Biologics 🧬 Flashcards

1
Q

Biologics (mAbs) formulation

A

• Biologics are either formulated as a liquid formulation or lyophilised powder (solid) (to be reconstituted before
Use)
• Administration is either subcutaneous, or i.v.
• Issues often linked to frequency of administration
• Adverse effects

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

Solid form advantages & disadvantages

A

• Dose and injection volume adjustable

• Can be developed as multi use formulations

• dual chambers allow precise volume & weight of protein

• Can be more expensive to couple a solid form to a delivery device (e.g., dual chambers)

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

Liquid form advantage & disadvantages

A

• More convenient to end user (precise concentration already prepared by company)

• Better patient compliance

• Better accuracy

• Chemical degradation hydrolysis thus less stable, limit shelf life, manipulation, etc..

• Physical stability more difficult to control aggregation (e.g., exposure to final fill finish operations) (step at which it could occur) (agglomeration has proteins are big flexible molecules that can lead to find conformational changes) Major obstacle as can start early on

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

mAbs conformation

A

Large molecules prone to conformational changes > may expose the hydrophobic acids buried inside

For mAbs and other therapeutic proteins in solution, the hydrophobic AA (amino acid) should be buried in the protein core and the hydrophilic AA should be in the shell. (Ensures stability but stress can disturb this)

Majority of all charged amino acids are on the surface of the mAb > makes antibody more hydrophilic

Majority of non polar & hydrophobic on the inside

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

mAbs stability

A

• mAbs (and other proteins) are not colloids
>Not a uniform distribution of the same charge
>Using DLVO theory (PHAR10100) is indicative at best.

• Need to consider attractive and repulsive forces but also solvation (ability to be in solution & contact in water)
> And consequently pH, buffer, salt (concentration and type) and concentration of co-solutes (to limit / maximise repulsion between the monoclonal antibodies). Consider PH of solution as want to increase solubility. mAbs isoelectric ph is 8 need it to be lower to PH 5-6 eg (not lower as it would irritate)

• Stress may lead to unfolding & ultimately to aggregation. (Hydrophobic parts exposed & will try to evade the contact with water)

• Aggregation not desirable

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

What stress can affect monoclonal antibodies?

A

Chemical degradation
• Oxidation, deamidation, hydrolysis
• May lead to instability then aggregation
>Exposure hydrophobic regions
>Exposure of cysteine residues of formation of disulfide bridges

Physical destabilisation
• Extreme pH (during formulation bio process)
• Shear forces (eg syringe & thin needle pressure but doesn’t happen when time administer just in earlier stages & an example to illustrate)
• Air-water interfaces (hydrophobic parts wants to get into contact with air)
• Adsorption to solid surfaces (wants to avoid contact with water) (eg wall of syringe)
• Freezing drying
• High pH or temperature changes

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

Formulation of proteins & mAbs

A

Most don’t need as small volume & conc & don’t need them stable for a long time eg not for infusions. Water only needed to reconstitute these but if need for a longer period of time:

Excipients
• Buffers
>Acetate, citrate, histidine or phosphate
>Formulations pH range often 5-6.5 (IgG pl approx. 8)
> Buffer concentration kept low to adapt to physiological pH upon administration

• Salt and tonicity modifiers
>Colloidal stability (attractive & repulsive forces. Changing salt amount in solution modulates charge of molecule & repulsion forces)
>i.v. injection requires isotonic preparation
>i.m. or s.c. injections may be able to handle hypertonic or hypotonic conditions
> Common excipient is NaCI, ArgI or sugars such as sorbitol. NaCl favours agglomeration however compared to the others. Sugars ensures solution is isosmotic

• Surface active agents (surfactants)
> mAbs flexible molecules with hydrophobic and hydrophilic regions
> Unfolding leads to aggregation
> Surfactants cover interfaces (air/liquid and solid liquid) thus limit unfolding
> Polysorbates 20 and 80 most common
> however polysorbates prone to degradation may contribute to aggregation & limit shelf life
> PS can under chemical degradation (oxidation & hydrolysis) as well as enzymatic degradation
> surfactants at concentrations above cmc (critical micelle concentration ) formation of micelles demonstrates that all interfaces have been covered

• Antioxidants
> Oxidation reaction catalysed by metals (may leach from surface of syringe)
> Use of EDTA to chelate metals to control oxidation
> Reducing agents such as glutathione can reverse oxidation

• Protein stabilizers
> Stabilisers are preferentially excluded (outside of the sphere of hydration of mAbs) : Lower interaction with protein but not hydrophobic (they are hydrophilic) leads to higher concentration of co-solute in bulk than in the solvation shell of the protein: increase the delta G unfolding > helps stabilise & important for higher volumes or lipholisation as these molecules pump the water molecules around and making the mAb more condensed. Requires far more energy to unfold the monoclonal body.
> Sugars e.g., sucrose
> Amino acids such as Arg (ArgHCI or ArgGlu) these provide both the isosmotic (shown previously) & protein stabiliser roles

• Lyophilisation development
> Use of PEG, sucrose, trehalose
> Same mechanism as stabilisers: exclusion by steric hindrance and maintained upon freezing
( when u lyophilise u pump out the water molecules which increases the concentration and the risk of unfolding increases as the encounter of molecules becomes more likely)

• Caveats to use of sugars as lyoprotectants
> Disaccharides susceptible to hydrolysis at low pH
> Hydrolysis of sucrose to glucose and fructose at pHs

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

Freezing mAbs (eg in plane in freezer)

A

• Low temperature extends shelf life

• But cold denaturation which happens when freezing sample (the sides & bottom & top of vile is where the cold comes from, water freezes first and all the rest of the molecules / ions concentrate at the centre of the vile) = more & more contact between mAb molecules

• Damage > results in change of pH, ionisation, solubility or H-bond energies.

• Repeated freezing and thawing cause aggregation by pH and concentration changes and by provision of nucleation points at ice water interface. Need to thaw & gently shake whole vile to ensure right concentration & dose to patient.

• Cryoprotection by sugars, polyhydric alcohols, AAs, work by preferential exclusion, lower cold denaturation and stabilise sample

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

Lyophilisation

A

Removal of water then adding it again before giving to patient. Removal of water preserves.

Loss of monomers happens as aggregates are created

• Lyophilised protein formulations have a greater long-term stability.

• These do undergo reversible conformational changes during the different steps of lyophilisation with render them prone to aggregation (and similarly again when reconstituted).

• Reactions and denaturation continue when lyophilised.

• Refrigerate lyophilised medicines to reduce aggregation rates

• Hygroscopic - sealed to avoid water vapour absorption.

• Advantage: allows us to modify concentration

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

mAbs mechanism of action

A

-Signalling pathway blocking: eg cetuximab / pantimab
Target: Receptors like EGFR (Epidermal Growth Factor Receptor) on cancer cells.(not necessarily on cell surface eg in RA adalinmumab binding to TNF alpha which is a soluble growth factor which moves in fluid & prevents it from binding to its receptor)
Mechanism: Prevents ligand binding to receptors.
Inhibits downstream signaling pathways (PI3K/AKT, RAS/ERK).
Outcome: Blocks cancer cell survival, proliferation, and growth.

  • Antibody-Dependent Cellular Cytotoxicity (ADCC): eg tafasitamab
    Target: Antigens like CD19 on cancer cells.
    Mechanism: Antibodies recruit NK cells by binding to Fc gamma RIII receptors.
    NK cells release cytotoxic molecules (granzyme and perforin). FC gamma RII receptor of natural killer cell bind to FC region of antibody and induces lysis of cell.
    Outcome: Cancer cells are lysed by NK cell-induced cytotoxicity.

-Complement-Dependent Cytotoxicity (CDC) : eg Naxitamab
Target: Antigens like GD2 on cancer cells.
Mechanism: Antibodies activate the complement system by recruiting C1q. (Compliment protein)
Induces formation of the Membrane Attack Complex (MAC).
Outcome: Cancer cells undergo lysis through complement-mediated action.

Antibody-Dependent Cellular Phagocytosis: (ADCP) eg tafasitamab
Target: Antigens like CD19 on cancer cells.
Mechanism: Similar to ADCC but now FC gamma receptor on macrophage recognises mAb FC receptor. Antibodies engage macrophages via FcγRI receptors. Macrophages engulf and digest cancer cells through phagocytosis.
Outcome: Cancer cells are destroyed by macrophages.

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

Antibodies & FcRn receptors

A

Antibodies bind to Fc receptors (specifically the neonatal Fc receptor, or FcRn) for recycling to extend their half-life and maintain their effectiveness in the body. Here’s why this process is crucial:

  1. Protection from Degradation
    Endocytosis: Antibodies, like IgG, are taken into cells through a process called endocytosis. Once inside, they enter acidic endosomes.
    Without FcRn binding, antibodies would be directed to lysosomes for degradation, leading to their breakdown and loss of function.
  2. FcRn-Mediated Recycling
    Binding in Acidic Environments: FcRn binds to the Fc region of antibodies in the acidic pH (around 6.0) of the endosome.
    Release at Neutral pH: After binding, FcRn protects the antibody from degradation and transports it back to the cell surface, where the neutral pH (7.4) causes the antibody to be released into circulation.
  3. Prolonged Circulation
    Recycling allows antibodies to avoid degradation and return to the bloodstream, significantly extending their half-life.
    IgG antibodies and albumin are examples of proteins whose longevity depends on FcRn recycling.
  4. Efficient Use of Resources
    Recycling reduces the need for constant production of new antibodies by the immune system.
    It ensures that functional antibodies are reused, optimizing immune defense mechanisms.
  5. Therapeutic Applications
    Monoclonal Antibodies (mAbs): Engineers often optimize the Fc region of therapeutic antibodies to enhance binding to FcRn, improving their stability and half-life.
    Drug Delivery: FcRn is used to extend the half-life of Fc-fusion proteins or antibody-drug conjugates.

In Simple Terms
Binding to Fc receptors like FcRn acts like a rescue system for antibodies. Instead of being destroyed inside cells, they are “rescued” and sent back into the bloodstream to continue their job of fighting pathogens. This process helps antibodies last longer and reduces the body’s burden of constantly making new ones.

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

Q: What is the signaling pathway blockade mechanism of mAbs?

A

mAbs bind to receptors or soluble ligands to block receptor activation and signaling.

Example: Adalimumab binds TNF-alpha, preventing receptor interaction.

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

Q: What is ADCC (Antibody-Dependent Cellular Cytotoxicity)?

A

Fc region of mAbs binds Fc gamma receptors on NK cells, triggering cell lysis.
Example: Tafasitamab targets CD19 receptors.

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

Q: What is CDC (Complement-Dependent Cytotoxicity)?

A

mAbs bind target receptors and recruit complement proteins (e.g., C1q).
Leads to membrane attack complex formation and cell lysis.

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

Q: What is ADCP (Antibody-Dependent Cellular Phagocytosis)?

A

Fc region of mAbs binds Fc gamma receptors on macrophages, triggering phagocytosis.

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

Q: What are the main routes of administration for mAbs?

A

IV, subcutaneous (SC), or intramuscular (IM).
SC and IM routes have slow absorption due to the large molecular size of mAbs.

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

Why do mAbs have restricted tissue penetration?

A

Large size (~165 kDa) limits diffusion; penetration relies on transcytosis or leaky vasculature.

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

Q: How are mAbs eliminated?

A

Proteolytic degradation in lysosomes after endocytosis.
No renal elimination due to large size (above glomerular filtration cutoff).

19
Q

Q: What is FcRn recycling and its role in mAb PK?

A

FcRn binds mAbs at acidic pH in endosomes, protecting them from degradation.
At neutral pH, mAbs are released back into circulation, extending half-life.

20
Q

Q: What is Target-Mediated Drug Disposition (TMDD)?

A

mAbs binding to antigens are internalized and degraded, influencing clearance.
High antigen levels increase mAb clearance.

21
Q

Q: What are Anti-Drug Antibodies (ADAs) and their types?

A

ADAs are immune responses against mAbs. Most biological drug products elicit some level of anti-drug antibody (ADA) response. This is because biological components are detected by the body and induce an immune (humurol) response resulting in the formation of antibodies.

Neutralizing ADAs: Bind Fab region (where they’re supposed to bind to antigen receptor) , blocking antigen interaction. (The upper Y section )
Non-neutralizing ADAs: Bind Fc region, preventing FcRn recycling.

22
Q

Q: What affects mAb half-life?

A

FcRn-mediated recycling extends half-life.
High antigen levels reduce half-life due to increased lysosomal degradation.

23
Q

Q: How do mAbs differ from small molecules in distribution?

A

Small molecules diffuse easily; mAbs rely on transcytosis.
mAbs have smaller distribution volume limited to extracellular space.

24
Q

Q: How are small molecules and mAbs cleared differently?

A

Small molecules are renally excreted; mAbs are cleared via lysosomal degradation.

25
Q: What is the bioavailability of mAbs by route of administration?
IV: 100%. SC and IM: Erratic, ranging from 20–95%.
26
Q: What is the role of FcRn in mAb recycling?
FcRn binds to the Fc region of mAbs, protecting them from degradation. Promotes recycling and extends half-life. Capacity limited
27
Q: What defines a biosimilar?
Must have the same amino acid sequence and similar PK/PD as the reference product. Differences in glycosylation can affect immunogenicity and clearance. Different clones / manufacturing
28
Challenges in mAb Therapy Q: What causes ADA formation in mAb therapy?
Non-humanized mAbs, impurities, excipients, or aggregation of the mAbs. Combination therapies (e.g., with radiation) can increase ADA formation.
29
Q: How does ADA formation affect mAb therapy?
Reduces mAb concentration and efficacy. Neutralizing ADAs prevent binding to the target antigen. Non-neutralizing ADAs block FcRn recycling.
30
Q: Why do pediatric patients differ in mAb PK?
Clearance rates and distribution volumes are significantly different compared to adults
31
Q: Why is renal elimination not applicable to mAbs?
Glomerular filtration cutoff is ~60 kDa; mAbs are ~165 kDa, preventing renal clearance.
32
Q: How does mAb size impact extravasation into tissues?
Large size restricts penetration; requires transcytosis or leaky vasculature for access.
33
Q: What is the main limitation of mAb absorption in SC and IM routes?
Low convective flow (15 mL/day in lymphatic system) delays uptake.
34
What is Target-Mediated Drug Disposition (TMDD)?
TMDD occurs when mAbs bind to their target antigens, leading to internalization and degradation. It is a significant pathway for mAb clearance.
35
Q: How does antigen expression affect TMDD?
High antigen levels increase mAb clearance as more mAbs bind and are degraded. Low antigen levels result in less TMDD, leading to longer circulation of mAbs.
36
Q: How does mAb concentration influence TMDD?
Low mAb concentration: TMDD contributes significantly to mAb clearance because antigen-binding sites are not saturated. High mAb concentration: TMDD becomes saturated, reducing antigen-mediated clearance and increasing circulating mAb levels.
37
Q: How does TMDD relate to dose optimization?
Dose adjustments must balance antigen levels and FcRn recycling to maintain efficacy and extend half-life. Too low a dose leads to rapid clearance; too high a dose may saturate receptors.
38
Q: How does TMDD affect pharmacokinetics?
TMDD often leads to non-linear pharmacokinetics. Low mAb concentrations show increased clearance due to antigen-mediated binding. High mAb concentrations result in reduced clearance as TMDD saturates.
39
Q: What is an example of TMDD in practice?
Adalimumab (targeting TNF-alpha): TMDD contributes to linear pharmacokinetics as TNF-alpha is a soluble ligand. Trastuzumab (targeting HER2): Non-linear pharmacokinetics due to membrane-bound receptor targeting.
40
Q: Why is TMDD capacity-limited?
Receptor availability for binding is limited. As mAb dose increases, receptors become saturated, reducing TMDD's impact on clearance.
41
Intracellular Expression of FcRn
In most cells (e.g., phagocytes, endothelial cells, epithelial cells), FcRn is primarily found in endosomes (intracellular vesicles). Functions: After IgG or albumin is internalized through endocytosis, FcRn binds these proteins at acidic pH (~6.0) within the endosome. Protects IgG and albumin from lysosomal degradation by recycling them back to the cell surface for release into the bloodstream at neutral pH (~7.4).
42
Unfolding
Eventually aggregates, can lead to immune reactions & ADAs
43
Humira (Adalimumab) success led to the marketisation of many biosimilars (Imraldi, Idacio, Amgevita, Hyrimoz and Yufilma). How are these mAbs formulated? How the formulation of these mAbs has evolved to reduce the side effects during delivery of the mAb?
Formulation of mAbs like Adalimumab and Biosimilars 1 .Active Pharmaceutical Ingredient (API): Adalimumab and its biosimilars are recombinant human monoclonal antibodies. They are highly sensitive proteins that require careful stabilization in formulation. 2. Excipient: 1)Buffer Systems: Typically phosphate or citrate buffers to maintain the pH (usually between 5-6.5), ensuring the stability and solubility of the mAb. 2)Salt and tonicity modifiers: Sodium chloride or other agents are added to adjust isotonicity to match the body's natural osmolarity, reducing injection site reactions. 3) Protein Stabilizers: Sugars (e.g., trehalose, sucrose, and polyols (e.g., sorbitol) help maintain protein stability during storage and transport. 4)Surface active agents: Polysorbate 20 and Polysorbate 80 are included to prevent aggregation and surface adsorption of the protein. 5)Antioxidants: Minimize oxidation of sensitive amino acid residues (e.g., methionine and tryptophan). 6)Lyoprotectants:use of sugar, PEG,same mechanism as stabilizers.In addition, sugar is susceptible to hydrolysis at low PH. 7)Preservatives: Multidose formulations may include antimicrobial agents to prevent contamination. 2 Evolution of mAb Formulations to Reduce Side Effects 1. High Concentration Formulations (HCFs): Challenge: Early formulations were dilute, requiring large injection volumes, leading to discomfort. Solution: Advances in solubility enhancers allowed the development of high-concentration formulations (HCFs) that reduce the injection volume without compromising stability, improving patient compliance. 2. Improved Stability: Protein Aggregation: Aggregates can trigger immunogenicity (unwanted immune responses). Modern formulations use advanced excipients and surfactants to mitigate aggregation. Oxidation and Deamidation: Antioxidants and optimized pH reduce these degradation pathways, improving stability and reducing inflammatory reactions. 3. Reduced Injection Pain and Site Reactions: Tonicity Adjustments: Early formulations were hypertonic, causing stinging or burning sensations. Current formulations match the body's isotonic conditions. Citrate-Free Formulations: Citrate buffers, while stabilizing, caused injection pain in some patients. Citrate-free formulations (e.g., for Humira biosimilars) eliminate this side effect. 4. Advances in Delivery Devices: Auto-injectors with thin, fine-gauge needles reduce the mechanical trauma during injection. Prefilled syringes and self-injection devices allow for precise delivery, minimizing dosing errors and discomfort. 5. Reduced Immunogenicity: Humanization of mAbs: Adalimumab is fully human, minimizing immune responses compared to chimeric or murine mAbs. Purity Improvements: Enhanced manufacturing processes reduce impurities and host cell proteins that might trigger an immune response. 6. Ready-to-Use (RTU) and Longer Shelf Life: Modern biosimilars offer RTU solutions that don't require reconstitution, simplifying use for patients and healthcare providers 7. pH Optimization: Modern formulations have optimized pH to balance stability and patient comfort, as extremes in pH can contribute to injection site discomfort.
44
What pre-screening is necessary before initiating a biologic?
Screen patients for: Latent tuberculosis (TB) Hepatitis B virus (HBV) infection Hepatitis C virus (HCV) infection Testing for TB and HBV is standard and should be performed in all patients who are preparing to start biologic therapy. There are no guidelines regarding HCV screening in this population, but testing is suggested. If a patient is having severe diarrhoea, or diarrhoea with fever and bleeding, then clinicians may want to check whether the patient is infected with Clostridium difficile or cytomegalovirus, as these infections could be driving symptoms in the absence of active IBD. Liver,kidney function tests FBC Appropriate screening prior to starting biologic therapy is very important, as reactivation of an infection could make the patient sicker. If a patient has recurrent TB, for example, administration of an immunosuppressant could exacerbate the infection and cause an overall worsening of the patient's condition. Once the clinician has decided that a patient truly has active IBD, then he or she needs to educate the patient about the risk-to-benefit profile of the drug. · Liver function, lipids, FBC Liver function The liver toxicity associated with ICI treatment is mostly immune-mediated hepatitis - hence liver function tests should be checked If liver enzyme levels rise significantly, the biological DMARD may need to be adjusted, or treatment may need to be paused to prevent further liver damage. Therefore, baseline LFTs ensure that any subsequent changes can be detected and managed early. Lipids Certain biological DMARDs, particularly TNF inhibitors (e.g., adalimumab, infliximab) and IL-6 inhibitors (e.g., tocilizumab), can interfere with lipid metabolism, leading to dyslipidemia—an abnormal level of lipids in the blood, such as high cholesterol or triglycerides. TNF inhibitors have been associated with increased total cholesterol and LDL (bad cholesterol) levels. IL-6 inhibitors can raise total cholesterol and triglyceride levels as well. This effect is believed to occur due to changes in inflammatory cytokine levels, which are involved in lipid metabolism. Impact → These changes in lipid levels can increase a patient’s risk of cardiovascular disease (CVD), including heart attacks, strokes, and other vascular complications. Cardiovascular risk is particularly important in patients with autoimmune diseases, who may already be at a higher risk due to the chronic inflammation associated with their condition. https://pmc.ncbi.nlm.nih.gov/articles/PMC4751079/ FBC Biological DMARDs can lead to conditions such as neutropenia (low white blood cells), thrombocytopenia (low platelet count), or anemia (low red blood cells). This can make patients more vulnerable to infections. A baseline FBC helps identify any pre-existing hematological issues and provides a reference point to monitor changes in blood cell counts during treatment. https://www.uhd.nhs.uk/services/rheumatology/medication#:~:text=Blood%20Monitoring,These%20are%20briefly%20explained%20below. · Myelinating disease Anti-tumour necrosis factor (TNF) agents like adalimumab are safe and effective for rheumatologic disorders, but they have been reported to cause demyelinating diseases like multiple sclerosis. Symptoms of myelinating disease to monitor: Vision changes. Tingling or numbness. Fatigue. Bladder or bowel problems. Trouble walking. Stiff or weak muscles. https://academic.oup.com/rheumatology/article/58/2/e3/5076446#130033813 · Contraception for individuals of child-bearing potential Many DMARDs have potential teratogenic effects or are newer agents with limited safety data in pregnancy. https://journals.lww.com/jclinrheum/abstract/2021/04000/contraceptive_use_in_women_of_childbearing_ability.3.aspx · Vaccinations The small dose of a live organism in live vaccines may be enough to cause symptoms of the disease in people who are immunosuppressed. For this reason, live vaccines aren't recommended if you're on certain DMARDs or biological therapies as these are immunosuppressive drugs.