CC8 - Biotech Flashcards

(53 cards)

1
Q

What are the three main areas of application for biotechnology?

A

The main areas of application for biotechnology are agricultural, medical, and industrial.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Describe the fundamental difference between biopharmaceuticals (biologics) and pharmaceuticals.

A

Biopharmaceuticals are produced by cells and generally have a high molecular weight (100s of kDa). They are sensitive to manufacturing processes, mostly protein-based and comparatively complex, less stable, and potentially immunogenic. Pharmaceuticals are produced by chemical reactions, have a low molecular weight, undergo robust manufacturing, are more stable, and mostly non-immunogenic.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What are some examples of protein-based biotherapeutics?

A

Examples include monoclonal antibodies (and derivatives), hormones, growth factors, fusion proteins, enzymes, and cell-based drugs.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What are the key factors to consider when designing protein-based drugs?

A

Factors include mechanism of action, characteristics of good/bad biotherapeutics (efficacy, safety—toxicity, off-target effects—and immunogenicity), and design strategies.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Name three common recombinant protein expression systems used in biotechnology and list a key characteristic of each.

A

E. coli: simple, cheap, fast, no glycosylation. S. cerevisiae: eukaryotic, intracellular/secreted proteins, yeast-type glycosylation. CHO cells: mammalian, human-like glycosylation, biotech workhorse.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What is the role of glycosylation in biotherapeutics, particularly monoclonal antibodies?

A

Glycosylation (especially N-glycosylation) impacts stability, solubility, bioavailability, pharmacokinetics, and immunogenicity. In mAbs, N-glycans at CH2 domain influence ADCC and CDC.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Describe the basic structure of an IgG monoclonal antibody and the function of its main domains.

A

Fab domain (light and heavy chains with CDRs) binds antigen; Fc domain (CH2, CH3) binds Fc receptors, activates complement, and induces ADCC.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

How can monoclonal antibody functions be modulated through engineering?

A

Fc-mediated functions can be enhanced (↑ADCC for oncology) or disabled (for immune checkpoint blockade) via amino acid mutations or glycosylation modulation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What is an antibody-drug conjugate (ADC)?

A

A monoclonal antibody chemically linked to a cytotoxic drug, delivering the drug specifically to cancer cells, reducing off-target effects.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What are bispecific antibodies (bsAbs)?

A

Engineered antibodies recognising two different epitopes/antigens, enabling new mechanisms like tumour-immune cell engagement.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

How was insulin, the first biotech drug, produced?

A

Human insulin was produced by recombinant DNA technology in E. coli.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Why is glycosylation important for the therapeutic protein Erythropoietin (EPO)?

A

Glycosylation, especially sialylation, determines EPO’s serum half-life. Non-sialylated EPO is rapidly cleared by hepatocytes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What are nucleic acid-based drugs and what is their potential in treating disease?

A

Nucleic acid drugs treat disease via gene inhibition, replacement, or editing, using gene transfer or molecules like ASOs and siRNA.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What are some key considerations when evaluating if a biotherapeutic is “good” or “bad”?

A

Efficacy, safety (toxicity, off-target effects), and cost.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What is a display library in the context of biotechnology?

A

A large collection of molecules displayed on a system linking the binder to its gene, used to screen for desirable properties.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What types of molecules might be found in a display library?

A

Antibodies, nanobodies, artificial binders, nucleic acids, or small chemical compounds.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What are some common display technologies used in library screening?

A

mRNA display, phage display, and yeast display.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Describe the general process of screening a library.

A

Selection of binders to a target protein, elution and replication, multiple enrichment cycles, followed by binder gene isolation and production.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What are some methods used for selecting binders from a library?

A

Surface panning, pull-down techniques (e.g., mRNA-protein fusion), magnetic bead sorting, and flow sorting.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What is the core principle of directed evolution?

A

Introducing diversity and selecting for improved traits iteratively, leading to molecules with enhanced properties.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Name three methods for introducing diversity into a library for directed evolution.

A

Mutagenesis, error-prone PCR, and synthetic DNA library construction.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

How does error-prone PCR contribute to creating diversity in directed evolution?

A

By using low-fidelity polymerases to introduce random mutations during amplification.

23
Q

What is DNA shuffling and how is it used in directed evolution?

A

Recombination of gene fragments to create diverse, chimeric genes for improved functionality.

24
Q

How can the stringency of selection be increased during directed evolution?

A

Tougher conditions (e.g., weaker binding buffers, fewer beads, altered gating) favour higher-affinity binders.

25
What is de novo protein design?
Designing entirely new proteins with sequences and structures not found in nature.
26
What are the two main categories of de novo protein design methods?
Energy minimisation approaches and artificial intelligence-based approaches.
27
How are energy optimization-based approaches (like Rosetta) used in protein design?
Using physics-based energy functions to identify sequences with minimum energy for a desired structure.
28
How are artificial intelligence-based methods used in de novo protein design?
Neural networks predict sequences or generate novel structures, e.g., AlphaFold, ProteinMPNN, RFdiffusion.
29
Provide an example of how de novo protein design has been used to create a novel protein topology.
Rosetta was used to design Top7, a novel protein fold confirmed by X-ray crystallography.
30
How are display libraries, directed evolution, and de novo protein design integrated to create new biomolecules?
De novo libraries are screened by display tech, and best molecules are improved by directed evolution (e.g., Adalimumab).
31
Describe how protein design can be used for thermal stabilization of proteins.
Using tools like PROSS to analyse sequence alignments, mutate variable sites, and select stabilising mutations via energy scoring.
32
How can protein design be used in vaccine development, specifically for Virus-Like Particles (VLPs)?
Designing VLPs by assembling trimeric proteins into precise geometries, mimicking viral surfaces to elicit strong immunity.
33
What is template grafting in protein design for vaccine development?
Grafting an epitope onto a matching template protein backbone, followed by design optimisation for proper folding.
34
What is topology building in protein design, and why is it used?
Building new protein folds around challenging epitopes when grafting fails, often followed by yeast display and selection.
35
How is rational design combined with directed evolution in enzyme engineering?
Rational active site design is followed by random mutagenesis and shuffling to evolve enhanced catalytic function.
36
What are the four main stages of pharmacokinetics (ADME)?
Absorption, Distribution, Metabolism, and Excretion.
37
What are some challenges that biotherapeutics face in the human body?
Proteolysis, pH denaturation, crowded molecular environments, and localisation difficulties.
38
Why is subcutaneous (SC) administration often preferred over intravenous (IV) administration for biologics?
SC is more convenient, faster, cheaper, enables self-administration, and allows fixed dosing.
39
How can nanoparticles be used to improve the delivery of biologics?
Nanoparticles encapsulate biologics, enhancing stability, cell uptake, circulation time, and enabling targeted delivery.
40
Name four main types of nanoparticles used for drug delivery.
Lipid-based nanoparticles (LNPs), polymeric nanoparticles (PNPs), inorganic nanoparticles (e.g., gold), and carbon-based nanoparticles.
41
42
What are adeno-associated viruses (AAVs) and why are they used for gene delivery?
AAVs are small, non-pathogenic viruses with a simple structure that can package genetic material (recombinant AAVs or rAAVs). They are used for gene delivery due to their low toxicity, non-pathogenic nature, and ability to target specific tissues, although they require a helper virus for replication.
43
What are some challenges associated with using AAVs for gene therapy?
Challenges include the presence of pre-existing neutralizing antibodies (NAbs) that can block receptor binding, premature degradation in endosomes triggering immune responses, degradation via the proteasome, and the potential for innate immune responses triggered by dsRNA formation in the nucleus.
44
What are some strategies to improve the stability and longevity of protein-based drugs?
Strategies include mutagenesis to create more stable variants, PEGylation (attachment of polyethylene glycol) to protect against proteolysis and immune recognition and increase half-life, using excipients to prevent surface adsorption and aggregation, supercharging proteins to enhance stability, glycoengineering to modify glycosylation patterns for better activity and half-life, and Fc fusion to increase half-life through increased size and FcRn recycling.
45
What are CAR T cells and how do they work in cancer immunotherapy?
CAR T cells are T cells engineered to express a chimeric antigen receptor (CAR). This CAR has an antigen recognition site (e.g., for a cancer-specific antigen) and intracellular signaling domains that activate the T cell upon binding. CAR T cells re-enable the patient’s own immune system to target and kill cancer cells.
46
What are some limitations and challenges associated with CAR T cell therapy?
Limitations include the risk of cytokine release syndrome ("cytokine storm"), challenges in targeting solid tumor microenvironments, and the high cost of therapy.
47
What are some potential future directions for CAR T cell therapies?
Future directions include tuning the ratio of CD4+ and CD8+ T-cell populations to improve efficacy, developing CAR-NK (natural killer) cells and allogeneic ("off-the-shelf") CAR T cells to reduce cost and time, and advancing 3rd and 4th generation CAR protein designs to enhance functionality.
48
What are some key considerations for the regulation of biotherapeutics by agencies like the FDA and EMA?
Regulatory agencies like the FDA and EMA aim to ensure studies are conducted under applicable laws and regulations and to protect the rights, safety, and welfare of human research subjects. They evaluate clinical data to assess safety and efficacy before approving drugs for commercialization.
49
Briefly outline the 5-step drug development and approval process in the US.
The process includes: 1) Discovery and Development (identifying new drug candidates), 2) Preclinical Research (testing for safety and toxicity in the lab), 3) Clinical Research (testing in human clinical trials across multiple phases), 4) FDA Review (evaluation of clinical trial data to determine safety and efficacy), and 5) FDA post-market safety monitoring (ongoing surveillance after approval).
50
What are Good Laboratory Practices (GLP) and Good Manufacturing Practices (GMP) in the context of biopharmaceutical development?
GLP includes guidelines and principles for laboratory operations to ensure the quality and integrity of preclinical research data, covering aspects like personnel, premises, equipment, and procedures. GMP encompasses all aspects of making a drug to ensure its safety, quality, and consistency, including production, quality control, and packaging.
51
What is bioprocessing in the context of industrial-scale protein production?
Bioprocessing is any manufacturing process involving living cells to generate a product, such as biopharmaceuticals. It occurs in highly controlled environments and typically includes upstream (cell growth and cultivation), downstream (purification), and fill-finish stages.
52
Briefly describe upstream bioprocessing.
Upstream bioprocessing refers to the steps related to cell growth and cultivation before purification. It involves optimizing media, cell inoculation, and growth kinetics in bioreactors, often scaling up from small to large volumes.
53
Briefly describe downstream bioprocessing.
Downstream bioprocessing encompasses the steps required to purify and formulate the product from the cell broth after upstream processing. This typically involves clarification (removing cells and debris), isolation/capture (recovering the product, often using chromatography), purification (removing remaining impurities), and polishing/formulation (preparing the drug for clinical administration with excipients and sterilization).