1/6
Looks like no tags are added yet.
Name | Mastery | Learn | Test | Matching | Spaced |
|---|
No study sessions yet.
Essay 1 INTRODUCTION
Modern oncology drug discovery has moved decisively away from untargeted cytotoxic screening towards a rational, mechanism-driven paradigm informed by molecular disease biology.
This shift is particularly important in breast cancer, which is not a single disease but a collection of biologically distinct subtypes, including hormone receptor–positive, HER2-amplified, and triple-negative breast cancers.
Each subtype is underpinned by unique signalling dependencies and therefore requires tailored therapeutic strategies.
As a result, screening novel small-molecule inhibitors can no longer be reduced to simple measurements of cytotoxic potency.
Instead, it represents a multidimensional process aimed at predicting clinical efficacy, selectivity, and safety as early as possible.
An exceptional screening strategy must therefore integrate robust validation of molecular target engagement with early assessment of drug-like behaviour and toxicity risk.
PARAMETER 1: Biochemical Potency & Selectivity
The foundation of any targeted drug discovery programme is confirming that a compound directly engages its intended molecular target.
Quantifying biochemical potency through measurements such as the IC₅₀ or inhibition constant (Kᵢ) against purified recombinant proteins provides an essential baseline assessment of binding affinity and inhibitory capacity.
However, reliance on a single potency value is insufficient for high-quality decision-making.
Exceptional screening strategies place equal emphasis on selectivity profiling; off-target activity remains one of the leading causes of late-stage failure.
Broad kinase or enzyme profiling platforms ,including full kinome panels , enable the systematic identification of unintended interactions, allowing selectivity to be quantified and refined during lead optimisation.
Selectively inhibiting PI3Kα while avoiding PI3Kβ is crucial ;the two isoforms have fundamentally different biological roles.
PI3Kα is the main driver of oncogenic PI3K signalling in many tumours, so its inhibition delivers the intended anticancer effect. However,PI3Kβ is essential for normal insulin signalling and glucose homeostasis.
Inadvertent inhibition of PI3Kβ disrupts metabolic control and leads to toxicities such as insulin resistance. .These metabolic liabilities are a hallmark of non‑selective PI3K inhibitors and frequently limit their clinical utility.
Without this level of profiling, compounds may progress with concealed polypharmacology that later compromises cellular efficacy that initially appeared promising.
PARAMETER 2: Cellular Efficacy & Mechanism of Action
While biochemical potency is necessary, it does not guarantee biological relevance.
A compound must also demonstrate activity within intact cells, where factors such as membrane permeability, intracellular metabolism, and active efflux can profoundly influence efficacy.
Measuring cellular EC₅₀ values, in disease-relevant breast cancer cell lines, therefore represents a critical next step.
Exceptional screening extends beyond viability assays and requires direct confirmation of pathway modulation.
This is achieved by quantifying downstream pharmacodynamic biomarkers ,linked to the target pathway.
For inhibitors of the PI3K/mTOR axis, reduced phosphorylation of ribosomal protein S6 provides a robust indicator of pathway suppression, whereas inhibition of CDK4/6 activity can be confirmed through decreased phosphorylation of retinoblastoma protein.
The use of genetically defined or isogenic cell line models further strengthens mechanistic validation.
Demonstrating selective cytotoxicity of PARP inhibitors in BRCA-deficient cells provides strong evidence of synthetic lethality and mirrors contemporary precision-medicine approaches.
This strategy ensures that observed cellular effects are causally linked to on-target activity rather than nonspecific toxicity.
PARAMETER 3: Selectivity & Therapeutic Index
Beyond efficacy, early assessment of safety and therapeutic index is essential.
A compound’s clinical value depends on its ability to selectively target tumour cells while sparing normal tissues.
Many oncogenic pathways are also active in healthy proliferating cells, meaning that unintended toxicity can easily emerge.
Early screening against non-transformed human cell types therefore provides valuable insight into potential dose-limiting liabilities.
Advanced screening programmes increasingly incorporate physiologically relevant primary cells, such as human hepatocytes or induced pluripotent stem cell-derived cardiomyocytes.
High-content imaging platforms and co-culture systems allow more nuanced assessment of compound effects on cell morphology, survival, and the tumour microenvironment.
This integrated safety strategy reframes selectivity as a tissue-specific concept rather than a simple on/off phenomenon.
PARAMETER 4: ADME / Drug-Like Properties
Suboptimal pharmacokinetic behaviour remains a dominant cause of attrition in drug development.
Screening for metabolic stability in human liver microsomes provides early predictions of hepatic clearance.
Permeability assays such as Caco-2 monolayers offer insight into likely oral bioavailability.
Measurement of plasma protein binding further informs estimates of free, pharmacologically active drug concentration.
For advanced or metastatic disease, particularly involving brain metastases, early evaluation of blood–brain barrier penetration and susceptibility to efflux transporters such as P-glycoprotein is especially important.
Profiling interactions with cytochrome P450 enzymes is essential, as breast cancer patients frequently receive long-term combination therapies that rely on these metabolic pathways.
PART (b) TECHNIQUE: CETSA
CETSA is based on the principle that ligand binding typically stabilises a protein’s folded structure, thereby increasing its resistance to thermal denaturation.
A shift in the protein’s thermal stability profile provides direct evidence of compound binding.
The major strength of CETSA lies in its ability to confirm that a compound engages its intended target under realistic intracellular conditions.
CETSA inherently incorporates membrane permeability, intracellular stability, competition with endogenous ligands, and binding to the target in its native conformation and cellular environment.
Thermal stabilisation serves as an indirect marker of binding and does not necessarily equate to functional inhibition.
The assay often relies on high-quality antibodies, and mass spectrometry-based approaches increase technical complexity.
CETSA is therefore most effective when integrated into a broader screening framework.
CONCLUSION
Screening strategies must move beyond linear workflows and adopt an integrated, iterative approach that simultaneously evaluates efficacy, selectivity, safety, and drug-like behaviour.
Robust decision-making emerges from triangulating data across biochemical potency, cellular mechanism of action, therapeutic index, and ADME properties.
Future advances are likely to arise from the convergence of direct target engagement tools such as CETSA with increasingly sophisticated disease models, including three-dimensional organoids and microfluidic systems.