carcinogens creates mutations in genes that regulate cell cycle and apoptosis. | • **TP53** loss → impaired DNA repair
• **PIK3CA** activating mutations → PI3K/AKT pathway activation
• **PTEN** loss → AKT hyperactivation |
| **Promotion** | Aberrant ER signaling, often due to increased estrogen exposure (e.g., early menarche, late menopause) or aromatase overexpression in adipose tissue. | • Upregulation of **ESR1**
• Increased local estrogen via **CYP19A1** (aromatase) |
| **Progression** | Acquisition of additional oncogenic mutations, chromosomal instability, and evasion of apoptosis. | • **TP53** loss or mutation → genomic instability
• Overexpression of anti‑apoptotic genes (**BCL2**, **MCL1**)
• Activation of growth factor pathways (EGFR, HER2) |
| **Metastasis** | Tumor cells disseminate through lymphatics and bloodstream; colonize lymph nodes, liver, bone. | • Upregulation of matrix‑degrading enzymes (**MMPs**, **ADAMTS**)
• Altered adhesion molecules (downregulation of E‑cadherin) |

---

## 2. Key Molecular Pathways in Breast Cancer Progression

| Pathway | Biological Function | Clinical Relevance | Potential Therapeutic Targets |
|---------|---------------------|--------------------|------------------------------|
| **ER/PR Signaling** | Estrogen- and progesterone-mediated transcription; cell proliferation, survival | Hormone‑responsive tumors (≈70% of breast cancers) | Aromatase inhibitors, selective estrogen receptor modulators (SERMs), ER degraders |
| **HER2/EGFR Family** | Receptor tyrosine kinase signaling → MAPK, PI3K/Akt pathways | HER2‑positive (~15–20%) | Trastuzumab, pertuzumab, lapatinib, neratinib |
| **PI3K/Akt/mTOR Pathway** | Cell growth, metabolism, survival; frequently mutated in breast cancer | PIK3CA mutations (~30%); PTEN loss | Alpelisib (PI3Kα inhibitor), everolimus (mTOR inhibitor) |
| **Ras/Raf/MEK/ERK Pathway** | Proliferation signaling cascade | KRAS/BRAF mutations rare in breast but targetable downstream | Trametinib, dabrafenib |
| **DNA Damage Response & Homologous Recombination** | BRCA1/2 defects; synthetic lethality with PARP inhibitors | BRCA-mutated or HRD tumors | Olaparib, talazoparib |

---

## 4. Translating Pathway Knowledge into Drug Discovery

### 4.1 Target Identification and Validation
- **Computational Filtering**: Combine pathway maps with omics data (RNA‑seq, proteomics) to shortlist genes/proteins that are overexpressed or mutated.
- **CRISPR/Cas9 Screens**: Systematically knock out genes in breast cancer cell lines to confirm essentiality for proliferation/survival.
- **Biomarker Correlation**: Ensure that the target’s activity correlates with clinical outcomes (e.g., poor prognosis, resistance).

### 4.2 Hit Discovery Strategies
| Strategy | Rationale |
|---|---|
| **Fragment‑based Screening** | Identify small chemical fragments binding to active sites; later merged into potent molecules. |
| **High‑Throughput Virtual Screening** | Use docking against the crystal structure of target (e.g., EGFR kinase domain) to prioritize compounds. |
| **Repurposing FDA‑Approved Drugs** | Screen libraries for off‑target activity on the new target; reduces time to clinical use. |
| **Phenotypic Assays in Cancer Cell Lines** | Detect functional inhibitors regardless of binding mode; may uncover novel mechanisms. |

- Example: Screening kinase inhibitor library against mutant EGFR shows compounds with nanomolar potency.

---

## 3. From Lead to Pre‑clinical Candidate

| Step | Goal | Typical Activities |
|------|------|--------------------|
| **Chemical Optimization** | Improve potency, selectivity, metabolic stability, and reduce toxicity | SAR studies, medicinal chemistry iterations, predictive ADME models |
| **In vitro ADMET Profiling** | Evaluate absorption (Caco‑2), metabolism (HepG2 microsomes), plasma protein binding | LC–MS/MS assays, in silico predictions |
| **Pharmacokinetic Studies** | Determine half‑life, clearance, volume of distribution | Rodent PK studies with IV and PO dosing |
| **In vivo Efficacy Models** | Confirm target engagement in disease models (e.g., tumor xenografts) | Tumor growth inhibition assays |
| **Toxicology Studies** | Acute and sub‑chronic toxicity in two species | Clinical pathology, histopathology |

---

## 3. Detailed Work‑Plan

| Week(s) | Activity & Deliverables | Responsible Person |
|---------|------------------------|--------------------|
| 1–2 | • Literature review on target biology and previous inhibitors.
• Selection of lead scaffolds (structure‑activity relationships).
• Draft proposal for synthesis plan. | Lead Chemist |
| 3–4 | • Design of synthetic route for selected scaffold.
• Procurement of reagents, kits, and instrumentation. | Synthetic Lead |
| 5–8 | **Synthesis Phase**:
- Step‑wise assembly of core structure (e.g., building block A).
- Functionalization at positions B & C via SN2 or Suzuki coupling.
- Purification by flash chromatography. | Synthesis Team |
| 9–10 | **Analytical Characterisation**:
- NMR (^1H, ^13C) to confirm structure.
- LC‑MS for mass confirmation.
- HRMS for exact mass.
- UV‑Vis & IR as supplementary data.
Compile full dataset. | Analytical Lead |
| 11–12 | **Quality Control**:
- Determine purity by HPLC (≥95%).
- Check solubility in DMSO and buffer.
- Store samples at −20 °C, protected from light. | QC Officer |
| 13 | **Documentation & Reporting**:
- Prepare final report summarising synthesis, characterization data, purity assessment.
- Upload to central repository; ensure all metadata (batch number, date, operator) are recorded. | Project Manager |

---

## 4. Contingency Planning

### 4.1 Failure Modes and Mitigations

| Potential Failure | Root Cause | Immediate Action | Long‑Term Fix |
|-------------------|------------|------------------|---------------|
| Low yield (<30 %) | Incomplete coupling, poor reagent quality | Verify stoichiometry, check reagent expiry dates, run small‑scale test reaction | Optimize coupling conditions (temperature, solvent), consider alternative coupling agents |
| Product impurity (>5 % by HPLC) | Side reactions (hydrolysis, racemization) | Adjust purification protocol, add ion exchange steps | Modify protecting groups, employ chiral chromatography if needed |
| Unexpected mass shift in MS | Incorrect formula or adduct formation | Re‑analyze with higher resolution MS, check solvent purity | Verify compound identity via NMR; consider isotopic labeling if necessary |
| Poor solubility in assay buffer | Inadequate salt form or pH | Prepare appropriate salt (e.g., HCl), adjust pH to 7.0–8.0 | Use co-solvents at ≤1 % DMSO; confirm solubility via visual inspection |

---

### 4. Summary

By systematically applying the provided nomenclature rules, https://www.soundofrecovery.org/rodgerb5438090 we can derive a concise SMILES representation for any target compound in this series, generate its InChI string, and verify that these identifiers are consistent with each other. The final step is to cross‑check these representations against experimental data such as HPLC retention times or NMR spectra to ensure the correct stereochemical configuration has been assigned. This workflow should enable rapid synthesis, characterization, and validation of all compounds in the series while minimizing errors arising from manual notation.
Kommentare