Exam 3 Flashcards

1
Q

What are the functions of the hindbrain?

A

Hindbrain - Contains medulla, pons, and cerebellum. Involved with non-conscious function.
- medulla: autonomic functions, including respiration, cardiac function, reflexes. Part of the reticular system (wakefulness, arousal). Part of the brain stem.
- pons: acts as a bridge. It relays signals from the forebrain to the cerebellum. Also part of the brain stem.
- cerebellum: governs motor coordination that produces smooth movements.

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

What are the functions of the midbrain

A

Midbrain - Contains the substantia nigra (SN).
- SN pars compacta: provides input to the basal ganglia and supplies dopamine to the striatum. It’s involved with voluntary motor control and some cognitive functions.
- SN pars reticulata: provides output functions. Relays signals from basal ganglia to the thalamus

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

What are the functions of the forebrain?

A

Forebrain - Contains the cerebral cortex, basal ganglia (not SN, yes striatum, globus pallidus, and subthalamic nucleus), limbic system (hippocampus, amygdala), and diencephalon (thalamus, hypothalamus)
- cortex: involved in processing and interpreting information.
- basal ganglia: includes the striatum, globus pallidus, and subthalamic nucleus. Involved in voluntary motor control and some cognitive functions.
- limbic system: includes amygdala that is tied to emotions and the hippocampus that is tied to memory.
- diencephalon: includes thalamus that acts as a ‘relay station’ to and from the cortex and the hypothalamus that is involved with non-concious functions (internal homeostasis, emotions, hormonal control, direct neural regulation).

Our senses receive information that is passed through the thalamus to the cortex and back. Within these cortico-thalamic loops, decisions about how to interpret and act on sensory information is made. Damage to the cortex can affect movement, speech, and personality.

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

What are the pathogenic conditions that involve the different brain regions?

A

cerebellum - undergoes neurodegeneration in spinocerebellar ataxias, meaning movements become jerky.

basal ganglia - the SN pars compacta undergoes cell loss during parkinson’s

cortex - schizophrenia is considered a disease of the frontal cortex and involves excess dopamine (?)

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

What is the structural basis of the blood-brain barrier?

A

Endothelial cells (facing blood) have very tight junctions in between them, which is the reason why we have the BBB in the first place. Beneath (on the brain side) the endothelial cells is a basement membrane.

Astrocytes are a key structural component. They form the lower part of the BBB.

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

What is the basic structure of a neuron? What are the roles of glial cells in the brain (astrocytes, oligodendrocytes, microglia)?

A

Structure -
- Dendrites are projections that receive signals from other neurons
- Soma contains the nucleus
- Axon connects soma to synaptic bouton
- Myelin sheaths insulate the axon
- Synaptic bouton contains the vesicle that fuses with the membrane in order to release the neurotransmitters into the synaptic gap/cleft. This release is triggered by electrical depolarization of the neuron (Na+)

Astrocytes - Provide growth factors and antioxidants to neurons. They also remove excess glutamate (too much glutamate can harm neurons). Also support the blood-brain barrier

Oligodendrocytes - Produce the myelin sheath that insulates axons.

Microglia - Similar to macrophages. They provide growth factors and clear debris by phagocytosis. Also play a role in neuroinflammation.

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

What are the principles of EPSPs, IPSPs, and action potentials?

A

Action potential - action potentials are always going to be the same size. The size of a signal is more about the # of action potentials (more rapidly or slowly). Larger diameter axons give faster rates (the rate is proportional to the diameter). The current is carried by a nerve fiber, or bundle of axons.

EPSP - Excitatory post-synaptic potential. It depolarizes the neuron (more positive), but not enough for an action potential. It’s like a little blip, but needs more Na+ to enter the cell in order to fully depolarize to trigger an action potential.

IPSP - Inhibitory post-synaptic potential. Makes the neuron more negative, so that the same signal that caused an action potential before is no longer able to. So what would’ve been an action potential is now just an EPSP.

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

What are the major amino acid neurotransmitters in the CNS?

A

Amino acid:
1. GABA - A major inhibitory neurotransmitter in the brain. It depresses neuronal excitability by increasing the flux of Cl- ions into the neuron. GABA-A is an ion channel. Drugs that interact with GABA are generally CNS depressants. Dysfunction of GABA transmission plays a role in epilepsy, spasticity, and addiction/alcohol.
2. Glycine (not talked about much) - Similar to GABA, but in the spinal cord
3. Glutamate (opp. of GABA) - A major excitatory neurotransmitter in the brain. Excess of glutamate can cause neuronal damage due to letting too much Ca2+ into the cell. Glutamate receptors are ion channels (AMPA, NMDA) and GPCRs. Too much glutamate may result in epilepsy and schizophrenia.

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

What are the major non-amino acid neurotransmitters in the CNS?

A

Non-amino acid:
1. Acetylcholine - ACh acts on muscarinic and nicotinic receptors. It is predominant within the basal forebrain, pons, cortex, and basal ganglia. Dysfunction in ACh transmission may result in cognitive function decline, nicotine dependence, and movement disorders. Drugs targeting ACh are cholinesterase inhibitors.
2. Dopamine (DA) - DA is predominant in the midbrain (SN pars compacta and ventral tegmental area). Dysfunction in DA transmission may result in schizophrenia, Parkinson’s, addiction, depression, and ADHD. Drugs will target D1-D5 receptors (GPCRs) and the DAT. Targeting DAT increases extracellular DA, which can produce euphoria (inc. risk of addiction). Too much DA can result in schizophrenia, not enough can result in Parkinson’s.
3. Norepinephrine - NE is predominant in the pons (locus coeruleus). Dysfunction in NE transmission may result in memory loss, depression, addiction, and pain. NET inhibitors are used to treat depression, since depression is thought to involve a NE deficiency.
4. Serotonin; 5-HT - Predominant in the midbrain/pons (raphe nuclei). Dysfunction in serotonin signaling may result in depression, mood disorders/anxiety, and schizophrenia. Drug targets are different serotonin receptors (14 GPCRs, one gated ion channel, and SERT) depending on the indication.

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

What are the mechanisms by which drugs can modulate neurotransmission in the CNS (with examples)?

A
  1. Post-synaptic receptor. Can be agonist or antagonist.
  2. Transporter. To control the amount of neurotransmitter in the synapse.
  3. Metabolism. To breakdown/slow down breakdown of neurotransmitter
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What are the symptoms and neuropathology of MS?

A

MS - an immune-related (inflammatory) disorder that involves destruction of the myelin sheath that surrounds neuronal axons, primarily in the spinal cord & brain. In place of this destruction, scarring accumulates.

Symptoms: Many different symptoms that vary from patient-to-patient. It depends on where this myelin breakdown is.
- Visual problems
- Numbness, tingling
- Fatigue, motor weakness
- Difficulty walking, gait problems, ataxia
- Pain
- Dizziness, vertigo

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

What are some environmental and genetic factors that are associated with elevated MS risk?

A
  • Age (peak incidence is people in their 30s)
  • Geography (lower exposure to sunlight, decreased 25-hydroxyvitamin D levels)
  • Viral infections (Epstein-Barr Virus: an infection can lead to an immune response against the antigen. That particular protein looks similar to one of our own proteins, so the immune response may also cross-react with one of our own proteins) People with a particular HLA phenotype have an increased risk of MS if they have EBN antigen
  • Environmental insults (urbanization) after age 15
  • Cigarette smoking increases risk and severity of MS
  • Genetics (highest risk in twins, which may indicate a polygenic mode of inheritance)

It is a connection of environment and genetics that results in MS.

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

What are the different clinical forms of MS?

A

Relapsing-remiting MS (RRMS) - About 85% of cases. Relapses of neurological dysfunction lasting weeks/months affecting the brain, optic nerves, and/or spinal cord. Initial symptoms disappear, but there’s less remission with each relapse

Secondary progressive MS (SPMS) - The damage is permanent and progressively worsening. There is less inflammation than RRMS. Little remission seen here.

Primary progressive MS (PPMS) - About 15% of cases. This at first resembles SPMS. These individals are already in the progressive phases that did not cross the threshold until the damage was permanent.

Clinically isolated syndrome (CIS) - The first auto-immune episode that crosses the threshold, leading to symptoms. This episode lasts longer than 24 hours.

**the progressive phase involves cytodegeneration where there is loss of myelin, axons, and oligodendrocytes.

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

What are the autoimmune and degenerative phases of MS?

A

Autoimmune phases - Antigens that are released from the CNS or cross-reactive foreign antigens are presented to B and T cells in the lymph nodes. More B and T cells migrate to the CNS and they carry out immune functions, releasing antibodies and cytokines, at the CNS sites.

Degenerative phases - Due to the immune function damage in the CNS, antigens are released that further prime immune cells to then cause more damage (also in periphery).

It’s a vicious cycle.

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

What autoimmune responses are involved in MS?

A

Dendritic cells present CNS antigens. Different T cells are activated from this antigen presentation. Dendritic cells also stimulate B cells from this. The activated B and T cells will then migrate to the BBB, but they can’t get in. α4-integrin mediated binding allows for the penetration of the BBB, so the B & T cells enter the brain. The B cells mature into plasma cells that produce antibodies that attack the antigens on the antigen presenting cells. The active T cells interact with antigen presented by MHC on the surface of neurons, oligodendrocytes, and microglia. Once the T cells bind, they release cytokines (inc. perforin and granzyme that lead to oligodendryocyte destruction) and stimulate macrophages, which leads to death to the myelin sheath.

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

What are the effects of demyelination on axon conductive properties?

A
  • Demyelination causes action potentials to slow down due to slowed propagation of the current from the loss of insulation from the myelin sheaths. So we no longer have sustained conduction.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What are the steps involved with remyelination? What role does remyelination play in MS pathology?

A
  • After the damage has been done, there is migration of neuronal stem cells and oligodendrocyte progenitor cells (OPC) to the lesion. This leads to the replacement of damaged neurons and oligodendrocytes that remyelinates the axon.
  • During demyelination, oligodendrocytes are lost and macrophages enter. Astrocytes enter and start to form scars.
  • OPCs are recruited and mature to oligodendrocytes. These then produce new myelin, but this ultimately fails, leading to scarring by astrocytes called astrogliosis.
  • The re-establishment of the myelin sheath is good, but it’s not as strong as the original myelin.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What are the pathogenic mechanisms that are current or potential targets for MS therapies?

A

Immune system targets:
- T cell binding/penetration of BBB -> α4-integrin antibodies, IFN-β
- T cell/APC interactions -> altered peptide ligands that interfere with this interaction
- cytokines -> antibodies that are specific to cytokines to neutralize them

Remyelination targets:
- Agents that facilitate OPC recruitment or promote OPC differentiation

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

How can gadolinium-enhanced MRI be used to visualize lesions in MS patients?

A

Gadolinium penetrates the brain in regions where the BBB is compromised. We can see where in the brain there is ongoing damage. This is useful for active damage and autoimmune events during a relapse phase, but it would recede during the remission phase as there is remyelination.

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

What are the symptoms, pathophys, and treatments of Guillain-Barré syndrome?

A

Acute, inflammatory neuropathy. This is peripheral, not in the CNS (like MS).

symptoms - starts in lower extremities. See weakness that ascends to proximal muscles and upper extremities. Can progress to total paralysis w/ death from respiratory failure in days. Generally peaks at 10-14 days.

pathophys - autoimmune attack on peripheral nerves by circulating antibodies, resulting in demyelination.

treatment - ventilation (if respiratory difficulty), plasmapheresis (to eliminate auto-antibodies), and intravenous immunoglobulin administration.

**recovery is slow, but most surviving patients recover completely (the rest have minor motor deficits)

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

What are the differences between MS drugs that are used to treat acute attacks vs. disease-modifying drugs?

A

Acute attacks are treated with anti-inflammatory drugs:
- Methylprednisolone
- Prednisone
- Adrenocorticotropic hormone (ACTH): stimulates anti-inflammatory steroid production.

Disease modifying therapies (DMTs) reduce relapse rates and may slow the progression of disability. These alter the course of the disease.

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

What DMDs are used as first-line drugs for MS? What are their mechanisms of action?

A

interferon β1a and β1b - act in the periphery to inhibit autoreactive lymphocytes, like T cells and dendritic cells. Also acts at the BBB to inhibit the penetration by matrix metalloproteinase (MMP), which breaks down components of the BBB, thus letting in lymphocytes. (not in CNS)

glatiramer acetate - a synthetic peptide that mimics antigenic properties of myelin basic protein, which binds to APCs, resulting in the inhibition of autoreactive lymphocytes, like T cells and dendritic cells. (only in periphery)

fingolimod - S1P receptor agonist, which stimulates oligodendrocyte survival and therefore remyelination. It also interferes with lymphocyte movement out of lymphoid organs. (in periphery (lymph node) and CNS)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What DMDs are used as second-line drugs for MS? What are their mechanisms of action?

A

natalizumab - monoclonal antibody that acts at the BBB and is specific for α4 integrin, which usually binds with β1 integrin to produce VLA-4. Interfering/neutralizing with VLA-4 binding with its ligand interferes with B and T cell movement into the CNS.

mitoxantrone - this is an anthracenedione with cytotoxic activity. it reduces lymphocyte numbers by causing DNA strand breaks and delaying DNA repair. (periphery)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What are the new DMDs for MS? What are their mechanisms of action?

A

dimethyl fumarate - works in the periphery and CNS and has multiple MOAs. In the CNS, it activates Nrf2 (neuroprotective transcription factor most common in astrocytes) that increases antioxidant and anti-inflammatory pathways (glutathione biosynthesis and detoxification). Also may promote remyelination and suppress activated T cells/dendritic cells in the periphery.

rituximab (also called ocrelizumab) - targets CD20 as well. It is effective for some PPMS patients!!! That’s unique.

alemtuzamab - targets CD20, which is on the surface of B cells. (in periphery)

cladribine - cytotoxic agent that is taken up by cells. It will be phosphorylated and integrated into DNA, resulting in cell death and lymphocyte depletion.

teriflunomide - cytotoxic agent that interferes with DNA metabolism, which inhibits proliferation of peripheral lymphocytes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What MS drugs are in clinical trials or in earlier experimental stages? What are their mechanisms of action?

A

firategrast - small molecule that targets α4 integrin (VLA-4). It’s active at the BBB.

amiloride - Only active in the CNS. It’s a small molecule that targets ASIC-1, which is an ion channel that is responsible for the entry of too much Ca2+. Antagonism of ASIC-1 leads to direct neuroprotective effects.

laquinimod - Only in the CNS. Not like the other imods. This stimulated BDNF, which leads to direct neuroprotective effects. Also shown immunomodulatory effects.

opicinumab - Only in the CNS. Interferes with LINGO-1, which is a protein that negatively regulates OPC differentiation. That is bad because we need more oligodendrocytes for MS.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What are features of drugs that limit their therapeutic effectiveness (ex. toxicities, induction of neutralizing antibodies)? (interferon, fingolimod, natalizumab, dimethyl fumurate, ocrelizumab, opicinumab)

A

PML - fingolimod, natalizumab, dimethyl fumurate

limited by neutralizing antibodies - interferon, natalizumab, ocrelizumab, opicinumab

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

How can you identify the structures of fingolimod, dimethyl fumarate, and cladribine?

A

fingolimod has a long fatty acid chain, which is what helps it get into the CNS

dimethyl fumarate looks like a mirror image of its self with methyls and esters on each end.

cladribine has a halogen on one of its rings.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

What is the diagnostic criteria and types/subtypes for multiple sclerosis?

A

There is no single feature or criteria that is sufficient to diagnose MS. It’s important to rule our other disorders. Two of the most important factors is a dissemination in time (new lesions 30 days apart) and dissemination in space (damage that is in more than one place (2-4 regions)).

Types: Clinically isolated syndrome, relapsing remitting, secondary progressive, primary progressive, and progressive relapsing.
- The disease modifying therapy is usually for RRMS

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

What is the rating scale that is associated with the diagnosis of multiple sclerosis?

A

The Expanded Disability Status Scale (EDSS) is the standard scale that providers use to assess MS disability.

0 = normal neurological function
10 = death

4 is the average for pts presenting with MS

5 = cane, 6 = walker, 7 = wheelchair

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

What is the clinical course, goals of therapy, and expected outcomes of treatment with drug and non-drug therapy for MS?

A

Goals of treatment:
- Start early. Hopefully we can reduce neurodegeneration by starting meds early.
- Treat acute attacks aggressively to promote more complete remission
- Use disease modifying drugs (start at CIS stage if possible). We want to target inflammation by various mechanisms.
- Increase quality of life!

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What oral drug therapy is used for MS (brand + generic), including side effects, and monitoring parameters?

A

Dimethyl fumurate (Tecfidera), Diroximel fumerate (Vumerity) - Capsule should not be opened. Monitor LFTs (hepatotoxicity) and CBCs with differential (neutropenia). It is associated with PML. May cause flushing, so pt can take an ASA 30 mins prior to the dose.

Fingolimod (Gilenya, Tascenso ODT) , Ozanimod (Zeposia), Ponesimod (Ponvory), Siponimod (Mayzent) - CV side effects!! Contraindicated with past arrhythmia diagnosis or other CV diagnoses in the past 6 months. Need to monitor ECG for the first 6 hours. Monitor CBC and pt needs routine eye exams due to risk of macular edema. D/c of the drug can result in worsening MS symptoms.
- Ozanimod: avoid use with MAO inhibitor
- Siponimod: CYP2C9 genotype testing is required before prescribing

Teriflunomide (Aubagio) - Contraindicated in pregnancy!! To the point that we will do accelerated elimination!

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What injection drug therapy is used for MS (brand + generic), including side effects, and monitoring parameters?

A

interferon beta-1a (Avonex, Rebif), peginterferon beta-1a (Plegridy), interferon beta-1b (Betaseron, Extavia) - Flu-like symptoms can occur after injection, so pt can take APAP or NSAID prior. Also evening dosing is good for this. See psychiatric side effects (depression, suicidal thinking)!!! Monitor LFTs and TSH.

Glatiramer acetate (Copaxone) - These have injection side effects, esp. immediately post-injections, such as flushing, sweating, dyspnea, chest pain, anxiety, and itching. Also need to rotate sites due to lipoatrophy. It may be preferred if treatment is necessary in pregnancy!

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What infusion drug therapy is used for MS (brand + generic), including side effects, and monitoring parameters?

A

Alemtuzumab (Lemtrada) - There are possible fatal infusion reactions and autoimmune conditions. Also associated with increased risk of malignancies. It is contraindicated in HIV infection due to prolonged decreased CD4 count. Need all vaccinations at least 6 weeks before starting treatment. Can premedicate prior to dose.

Natalizumab (Tysabri) - Significant association with PML. Need all vaccinations at least 6 weeks before starting treatment. Can premedicate prior to dose.

Ocrelizumab (Ocrevus) - Only drug FDA-approved for PPMS!! Contraindicated in active Hep B. Also associated with increased risk of malignancies. Need all vaccinations at least 6 weeks before starting treatment. Can premedicate prior to dose.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

What are the boxed warning and REMS programs that are associated with drug therapy for MS?

A

REMS (risk evaluation mitigation strategies): Alemtuzumab, Natalizumab

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

What can be used to treat gait disturbances, bladder dysfunction, cognitive dysfunction, and spasticity for MS patients?

A

Gait disturbances - Dalfampridine (Ampyra) may improve walking speed by 3 seconds.

Bladder dysfunction - anticholinergics like oxybutynin and tolteridone. BUT these may worsen cognitive dysfunction

Cognitive dysfunction - Cholinesterase inhibitors and memantine.

Spasticity - Baclofen is approved. Also physical therapy, stretching, exercise, etc.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

What type of vaccines are preferred for MS patients?

A

Inactivated vaccines are preferred for people with multiple sclerosis. These can be given at anytime.

It is preferred that the pts get the varicella vaccine, especially if they are going to start MS drugs that suppress cell-mediated immunity (ex. fingolimod, alemtuzumab).

If they haven’t started anything yet, we want to get their vaccines in about 6 weeks before starting immunological therapy.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

How long after stopping these drugs would you need to continue contraception for MS? Fingolimod, Ozanimod, Ponesimod, Siponimod, Orelizumab, Cladribine

A

Ponesimod - 7 days
Siponimod - 10 days
Fingolimod - 2 months
Ozanimod - 3 months
Ocrelizumab - 6 months
Cladribine - 6 months

Please
Stop
Fingolimod
Or
Ocean
Clash

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

What are the physical symptoms and neuropathological underpinnings of PD, including the Braak staging of the disease?

A

Symptoms - resting tremor, ridigity, akinesia/bradykinesia, postural instability, mask-like appearance, speech difficulties, cognitive deficits, depression

  • chronic, progressive, irreversible, disease that results from a deficit in the extrapyramidal system (basal ganglia), which is involved in noncortical voluntary motor control.
  • We see Lewy bodies in the neurons and loss of neurotransmission through the nigrostriatal system (due to loss of dopamine-releasing neurons)
  • there is Braak staging that represents the spreading of PD from the lower brainstem through the entire brain. There is a symptomatic threshold in stage 3 where Lewy bodies are present in the substantia nigra.
  • by the time pts present with symptoms, about 50% of the nigral dopamine neurons, or 70-80% of the nerve terminals in the striatum are already lost.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

How does neurotransmission from the substantia nigra to the basal ganglia get disrupted in PD?

A

There are two types of neurons in the striatum that lead to two different pathways:
1. D1 receptors (direct): shortest pathway.
2. D2 receptors (indirect): slower pathway, involves additional components.

In a healthy person, dopamine will be released from neurons to go onto signal in the striatum through these two pathways. This signaling favors movement (increase thalamocortical signaling). So, the loss of dopaminergic neurons in PD will disrupt this signaling to cause the motor dysfunction.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

What is the role of anti-muscarinic drugs in PD therapy?

A
  • The D2 (indirect) pathway is antagonized by acetylcholine (balance). So if we inhibit this muscarinic/cholinergic signaling, it will help regain the balance of muscarin and dopamine signaling.

These are used as adjunct therapy for tremor in PD. But need to keep the dose low in order to prevent the cognitive deficit side effect.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

Why do we treat PD patients with L-DOPA and carbidopa?

A

L-DOPA is the gold standard for PD therapy. It is orally active and can enter the CNS (plain dopamine can’t enter the BBB). L-DOPA is neutral at a pH of 7, dopamine is not.

This boosts dopamine!

carbidopa is a peripherally-acting DOPA decarboxylase (DDC) inhibitor. This means it stops the peripheral breakdown of L-DOPA into dopamine, which allows us to decrease the dose of L-DOPA by 4x! This is especially good because it means more dopamine in the CNS and it reduces the risk of side effects, like nausea, HTN, and psychosis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What are the challenges that are associated with L-DOPA therapy?

A

On/off:
- Won’t work forever. After years of L-DOPA treatment, we will see large peaks and troughs from the drug, which can produce dyskinesias and off-states.
- These dyskinesias and on/off effects are major problems in long-term therapy with levodopa.
- We can alleviate this by infusing L-DOPA (or just continuous) instead of taking orally (pulsatile).

Prodrug conversion:
- L-DOPA is a prodrug that but be converted to dopamine by DDC in the surviving dopaminergic neurons, but because the disease is progressive, this conversion will eventually stop happening.
- We can address this by using dopamine receptor agonists, which can target postsynaptic dopamine receptors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

What are the PD drugs that are ergoline and non-ergoline dopamine receptor agonists? Which ones are used for adjunct treatments vs. monotherapy?

A

ergoline:
- bromocriptine: D2 agonist. Used as an adjunct with L-DOPA to keep the dose down

non-ergoline: used more often
- ropinirole, pramipexole, rotigotine: D2/D3 agonists with fewer side effects. Usually monotherapies for early-stage PD, efficacy may last for 2-4 years.
*rotigotine is given through transdermal patch which allows for continuous drug delivery.

  • apomorphine: mixed D1/D2 agonist. Can provide rapid relief of the on/off state, but has vomit-inducing side effects, so it’s use is limited.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

What are the PD drugs that interfere with dopamine metabolism?

A

MAO-B converts active DA into inactive DOPAL, so we want to inhibit MAO-B. COMT also destroys DA and L-DOPA.

irreversible MAO-B inhibitors: selegiline, rasagiline
- Can be used initially as a monotherapy to delay the use of L-DOPA or as adjunct to decrease dose of L-DOPA

reversible MAO-B inhibitor: safinamide
- no proparygylamine. Used as an adjust to L-DOPA/carbidopa to reduce the dose. This is particularly useful during off episodes

COMT inhibitors: entacapone, tolcapone, opicapone
- entacapone/opicapone decrease metabolism of L-DOPA in the periphery, allowing more to reach the brain
- tolcapone works in the CNS!!! which allows levels of CNS dopamine to remain higher and increases dopamine elimination time.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

What are the chemical structures of dopamine, L-DOPA, carbidopa, and selegiline?

A

L-DOPA - looks like dopamine but has a carboxyl group, which is what allows its access to the CNS

Carbidopa - looks like L-DOPA, but has one more amino group

selegiline - has the propargylamine (triple bond) which results in irreversible inhibition of MAO-B.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

How would we recommend a patient-specific treatment plan for the management of PD? What is first line and second line?

A

Goals of therapy - Minimize/manage motor and non-motor symptoms, maintain highest QOL possible, preserve activities of daily living, minimize/manage adverse drug reactions

Want to rule out drug-induced PD (ex. typical antipsychotics). Start treatment when PD interferes with ADL, employment, or QOL.

  1. First line: Dopamine precursor (L-DOPA)
    1.5 Dopamine agonist, MAO-B inhibitors (can be first line in certain patients to delay levodopa use
    - Use dopamine agonist maybe if less than 60 years of and at a higher risk for dyskinesia, but avoid if over 70 years old, if they have impulse control disorder, hallucinations, cognitive impairment, or excessive daytime sleepiness (these are side effects of dopamine agonists)
    - Efficacy: Levodopa/Carbidopa > DA > MAOB-I
  2. COMT inhibitors, amantadine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

How can we therapeutically manage levodopa-induced motor fluctuations?

A

There are four types of LD motor fluctuations:
1. Wearing off: before the next dosing intervals, see signs of motor symptoms -> increase dose or frequency, add DA agonist, MAOI, or COMTI, switch to CD/LD

  1. Freezing: Inability to move due to insufficient or fluctuating DA levels -> increase dose or frequency, add DA agonist (apomorphine), add ODT CD/LD
  2. Delayed onset: therapeutic benefits delayed -> take LD/CD on an empty stomach, change to ODT LD/CD, avoid CR/XR LD/CD
  3. Peak-dose dyskinesias: involuntary body movement caused by high DA levels -> decrease dose of DA or CD/LD, add amantidine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

How can we manage non-motor symptoms for a PD patient? ex. anxiety/depression, constipation, dementia, insomnia, orthostatic HTN, psychosis/delirium, sexual dysfunction

A

Non-motor symptoms - anxiety/depression, constipation, dementia, insomnia, orthostatic HTN, psychosis/delirium, sexual dysfunction

constipation - increase fluid intake, physical activity, stool softeners/laxatives, probiotics

insomnia - non-pharm counseling, melatonin, AVOID benzodiazepines

orthostatic hypotension - non-pharm counseling, midodrine, droxidopa, medical equipment

anxiety/depression - cognitive behavior therapy, SSRI, SNRI, AVOID benzodiazepines

dementia - cholinesterase inhibitor (donepezil, rivastigmine), AVOID anticholinergics, benzodiazepines, antihistamines, sedatives

psychosis/delirium - reduce PD medication doses (if appropriate), pimavanserin!!, atypical antipsychotics (clozapine, quetiapine), AVOID haloperidol, olanzapine, paliperidone, risperidone

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

What is the non-pharm therapy for PD?

A
  • Exercise/physical therapy: it’s been shown that we should start exercise before pharmacotherapy.
  • Nutritional counseling (swallowing may be difficult)
  • Occupational therapy
  • Psychotherapy/support groups
  • Speech therapy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What are the side effects and clinical pearls for dopamine precursors, dopamine agonists, MAO-B inhibitors, COMT inhibitors, amantadine, anticholinergics

A

Dopamine precursors - levodopa
- Side effects: LD motor fluctuations/dyskinesia, hallucinations, N/V
- Pearls: Increased absorption on an empty stomach, but food helps with nausea. Start low dose and titrate up as side effects allow.

Dopamine agonists - non-ergot (used more), ergot
- Side effects: N/V, impulse control disorder (ICD), orthostatic HTN
- Pearls: Very low starting doses, causes fewer motor fluctuations and there are long-acting formulations

MAO-B inhibitors - rasagiline, selegiline, safinamide
- Side effects: N/V, headache, insomnia (selegiline is an amphetamine derivative), hypo/hypertension
- Pearls: dietary restrictions for tyramine-rich foods, risk of serotonin syndrome if used will serotonergic antidepressants, dextromethorphan, and serotonergic opioids

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

What are the side effects and clinical pearls for COMT inhibitors, amantadine, anticholinergics

A

COMT inhibitors - entacapone, opicapone, tolcapone
- Side effects: N/V, urine discoloration (entacapone), hepatoxicity (tolcapone)
- Pearls: used to manage LD/CD fluctuation, no benefit in early PD.

amantadine -
- Side effects: insomnia, confusion/hallucinations, livedo reticularis (rash that can be life threatening)
- Pearls: Can be used for an isolated tremor, limited use due to cognitive side effects

Anticholinergics - benztropine, trihexyphenidyl
- Side effects: confusion/dementia, blurry vision, urinary retention, dry mouth, constipation
- Pearls: limited use due to confusion & side effects. Avoid if over 65 years old. Can help with tremor symptoms.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

What are the symptoms and neurological underpinnings of AD? What happens to brain volume and neurotransmitter level? What part of the brain is most impacted?

A

Symptoms - short term memory loss, impaired ability to learn, reason, impaired ability to carry out daily activities, confusion, untidiness, anxiety, suspicion, hallucinations, motor dysfunction

  • With AD, there is a loss of brain volume.
  • amyloid plaques: extracellular, consist of amyloid-β
  • neurofibrillary tangles: intracellular, consist of hyperphosphorylated tau
  • destruction of synapses occurs, which results in reduced levels of neurotransmitters (esp. ACh, serotonin, norepi, and DA). We also see excitotoxicity and neurotoxicity from dysregulated glutamate.

*basal forebrain is an important place that AD affects. It is involved with learning. Eventually, the disease will spread throughout the entire neocortex

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

What are the environmental and genetic factors associated with elevated AD risk or early-onset AD?

A

environmental factors - age, low educational level, reduced mental and/or physical activity in late life, risks for vascular disease, head injury

genetic - there is a key role for Aβ. Mutations in the gene encoding the Aβ precursor, APP, are linked to early onset AD. Mutations in the gene encoding presenilin proteins that are involved in cleaving Aβ from APP are also linked to early onset AD. Also Down’s syndrome is associated with AD.

54
Q

How is amyloid-β generated from the amyloid precursor protein (APP)?

A

Aβ is attached to APP within the cell membrane. β-secretase and γ-secretase cleave the attachment points, thus releasing Aβ into the extracellular space. This is bad because it can lead to a build up of amyloid plaques.

Also, α-secretase can cleave Aβ. It cleaves Aβ right down the middle, which is desirable because it ensures that the Aβ cannot function.

55
Q

How is Aβ accumulation related to neurofibrillary tangle formation and microglial activation/neuroinflammation?

A

Aβ starts to form oligomers, that go on to form plaques OR act on cell-surface receptors. If they act on these receptors, they can activate kinases that phosphorylate tau. Tau is usually sitting on microtubules, so the phosphorylation of tau can cause a disassociation of tau from the tubules, then that can lead to neurofibrillary tangle formation and the disruption of cytoskeleton.

Aβ is also thought to induce neurotoxicity by triggering microglial activation. Activated microglia cause neuroinflammation and release reactive nitrogen species that cause oxidative stress.

56
Q

How does ApoE genetic status modulate AD risk?

A

ApoE is responsible for transporting cholesterol in the brain (LDL). Altered ApoE function can affect Aβ aggregation or clearance.

There are 3 ApoE isoforms: ApoE2, ApoE3, ApoE4
- Individuals with ApoE2 are less likely to have AD
- Individuals with ApoE4 have an increased risk of AD

57
Q

What is the role of cholinesterase inhibitors and anti-glutamatergic agents in AD therapy?

A

Cholinesterase inhibitors - Block the breakdown of ACh.
- Donepezil: reversible inhibitor of AChE
- Rivastigmine: inhibits AChE and butytylcholinesterase (in the blood); oral or patch
- Glantamine: inhibits AChE AND enhances action of ACh on nicotinic receptors!!

Anti-glutamatergic agents -
- Memantine: noncompetitive NMDA antagonist that blocks glutamatergic neurotransmission
- Too much glutamate leads to excitotoxicty, which can result in neuronal death. But we need glutamate for memory formation. So this is a balancing act.

58
Q

What are the 6 pathogenic mechanisms that are targeted in the development of new AD therapies? Which one has evidence of success so far?

A
  1. Aβ generation - β and γ-secratase inhibitors)
  2. Aβ aggregation
  3. Aβ clearance - vaccines, Aβ antibodies!!!! There is evidence that these decrease plaques in the brain and improve symptoms!!
  4. tau kinase inhibitors
  5. glutamate-mediated excitotoxicity
  6. inflammation or oxidative stress
59
Q

How are Florbetapir (18F) and other imaging agents used to monitor amyloid formation in living individuals?

A

Florbetapir binds β-amyloid. It may help clarify the role of amyloid in pathophys and lead to improved treatments.

18F-Flortaucipir is a radiolabeled agent that is specific for tau.

There are also AD blood tests we can do to detect Aβ levels.

60
Q

What are the similarities and differences between AD and other dementias (vascular dementia, DLB, FTD)?

A

vascular dementia - occurs as a result of brain injury associated with vascular disease or stroke.

dementia with lewy bodies (DLB) - combination of cognitive decline and parkinson’s. Visual hallucinations are a core diagnostic feature.

frontotemporal dementia (FTD) - Neuropathology is characterized by the presence of tau accumulations. There are disturbances in executive functioning. See disinhibited behavior.

61
Q

What are the 6 neurocognitive domains that are the basis for diagnostic criteria for the NCDs?

A
  • Complex attention
  • Learning and memory - More common
  • Perceptual/Motor
  • Executive function - More common
  • Language
  • Social cognition
62
Q

What defines mild and major neurocognitive disorders?

A

Mild:
- Modest cognitive decline from a previous level of performance in one or more cognitive domains
- Does NOT interfere with independence
- Not attributed to a delirium episode (these are acute, temporary cognitive decline)
- Not better explained by another mental or medical disorder

Major:
- Significant cognitive decline from a previous level of performance in one or more domains
- Cognitive deficits interfere with independence
- Not attributed to a delirium episode
- Not better explained by another mental or medical disorder

63
Q

What 4 class of drugs are the most frequent to cause drug-induced cognitive impairment?

A
  • Skeletal muscle relaxants
  • Tricyclic antidepressants
  • Bladder antispasmodics
  • Antihistamines (OTC allergy/cough cold, rx anti-emetics)
64
Q

What are the differences in the course of dementia by dementia type? (alzheimer’s, vascular, lewy body)

A

Alzheimers - progressive, slow, constant decline

Vascular - lots of plateaus until the vascular event (ex. stroke), then it drops and plateaus again

Lewy body - big fluctuations that end up with overall decline

65
Q

What are the differences between these rating scales for dementia: MMSE, ADAS, MoCA, SLUMS

A

MMSE - evaluates orientation, memory, attention, naming, comprehension, spatial orientation. Influenced by age and educational level. Clinically used to follow scores over time.

ADAS - evaluates severity of dysfunction over time. Higher scores indicate worse cognitive performance. (avg decline for pt with severe AD is 6-11 per year)

MoCA - differentiates mild cognitive impairment

SLUMS - tells a story to assess recall

66
Q

What are the cholinesterase inhibitors that we use for AD? What are their dosing, side effects, and interations?

A
  • cholinesterase inhibitors are first line with no preference as to which agent

Donepezil - Start at 5mg qHS, then 10mg qHS after 4-6 weeks.
- Side effects: GI bleeding, N/V/D, bradycardia, syncope, insomnia, weight loss
- P450 2D6 and 3A4 substrate

Galantamine - BID dosing with breakfast and dinner. Doses above 16mg/day are not recommended.
- Side effects: GI bleeding, weight loss, N/V/D, bradycardia, syncope, insomnia
- P450 2D6 and 3A4 substrate

Rivastigmine - BID dosing. Take with meals to minimize GI effects. Also patch form
- Need to remove the patch at the right time or else toxicity! There has been esophageal rupture!!
- No P450 interactions

67
Q

What are the NMDA receptor antagonists that can be used for AD? What are their dosing, side effects, and interactions?

A
  • These do not slow or prevent neurodegeneration
  • FDA approved in moderate to severe dementia only (NOT for mild)

Memantine - Only IR available as generic. Dose adjustment needed in CrCL 5-29 (5mg BID max dose in this case, usually 10mg BID is target)
- Side effects: Caution in pts with seizure disorder, hallucinations, insomnia, confusion
- Caution with carbonic anhydrase inhibitors and sodium bicarbonate. No P450 interactions

Donepezil/Memantine - Different once daily dosing depending on prior dose of donepezil or memantine (seems like want to slowly increase the memantine)
- Side effects: bradycardia, heart block, GI ulceration, N/V/D, bladder outflow obstructions

68
Q

What are the amyloid-targeted immunotherapies we use for AD? Side effects? What is needed before initiating treatment?

A

Aducanumab - Every 4 weeks
- Side effects: ARIA 40% -> requires MRI of brain w/in 1 year of starting treatment, then before 7th and 12th doses

Lecanemab - Every 2 weeks
- Side effects: ARIA 30% -> requires MRI of brain w/in 1 year of starting treatment, then before 5th, 7th, and 14th doses

**These require presence of amyloid beta pathology PRIOR to initiating treatment.

69
Q

How do we treat psychosis and depression in AD patients?

A

Psychosis -
- Use for psychosis or severe behavioral problems, NOT for behaviors like yelling or wandering
- Atypical antipsychotics, such as quetiapine and risperidone
- Increased risk of death or stroke in older adults with dementia if using these

Antidepressants -
- SSRIs usually first line, but avoid paroxetine
- Mirtazapine, venlafaxine, buproprion may also be considered

70
Q

What treatment is recommended for vascular dementia and lewy body dementia?

A

Vascular -
- Treat the vascular condition
- Cholinesterase inhibitors are recommended (donepezil, rivastigmine, galantamine)

Lewy body -
- Cholinesterase inhibitors and memantine MAY be helpful
- Very sensitive to side effects of antipsychotics (use quetiapine if needed)
- Visual hallucinations are common

71
Q

What are the definitions of seizure, convulsion, and epilepsy?

A

seizure - a sudden disorder of the CNS that is characterized by abnormal cerebral neuronal discharges with or without loss of consciousness.

convulsion - seizure type where the attack manifests as involuntary muscle contractions

epilepsy - repeated seizures due to damage, irritation, and/or chemical imbalance in the brain which leads to sudden, excessive, synchronous electrical discharge

72
Q

What is the psychological basis of a seizure? What are the potential harmful consequences of seizure activity? Do genetics play a part in seizures?

A

Seizures are a result of disordered, synchronous, and rhythmic firing of a population of brain neurons (aka synchronized hyperexcitability). It originates from the gray matter of any cortical or subcortical area of the brain. During a seizure, the brain is using more energy than it can manufacture, which is why prolonged seizures may result in cell ischemia.

  • Genetics plays a big part in seizures. (ex. SCN, GABR). Genetic testing for infantile epilepsy is becoming more of a standard diagnostic tool. It also gives us a genetic target to aim our treatment at
73
Q

What are the different seizure classifications and differences among different types of seizures (inc. differences in modes of propagation)?

A

Focal onset - Seizure starts in one area of the brain (ex. temporal lobe). Usually caused by a lesion (ex. head trauma, tumor, stroke, etc.). These frequently progress to generalized seizure, which is called ‘focal to bilateral’.
- Starts from a focal point and starts to spread and progress to the thalamus, which is when it turns bilateral.
- On an EEG, there are isolated hyperactivities
- Aware type - no loss of consciousness
- Impaired awareness type - clouding of consciousness

Generalized onset - Seizures that involve both hemispheres of the brain. There is a loss of consciousness. This is referred to as ‘primary’ or ‘idiopathic’ because it’s not due to a lesion. Most of these are presumed to be genetic.
- Starts somewhere in the connections between the thalamus and the cortex and projects into both hemispheres.
- On an EEG, all electrodes are hyperactive

74
Q

What are the definitions of aura and postictal state?

A

Aura - abdominal discomfort, sense of fear, unpleasant smell
- Due to abnormal electrical activity
- aware and impaired awareness type focal seizures experience this

Postictal state - the period after a seizure when the patient doesn’t know what’s going on. It can last for seconds to hours depending on the area of the brain affected, the length of seizure, use of anti-epileptic drugs, and age.
- Often see confusion, disorientation, and anterograde amnesia
- Can occur in an impaired awareness type seizure

75
Q

What are the characteristics of status epilepticus and what are the therapeutic goals when treating this state?

A

Status epilepticus - more than 5 mins of continuous seizure activity OR recurrent seizure activity w/o appropriate return to baseline

Goals of therapy -
- To bring the seizures under control within 60 minutes to minimize neurologic and CV complications. These complications include abnormal glucose utilization, compromised CNS blood flow, lactic acid accumulation, CV collapse (arrhythmia), long term impact on cognitive function, worsening of seizure disorder. All of these can end up leading to injured brain tissue.

76
Q

What is the meaning of ‘paroxysmal depolarizing shift’ and what is the electrophysiological basis of depolarization and hyperpolarization?

A

Paroxysmal depolarizing shift - Large depolarization that triggers a burst of action potentials. This depolarization involves glutamate activating AMPA and NMDA and voltage gated calcium channels, leading to an influx of cations that make the neuron more positive.

After this depolarization, there is hyperpolarization that involves the activation of GABA receptors (causing influx of Cl-) and voltage/calcium dependent K+ channels, that lead to an efflux of K+.

Clearly there has been some excitation/inhibition balance disruption to cause this.

77
Q

What is the electrophysiological basis of surround inhibition and the tonic and clonic phases of generalized tonic-clonic seizure?

A

Inhibitory surround - When electrical discharge spreads through the seizure focus, but is contained due to inhibition in a neighboring zone (the inhibitory surround) by GABAergic interneurons.

Focal-to-bilateral seizures happen due to a loss of hyperpolarization and surround inhibition.

Tonic phase - GABA inhibition disappears, glutamate AMPA & NMDA receptor activity increases (muscle contraction from all the excitation)

Clonic - GABA inhibition via Cl- gradually returns, leading to oscillations. (jerking stage)

78
Q

What underlying conditions or environmental perturbations can trigger epileptic seizures?

A

Underlying conditions:
- Prenatal injury, cerebrovascular disease, brain tumors, head trauma, infection, hemorrhage, anoxia, drugs

General perturbations:
- metabolic disturbances, like hyperventilation, blood gas, pH, hypoglycemia
- sleep deprivation
- stress
- alcohol withdrawal
- withdrawal from anti-epileptic drugs (especially when sudden)
- strobe lights

79
Q

What drugs (7) can aggravate or increase the risk of epileptic seizures?

A
  • alcohol
  • theophylline
  • CNS stimulants
  • bupropion
  • oral contraceptives
  • withdrawal from depressants
  • clozapine
80
Q

What are the types of generalized seizures? (ex. absense [typical and atypical], myoclonic, tonic, clonic, atonic, and tonic-clonic)

A

absense - can be typical (petit-mal) or atypical. Typical includes a brief loss of consciousness, but no convulsions, aura, or postictal period. Atypical has a slower onset than typical.

myoclonic - shock-like contraction of muscles. There is isolated jerking of the head, trunk, and body.

tonic - involves rigidity as a result of increased tone in the extensor muscles. These occur in children.

clonic - involve rapid, repetitive motor activity. These occur in babies and young children.

atonic - Sudden loss of muscle tone. A patient would fall if they were standing.

tonic-clonic (grand mal) - Tonic phase immediately followed by clonic phase. No aura. The tonic phase lasts 15-30 seconds and is then followed by the clonic phase for 1-2 minutes. After this, pt is usually stunned or confused. If the seizure started as a focal seizure, the patient may have had brief aura.

81
Q

What are the 4 molecular targets of anticonvulsant drugs?

A
  1. decrease sodium influx, prolong inactivation of Na+ channels
    - ex. carbamazepine, oxcarbazepine, phenytoin, lacosamide, lamotrigine, valproate
  2. reduction of calcium influx (critical for absence seizures)
    - ex. ethosuximide, lamotrigine, valproate
  3. enhance GABA-mediated neuronal inhibition
    - ex. barbiturates, benzodiazepines, valproate, gabapentin, bigabatrin, tiagabine
  4. antagonism of excitatory transmitters (glutamate)
    - ex. felbamate, topirimate

**levetiracetam has other targets

82
Q

Phenytoin for seizures: MOA, pearls, interactions, side effects

A

phenytoin - treats focal AND generalized
- MOA: binds and stabilizes the inactivated state of Na+ channels (binds in the brain and other parts of the body)
- Pearls: fosphenytoin has similar MOA and is an injectable prodrug. Elimination kinetics are dose dependent (non-linear kinetics)
- interactions: can be displaced from plasma proteins (ex. valproate). Also induces P450 enzymes
- side effects: arrhythmia, visual, ataxia, GI symptoms, gingival hyperplasia, hirsutism, hypersensitivity reactions

83
Q

Carbamazepine & oxcarbazepine for seizures: MOA, pearls, interactions, side effects

A

carbamazepine/oxcarbazepine - treats focal AND generalized
- MOA: binds and stabilizes the inactivated state of Na+ channels
- Pearls: oxcarbazepine has less toxicity than carbamazepine
- interactions: induces P450 enzymes, which reduces the rate of metabolism of itself
- side effects: blurred vision, ataxia, GI disturbances, sedation at high doses, SJS!!! DRESS!!

84
Q

lacosamide for seizures: MOA, pearls, interactions, side effects

A

lacosamide - treats focal AND generalized
- MOA: enhances inactivation of voltage-gated Na+ channels
- side effects: dermatological reactions, cardiac risks (PR interval prolongation), visual disturbances

85
Q

barbiturates: phenobarbital and primidone for seizures: MOA, pearls, interactions, side effects

A

phenobarb - treats focal AND generalized
- MOA: binds to allosteric site on GABAa receptor, which increases the duration of Cl- channel opening
- interactions: induces P450 enzymes
- side effects: sedation, physical dependence (abuse)

primidone - treats focal AND generalized
- MOA: similar to phenytoin (inactivation of Na+ channels)

86
Q

benzodiazepine - diazepam and clonazepam for seizures: MOA, pearls, interactions, side effects

A

diazepam - treats focal AND generalized
- MOA: binds to allosteric side of GABAa to increase frequency of Cl- channel opening
- Pearls: especially useful for tonic-clonic status epilepticus. Often administered as a rectal gel for acute control of seizure activity.
- side effects: sedation, physical dependence, not useful for chronic treatment

clonazepam -
- similar to diazepam
- useful for acute treatment of seizures and absence seizures

87
Q

gabapentin and pregabalin for seizures: MOA, pearls, interactions, side effects

A

gabapentin/pregabalin - treats focal AND generalized
- MOA: increases GABA release and decreases presynaptic Ca2+, which reduces glutamate release
- Pearls: used as adjunt for anti-seizure therapy
- side effects: sedation, ataxia, behavioral changes

88
Q

vigabatrin and tiagabine for seizures: MOA, pearls, interactions, side effects

A

vigabatrin - treats focal AND generalized
- MOA: IRREVERSIBLE inhibitor of GABA-T (GABA transaminase), which is the enzyme responsible for degrading GABA !!!
- Pearls: used as adjunct for refractory pts
- side effects: sedation, depression, visual field defects

tiagabine - treats focal AND generalized
- MOA: inhibits GAT-1 (GABA transporter) !!!!
- Pearls: used as adjunct therapy
- side effects: sedation, ataxia

89
Q

felbamate and topiramate for seizures: MOA, pearls, interactions, side effects

A

felbamate - treats focal AND generalized
- MOA: NMDA receptor antagonist
- Pearls: 3rd line drug for refractory cases (esp. focal)
- side effects: severe hepatitis (this is why it’s 3rd line)

topiramate - treats focal AND generalized
- MOA: AMPA and kainate receptor antagonist
- Pearls: used as monotherapy or adjunct
- side effects: confusion, cognitive dysfunction, sedation, vision loss

90
Q

What drugs are used to treat absence seizures only? What are their MOAs?

A

ethosuximide - blocks T-type Ca2+ channels in thalamic neurons. These T-type Ca2+ channels are thought to be involved in generating the rhythmic discharge of an absence attack.
- Side effects: GI distress, sedation, psychiatric disturbances

91
Q

What drugs are used to treat focal seizures, generalized tonic-clonic seizures, AND absence seizures? What are their MOAs?

A

clonazepam - binds to allosteric site of GABAa receptor
- side effects: sedation, physical dependence

lamotrigine - inhibits Na+ and Ca2+ channels. Also disrupts synaptic glutamate release
- side effects: sedation, ataxia, SJS!!

valproate - inhibits Na+ and Ca2+ channels. Also potentiates GABA signaling.
- interactions: displaces phenytoin. interferes with metabolism of phenytoin, lamotrigine, phenobarb, and carbamazepine
- side effects: GI distress, hyperammonemia, hepatotoxicity

levetiracetam - binds the synaptic vesicular protein SV2A, which interferes with synaptic vesicle release and neurotransmission. Also interferes with Ca2+ channels.
- Usually 2nd or 3rd line
- Can be used for refractory status epilepticus.

92
Q

What medications can lower the seizure threshold? Which ones at usual doses and which ones at high doses/impaired renal function? (BCTVPC, CLMPQT)

A

Usual doses:
Bupriopion
Clozapine
Theophylline
Varenicline
Phenothiazine antipsychotics
CNS stimulants

At high doses & impaired renal function:
Carbapenems
Lithium
Meperidine
Penicillin
Quinolones
Tramadol

93
Q

What do we use for quality of life monitoring when treating seizures? (7)

A
  • seizure frequency
  • functional status
  • social functioning (driver’s license)
  • emotional status (depression?)
  • cognition (many antiseizure meds can have cognitive side effects
  • number of doses of drug per day
  • cost of drug therapy
94
Q

What are the 9 risk factors for seizure recurrence?

A
  • less than 2 years seizure free
  • onset of seizures after age 12
  • history of atypical febrile seizures
  • 2-6 years before good seizure control in treatment
  • more than 30 seizures before control was achieved
  • partial seizures (most common type)
  • abnormal EEG throughout treatment
  • organic neurological disorder (ex. traumatic brain injury, dementia)
  • withdrawal of phenytoin or valproate
95
Q

What is drug-resistant epilepsy? What are the possible reasons for treatment failure? How can we manage it?

A

drug-resistant epilepsy: failure of at least 2 trials of antiseizure medication trails of adequate dose and duration

reasons -
1. failure to reach the CNS target
2. alternation of drug targets in the CNS
3. drugs missing the real target

management -
1. rule out pseudo-resistance (EEG normal, so something else is going on)
2. combination therapy
3. electrical/surgical intervention

96
Q

What is status epilepticus? What are the drug therapy options?

A

status epilepticus - continuous seizure for more than 5 minutes OR 2 or more discrete seizures with incomplete recovery between seizures

Treatment:
- Benzodiazepines!! IV lorazepam, IV/IM midazolam (or other nasal dosage forms)

97
Q

What are the boxed warnings and REMS programs associated with drug therapy for seizure disorders?

A

Carbamazepine or like derivatives - HLA-B*1502 allele -> anticonvulsant hypersensitivity syndrome

Lamotrigine - arrhythmia

perampanel - serious and/or life threatening neuropsychiatric events (don’t use with pre-existing psychosis, okay with depression/anxiety)

98
Q

What antiseizure drugs are teratogenic? Which drug is associated with a decrease in IQ of the offspring? What should be supplemented during pregnancy?

A

3A4 inducers are teratogenic - carbamazepine!!, phenytoin, phenobarbital, clonazepam, fosphenytoin, primidone, topiramate!!
*need extra contraception in these cases, since 3A4 also metabolized birth control

  • valproate is not recommended in pregnanct due to neural tube defects and it is also associated with decreased IQ in the offspring
  • supplemental folic acid 5mg daily should be considered during pregnancy
99
Q

What are some of the contraceptive drug interactions with antiseizure drugs?

A

P450 3A4 induction! estrogen compounds are major 3A4 substrates.

  • higher-dose estrogen contraceptives can minimize this, but there is increased thromboembolism risk
  • can use progestin only contraceptives
  • estrogen can significantly decrease lamotrigine serum concentration (50%) and lamotrigine decreases estrogen concentrations (bad two way street)
100
Q

What are the non-drug therapies for seizure disorders? When should non-drug therapies be recommended?

A

medical marijuana (cannabidol)- good for lennox-gastaut syndrome, dravet syndrome

keto diet - 3:1 or 4:1 fats:carbs/proteins
- side effects: hyperlipidemia, weight loss, constipation, kidney stones, decreased bone mass/growth
- adults only seem to respond while on the diet, but the effects in children may continue after the diet is discontinued.

101
Q

Why is depression such a problem in epilepsy?

A

All antiseizure drugs carry a warning for increased risk of suicidal thinking and/or behaviors during treatment.

Antidepressants also have increased risk of suicidal thinking and behaviors during treatment in patients under 24 years of age.

AVOID use of bupropion in patients with uncontrolled seizure disorders (it can increase risk of seizures and seizure frequency)

102
Q

What is the dose titration schedule for lamotrigine monotherapy, lamotrigine with valproic acid, and lamotrigine with carbamazepine?

A

VERY WORRIED ABOUT SJS!! lamotrigine is a UGT substrate

if no UGT drug interactions:
25mg once daily x 14 days
50mg once daily x 14 days
100mg once daily x 7 days
200mg once daily

UGT inducer - carbamazepine, phenytoin
50mg once daily x 14 days
100mg once daily x 14 days
200mg once daily x 7 days
400mg once daily

UGT inhibitor - valproate
25mg every other day x 14 days
25mg once daily x 14 days
50mg once daily x 7 days
100mg once daily

103
Q

Why do we need to monitor for HLA-B1502 and HLA-A3101 alleles when using certain anticonvulsants and antipsychotics? Who is this testing most important for?

A

worried about anticonvulsant hypersensitivty syndrome with carbamazepine, oxcarbazepine, eslicarbazepine, etc.

  • if pt has HLA-B*1502 allele, pt has HIGH risk for AHS
  • if positive, DO NOT use carbamazepine or like derivatives
  • common in pts of asian descent
  • need to screen for this
104
Q

How can we calculate loading doses for anticonvulsants for status epilepticus?

A

fosphenytoin - prodrug of phenytoin, better IV tolerance
20mg PE (phenytoin equivalents)/kg IV, may give additional dose 10 minutes after the load.
- Can give up to 150mg PE/minute IV infusion
(hypotension seen with phenytoin, which limits infusion rate)
*requires cardiac monitoring due to purple glove syndrome

valproate - The IV to PO conversion is 1:1mg/mg
- desired serum concentration is 80mcg/mL

105
Q

What are important phenytoin dosing considerations?

A

You MUST get both phenytoin serum concentration AND serum albumin in the same blood draw

therapeutic serum concentration range is 10-20mcg/mL

106
Q

What changes were made to the epilepsy classification by the ILAE from 2011 to 2017?

A

2011: partial (split into simple and complex) & generalized

2017: focal, generalized, unknown - based on where the seizure started and if they were aware

107
Q

Which antiseizure drugs are 1A2 inducers, 2C9 inducers, 3A4 inducers, and UGT inhibitors?

A

1A2 - CPP
- carbamazepine, phenobarbital, phenytoin

2C9 - CPP
- carbamazepine, phenobarbital, phenytoin

3A4 - CPPLOT (teratogenic)!
- carbamazepine, phenobarbital, phenytoin
- lamotrigine, oxcarbazepine, topiramate

UGT inhibitor -
- Valproate

108
Q

What seizure medications are associated with DRESS syndrome? When does it usually occur? What allele increases risk?

A
  • carbamazepine, phenobarbital, phenytoin, cenobamate, lamotrigine, valproate, zonisamide

This can be life threatening. 10% mortality rate.

Generally occurs 2-6 weeks after initiation of drug therapy or dose increase.

Pts with HLA-A*3101 allele are at higher risk (usually asian or northern european descent)

109
Q

What is antiseizure drug withdrawal syndrome?

A

when antiseizure drugs are discontinued, it may cause a recurrence of seizures. For this reason, the doses of antiseizure meds should always be tapered for discontinuation.

*additional withdrawal syndromes may include anxiety, agitation, and physical complaints

110
Q

What antiseizure drugs are associated with CV adverse effects? (arrhythmia, QTc shortening, PR interval changes, heart block, valvular heart disease)

A

arrhythmia - lamotrigine, phenytoin/fosphenytoin (contraindicated if heart block)

QTc shortening - rufinamide, cenobamate

PR interval changes - lacosamide, pregabalin

heart block - lacosamide

valvular heart disease - fenfluramine (pulled off the market)

111
Q

What antiseizure drugs are associated with electrolyte abnormalities, metabolic acidosis, and bone loss?

A

carbamazepine/eslicarbazepine/oxcarbazepine - hyponatremia, syndrome of inappropriate antiduretic hormone (SIADH)

phenytoin - altered vit D metabolism, decreased Ca2+ concentrations, which can lead to osteoporosis with long-term use

topiramate - oligohydrosis, heat intolerance, decreases serum bicarb which can lead to metabolic acidosis (monitor)

112
Q

What are the 4 antiseizure medications cause psychiatric side effects?

A

Levetiracetam - seen in children and adolescents most often. psychosis, suicidal thoughts/behaviors, unusual mood changes, worsening depression

Perampanel - boxed warning! serious and/or life threatening neuropsychiatric events

Valproate - acute mental status changes, often related to hyperammonemia (differentiate from sedation side effect)

Topiramate - cognitive dysfunction if the dose is increased too rapidly, so make sure to use a slow dose titration

*also tiagabine and cenobamate

113
Q

Which 2 antiseizure drugs are known to cause visual abnormalities?

A

topiramate - vision loss, myopia, retinal detachment

vigabatrin - contraindicated in patients who have other risk factors for irreversible vision loss.

114
Q

What is the biggest risk with gabapentin and pregabalin?

A

Respiratory depression.

Need to evaluate the appropriateness of gabapentin or pregabalin use in patients who are taking other CNS depressants, if they have pulmonary disease, or if they are eldery.

115
Q

What are the differences between migraine without aura vs. migraine with aura?

A

migraine without aura -
- at least five attacks with a heading lasting 4-72 hours
- unilateral, pulsating, moderate/sever pain, aggravation by routine activity
- nausea and/or vominting, photophobia, phonophobia

migraine with aura -
- at least two attacks
- at least one fully reversible aura
- no aura lasting more than 60 minutes, then headaches follow aura within 60 minutes
- aura symptoms: visual, sensory, speech/language, motor, brainstem, retinal

116
Q

What are the 4 migraine headache phases?

A
  1. prodrome - hours or days before headache onset. pt may experience euphoria, depression, irritability, food craving, constipation, etc.
  2. aura - (15-30% of pts) commonly visual, maybe sensory, verbal, or motor. Happens before or during the migraine, lasts less than 60 minutes. Photpsia (flash of light), scotoma (blind spot), zigzag lines, numbness/tinging in arms, less, face
  3. migraine headache - dull ache that intensifies. Unilateral and throbbing. Phonophobia and/or photophobia.
  4. postdromal - may last several days after the headache has ended. Tiredness, head pain, GI distress, mood changes, weakness, cognitive difficulties.
117
Q

What are some migraine triggers?

A

medication overuse - associated with analgesics (avoid overuse!), ergots, triptans (rebound headaches)

medications - oral contraceptives, hydralazine, nitroglycerin, nifedipine, cocaine

diet - chocolate, oranges, alcohol, caffeine, etc.

additives/preservatives - aspartame, monosodium glutamate

environment - altitude/weather changes, perfumes, tobacco smoke, loud noises, flickering light, etc.

others - too much/little sleep, skipping meals, stress, hormone changes

118
Q

When do we use abortive vs preventative treatment for migraines?

A

abortive - when the migraine starts. It’s important not to overuse abortive medicines.

preventative - use when pt is having four or more attacks/month with a disability at least 3 days/month

119
Q

What are the first and second line drugs used for abortive treatment in migraines?

A
  1. triptans, NSAIDs (ASA, diclofenac, ibuprofen, naproxen)
  2. NSAIDs (ketoprofen, IV and IM ketorolac, flurbiprofen)
120
Q

What are the first and second line drugs used for prevention treatment in migraines?

A
  1. valproate, topiramate, metoprolol, propranolol
  2. amitriptyline

*monotherapy

121
Q

What non-pharm therapy is available for migraines?

A
  • biofeedback and relaxation therapy
  • cognitive behavioral therapy
  • diet (eliminate triggers)
  • sleep (consistency)
  • TENS (transcutaneous electrical nerve stimulation) device
  • exercise, massage, acupuncture, thermal biofeedback, heat/cold applications
  • headache diary
122
Q

What contraindications and warnings/precautions are there for triptans for abortive treatment of migraines? If pt has n/v, how does that change the treatment?

A

Contraindications - use of an ergot or other triptan within 24 hours, use of MAO-A inhibitor in the last 2 weeks (rizatriptan, frovatriptan), ischemic heart disease, angina, hx of stroke or TIA, hemiplegic/basilar migraine, arrhythmias, peripheral vascular disease, uncontrolled HTN, ischemic bowel disease, severe hepatic impairment

warning/precautions - MI, pain/pressure/tightness in chest/throat/neck/jaw, CVA, HTN, GI ischemic reactions or peripheral vasospasm, medication overuse headache, serotonin syndrome

*if nausea/vomiting is a common symptom, use non-oral triptan and consider anti-emetic

123
Q

What is the most important drug interactions with triptans for abortive treatment of migraines?

A

SSRIs/SNRIs - increased risk of serotonin syndrome. This is extremely rare.

also MAO inhibitors

124
Q

What is an important drug interaction with ergot alkaloids?

A

3A4 inhibitors can cause ergot toxicity. This is bad because it can cause ergotism, which results in cramps, spasms, etc.

There are also vascular complications seen with ergot alkaloids.

125
Q

When using rimegepant and ubrogepant for abortive treatment (3rd line), what are the important drug interactions/contraindications? What’s the main side effect?

A

These are CGRP antagonists, which blocks neurogenic inflammation.

Contraindicated with strong 3A4 inhibitors!!
Also pgp inhibitors.

side effect: nausea

126
Q

what is lasmiditan used for? what are the precautions?

A

abortive therapy for migraines.

Warning/precautions - medication overuse headaches, sedation, dizziness, serotonin syndrome, driving impairment

127
Q

what is butorphanol nasal spray used for?

A

it can be used as a rescue medication for abortive therapy in migraines

128
Q

which beta blockers and tricyclic antidepressants can be used for preventative therapy in migraines?

A

beta blockers - propranolol, metoprolol, timolol
- contraindicated in asthma and Raynaud’s syndrome

tricyclic antidepressants - amitriptyline

129
Q

which CGRP antagonist can be used for preventive treatment in migraines?

A

atogepant

130
Q

which antiseizure drugs can be used for preventative drug therapy in migraines?

A

valproate, topiramate

**not recommended in women of child-bearing age unless using contraception (3A4 inducer & teratogenic)

131
Q

what is the one natural product that has shown to work for preventative therapy in migraines?

A

butterbur/petasites 150mg/day

132
Q

Which 6 drugs are okay to use in children for migraines? Which drug is the only one indicated for children down to 6 years old?

A

almotriptan - 12 years+
zolmitriptan nasal spray - 12 years+
sumatriptan nasal spray - not FDA approved
sumatriptan/naproxen - 12 years+
topiramate - 12 years +

rizatriptan - 6 years+!!