CCPP Module 30, Section 2: Psychiatric Pharmacogenomics Applications
MODULE 30: PHARMACOGENOMICS IN DISEASE-STATE MANAGEMENT

Section 30.2: Psychiatric Pharmacogenomics Applications

Exploring the use of multi-gene PGx panels to guide antidepressant and antipsychotic selection, moving beyond trial-and-error prescribing for conditions like major depressive disorder and schizophrenia.

SECTION 30.2

Psychiatric Pharmacogenomics Applications

From Trial-and-Error to Genetically-Informed Precision Prescribing.

30.2.1 The “Why”: Ending the Agonizing Cycle of Trial-and-Error in Mental Health

As a pharmacist, you have been on the front lines of the mental health crisis. You have filled prescriptions for antidepressants and antipsychotics, provided counseling on their delayed onset of action, and managed the frustrating and often debilitating side effects that accompany them. More than anyone, you have witnessed the painful and inefficient process of “trial-and-error” prescribing that has defined psychopharmacology for decades. A patient with major depressive disorder (MDD) might try one SSRI for 6-8 weeks, only to find it ineffective. They then switch to another, wait another two months, and perhaps experience intolerable side effects. This cycle can repeat for months, or even years, leaving the patient feeling hopeless and the provider feeling helpless. The statistics are sobering: up to 50% of patients with MDD do not respond to their first-line antidepressant, and many will try multiple medications before finding one that provides even partial relief.

This is not a failure of the drugs themselves, nor is it a failure of the providers. It is a failure of the “one-size-fits-all” model in a field where individual biological variability is the rule, not the exception. The neurochemical pathways our psychiatric medications target are profoundly complex, and the enzymes that metabolize these drugs are notoriously variable from person to person. Two patients with identical symptoms can have wildly different responses to the same dose of sertraline, not because of their willpower or the severity of their illness, but because of unseen variations in their genetic code that dictate how the drug is processed in their bodies.

Psychiatric pharmacogenomics offers a powerful beacon of hope to break this cycle. By analyzing key genes involved in drug metabolism (pharmacokinetics) and drug targets (pharmacodynamics), we can get a biological “preview” of how a patient is likely to respond to a medication. This is not a crystal ball that perfectly predicts the single “best” drug. Rather, it is an invaluable decision-support tool that helps us build a more intelligent list of options. It allows us to identify, with a high degree of confidence, which medications are likely to have abnormal metabolism, leading to a higher risk of side effects or a higher likelihood of therapeutic failure. By ruling out these genetically mismatched options from the start, we can significantly increase the probability of choosing a medication that is both safe and effective on the first or second try.

Your role in this new paradigm is indispensable. You are the medication expert who can bridge the gap between a complex genetic report and a practical clinical decision. You will be the one to translate a `CYP2D6` diplotype into a specific dosing recommendation for an antipsychotic, or to explain to a provider why their patient’s `CYP2C19` status makes citalopram a potentially risky choice. In this section, we will equip you with the knowledge and tools to become a leader in psychiatric PGx. We will move beyond single gene-drug pairs and explore the use of comprehensive multi-gene panels to create a holistic, personalized medication strategy for patients suffering from mental illness. This is your opportunity to help transform psychiatric care from an art of guesswork into a science of precision.

Pharmacist Analogy: Tuning a High-Fidelity Radio

Think of a patient’s brain as a complex, high-fidelity radio receiver, and each antidepressant or antipsychotic as a specific radio station broadcasting on a unique frequency. The goal of treatment is to find the station that comes in “loud and clear” for that individual patient, providing a therapeutic signal without static (side effects).

  • Traditional Prescribing is like tuning an old analog radio in the dark. You turn the dial slowly (trying one drug), listen for a while (wait 6-8 weeks), and if all you get is static or a weak signal (side effects or no efficacy), you turn the dial again, hoping to stumble upon the right frequency. It’s a slow, frustrating process based entirely on guesswork.
  • A Pharmacogenomic Test is like getting the owner’s manual for that specific radio. The manual doesn’t tell you which station broadcasts the “best” music (the most effective drug), but it gives you critical technical specifications.
    • The `CYP2D6` and `CYP2C19` results are like the radio’s “Antenna and Signal Processor” report. It tells you which frequencies (drugs) the radio is built to receive well. A “Poor Metabolizer” result is like a note saying, “This radio’s processor will be overwhelmed by signals on the 98.7 FM frequency (e.g., paroxetine), causing loud, distorted static (toxicity).” An “Ultra-rapid Metabolizer” note might say, “The antenna for the 101.5 FM frequency (e.g., sertraline) is too efficient; the signal will be processed and discarded so quickly it will be barely audible (lack of efficacy).”
    • The `SLC6A4` or `HTR2A` results are like the “Speaker Quality” report. It might indicate that even if a signal is received perfectly, the speakers (receptors) are predisposed to distorting certain bass frequencies, leading to undesirable audio artifacts (side effects like weight gain or sexual dysfunction).

As the pharmacist with the PGx “manual,” you are no longer guessing. You can now advise the provider: “Based on the manual, this patient’s radio is poorly equipped to handle the 98.7 FM station. However, it’s perfectly built to receive the 103.3 FM station (e.g., a different medication). Let’s start there. We have a much higher chance of getting a clear signal from the beginning.”

30.2.2 The Key Players: A Deep Dive into the Genes on a Psychiatric PGx Panel

Unlike in cardiology where we often focus on a single gene for a single drug, psychiatric PGx almost always involves a multi-gene panel. This is because the response to these medications is complex, involving both metabolism (pharmacokinetics) and the drug’s effect at the receptor level (pharmacodynamics). A comprehensive panel provides a more complete picture. Let’s break down the most important genes you will encounter.

The Pharmacokinetic Powerhouses: `CYP2D6` and `CYP2C19`

These two cytochrome P450 enzymes are the undisputed stars of psychiatric PGx. Together, they are responsible for the metabolism of approximately 70-80% of all commonly prescribed psychotropic medications. An individual’s genetically determined function of these two enzymes is the single most powerful predictor of their risk for side effects and therapeutic failure.

`CYP2D6`: The Most Important Enzyme in Psychopharmacology

`CYP2D6` is involved in the metabolism of roughly 25% of all drugs on the market, but its impact is especially profound in psychiatry. It metabolizes many tricyclic antidepressants (TCAs), many SSRIs and SNRIs, and the vast majority of antipsychotics. The `CYP2D6` gene is extremely polymorphic, with over 100 known alleles, leading to a wide spectrum of enzyme activity.

`CYP2D6` Phenotype Metabolic Capacity Example Genotypes Clinical Implication for a `CYP2D6` Substrate
Poor Metabolizer (PM) Absent enzyme function. `*4/*4`, `*4/*5`, `*5/*5` High risk of toxicity. Drug accumulates to dangerously high levels at standard doses. Leads to severe side effects. Requires significant dose reduction or avoidance of the drug.
Intermediate Metabolizer (IM) Reduced enzyme function. `*1/*4`, `*1/*41`, `*4/*10` Increased risk of side effects. Drug levels will be higher than expected. Often requires a dose reduction (e.g., 25-50% of standard dose) and careful titration.
Normal Metabolizer (NM) Normal, expected enzyme function. `*1/*1`, `*1/*2` Expected response. Standard dosing is appropriate.
Ultra-rapid Metabolizer (UM) Increased enzyme function due to gene duplication. `*1/*1xN`, `*1/*2xN` (where N > 2) High risk of therapeutic failure. Drug is cleared so rapidly that it may not reach therapeutic concentrations at standard doses. Requires a significant dose increase or avoidance of the drug.
Critical Safety Alert: TCAs and `CYP2D6` Poor Metabolizers

Tricyclic antidepressants like nortriptyline, amitriptyline, and desipramine have a narrow therapeutic index and are major substrates of `CYP2D6`. Administering a standard dose of a TCA to a `CYP2D6` Poor Metabolizer is extremely dangerous. The drug will accumulate to toxic levels, leading to a high risk of cardiotoxicity (QTc prolongation, arrhythmias) and anticholinergic crisis. The CPIC guidelines have a “Red Light” recommendation for this combination, stating that alternative agents not metabolized by `CYP2D6` should be used. This is one of the most critical, life-saving interventions a pharmacist can make with a PGx report.

`CYP2C19`: The SSRI Specialist

While `CYP2D6` gets much of the attention, `CYP2C19` is equally critical, particularly for some of the most widely used antidepressants in the world, including citalopram, escitalopram, and sertraline. It also metabolizes several TCAs. The phenotypes for `CYP2C19` mirror those of `CYP2D6`.

`CYP2C19` Phenotype Clinical Implication for Citalopram / Escitalopram
Poor Metabolizer (PM) High risk of QTc prolongation. Citalopram and escitalopram levels can increase significantly, raising the risk of cardiac arrhythmias. The FDA and CPIC recommend a 50% dose reduction or choosing an alternative agent. For citalopram, the maximum recommended dose is 20 mg/day.
Intermediate Metabolizer (IM) Increased risk of side effects. Drug levels may be elevated. Consider a 25% dose reduction or slower titration. Careful monitoring is advised.
Normal Metabolizer (NM) Expected response. Standard dosing is appropriate.
Ultra-rapid Metabolizer (UM) High risk of therapeutic failure. The drug may be cleared too quickly to be effective. CPIC recommends selecting an alternative agent not primarily metabolized by `CYP2C19` (e.g., paroxetine, fluvoxamine).

The Pharmacodynamic Genes: A More Nuanced Picture

While the pharmacokinetic genes tell us about drug *exposure*, pharmacodynamic genes give us clues about the drug’s *effect* at its target. The evidence for these genes is generally less direct than for metabolism, but they can provide valuable secondary insights, especially when choosing between mechanistically similar drugs.

  • `SLC6A4` (Serotonin Transporter): This gene codes for the serotonin transporter (SERT), the primary target of SSRIs. A polymorphism in the promoter region of this gene, known as `5-HTTLPR`, exists as a “short” (S) or “long” (L) allele. For many years, it was thought that individuals with one or two copies of the S allele were less likely to respond to SSRIs. Current View: The evidence for this has become much weaker and more controversial over time. Most modern PGx panels report this but advise caution in its interpretation. It should not be used as the sole reason to avoid an SSRI but can be a “tie-breaker” when choosing between mechanistically different classes of antidepressants in a patient who has failed multiple SSRIs.
  • `HTR2A` / `HTR2C` (Serotonin Receptors): Variations in these genes, which code for serotonin receptors, have been associated with the risk of side effects from antidepressants and antipsychotics. For example, certain variants in `HTR2C` have been linked to a higher risk of antipsychotic-induced weight gain. This information can be useful in selecting an agent with a lower metabolic risk profile (e.g., aripiprazole, lurasidone) for a genetically predisposed patient.
  • `MTHFR` (Methylenetetrahydrofolate Reductase): This is not a classic PGx gene, but it’s often included on panels. `MTHFR` is a key enzyme in folate metabolism, which is essential for the synthesis of neurotransmitters (serotonin, dopamine, norepinephrine). Patients with reduced `MTHFR` function may have lower levels of the active form of folate (L-methylfolate) in the brain. This has been linked to a poorer response to antidepressants. Clinical Action: For a patient with MDD and a reduced-function `MTHFR` variant, adding L-methylfolate (Deplin) as an adjunct to their antidepressant may improve their response.

30.2.3 Masterclass Case Study 1: Treatment-Resistant Depression

Patient: Ms. Sarah Jenkins, a 34-year-old female with a 5-year history of Major Depressive Disorder (MDD). She presents to a new psychiatrist for consultation.

Medication History:

  • Sertraline (Zoloft): Tried for 12 weeks, titrated up to 200 mg/day. Reported zero improvement in mood, but significant insomnia and agitation.
  • Venlafaxine (Effexor XR): Tried for 10 weeks, titrated to 150 mg/day. Discontinued due to extreme nausea and a “racing heart.”
  • Bupropion (Wellbutrin XL): Currently taking 300 mg daily. Reports it helps with energy but provides minimal benefit for her core depressive symptoms of sadness and anhedonia.

The Dilemma: The patient is frustrated and feels “treatment-resistant.” The psychiatrist is considering augmenting the bupropion with another agent or switching entirely, but is unsure which direction to go. They order a comprehensive psychiatric PGx panel to guide the next choice.

The PGx Report Highlights:
Gene: `CYP2D6` | Genotype: `*4/*5` | Predicted Phenotype: Poor Metabolizer
Gene: `CYP2C19` | Genotype: `*1/*17` | Predicted Phenotype: Rapid Metabolizer
Gene: `SLC6A4` | Genotype: S/S | Predicted Phenotype: Lower likelihood of response to SSRIs

The Pharmacist’s Tutorial: Solving the Puzzle

As the consulting pharmacist, this PGx report is a Rosetta Stone that explains Ms. Jenkins’ entire medication history. Let’s deconstruct it piece by piece.

  1. Why did Sertraline fail? Sertraline is primarily metabolized by `CYP2C19`. Ms. Jenkins is a `CYP2C19` Rapid Metabolizer (`*1/*17`). This means she was likely clearing the drug from her system so quickly that it never reached a stable, therapeutic concentration in her brain, even at the maximum dose of 200 mg. The agitation she experienced could have been from the rapid fluctuations in drug levels. The `SLC6A4` S/S genotype might also have contributed to the lack of efficacy. Conclusion: The failure was pharmacokinetic, not a failure of the SSRI mechanism itself.
  2. Why did Venlafaxine cause toxicity? Venlafaxine is primarily metabolized to its active metabolite, desvenlafaxine, by `CYP2D6`. Ms. Jenkins is a `CYP2D6` Poor Metabolizer (`*4/*5`). This means she has virtually no functional `CYP2D6` enzyme. When she took venlafaxine, she was unable to metabolize it effectively. The parent drug likely accumulated to toxic levels, causing the nausea and tachycardia (a known side effect of noradrenergic overstimulation). Conclusion: A clear case of genetically-driven adverse drug reaction.
  3. Why is Bupropion partially effective? Bupropion is metabolized by `CYP2B6` (not usually on standard panels) to its active metabolite, hydroxybupropion. Critically, it is not a substrate of `CYP2D6` or `CYP2C19`. Therefore, her genetic variations did not significantly impact its metabolism, allowing her to tolerate it and receive some benefit.
Formulating the Recommendation: A Genetically-Informed Strategy

You now have the data to provide a powerful, evidence-based recommendation to the psychiatrist.

  • Step 1: Identify Drugs to Avoid. Based on her `CYP2D6` Poor Metabolizer status, you create a “Red Light” list. This includes most TCAs (nortriptyline, desipramine), and SSRIs/SNRIs that are heavy `CYP2D6` substrates like paroxetine, fluvoxamine, and duloxetine. Venlafaxine is already on this list from her experience.
  • Step 2: Identify Drugs to Use with Caution. Based on her `CYP2C19` Rapid Metabolizer status, you create a “Yellow Light” list. This includes citalopram, escitalopram, and sertraline. While they could theoretically be used at very high doses, it’s more logical to choose an agent that doesn’t have this built-in pharmacokinetic problem.
  • Step 3: Identify a “Green Light” Path Forward. You need to find a medication that avoids both `CYP2D6` and `CYP2C19` metabolism.
    • Mirtazapine (Remeron): An excellent choice. It works via a different mechanism (alpha-2 antagonist) and is metabolized by multiple enzymes (`CYP1A2`, `CYP3A4`, `CYP2D6`), so a deficiency in one pathway (`CYP2D6`) is less impactful. It would also be sedating, which could help her sertraline-induced insomnia. Augmenting her current bupropion with mirtazapine (a combination sometimes called “California Rocket Fuel”) is a well-established strategy.
    • Desvenlafaxine (Pristiq): Another excellent choice. Desvenlafaxine is the active metabolite of venlafaxine. By giving the active metabolite directly, you completely bypass the need for `CYP2D6` metabolism. This would give her the benefits of an SNRI without the toxicity she experienced with the parent drug.
    • Vortioxetine (Trintellix): A newer agent with multimodal serotonergic activity. While it is a `CYP2D6` substrate, the CPIC guidelines provide a clear recommendation for PMs: “Consider a 50% reduction of the recommended starting dose and titrate to response, maximum dose 10 mg/day.” This makes it a viable, genetically-guided option.

The Final Recommendation (SBAR): “Dr. Smith, the PGx results for Sarah Jenkins provide a clear explanation for her past medication failures. She is a `CYP2D6` Poor Metabolizer, which explains the venlafaxine toxicity, and a `CYP2C19` Rapid Metabolizer, explaining the sertraline failure. (A) Given this profile, I would strongly advise against using TCAs, paroxetine, or duloxetine. (R) A safer and more effective strategy would be to either augment her current bupropion with a low dose of mirtazapine, or to switch her from bupropion to either desvenlafaxine 50 mg daily or vortioxetine starting at 5 mg daily. Both options bypass her key genetic defects.”

30.2.4 Masterclass Case Study 2: First-Episode Psychosis

Patient: Mr. Kevin Johnson, a 22-year-old male, is brought to the inpatient psychiatric unit by his family after experiencing several weeks of paranoid delusions, auditory hallucinations, and social withdrawal. He is diagnosed with schizophrenia.

The Clinical Challenge: The treatment team needs to select a first-line second-generation antipsychotic (SGA). The goals are to rapidly control his psychotic symptoms while minimizing the risk of debilitating side effects like extrapyramidal symptoms (EPS), metabolic syndrome, and hyperprolactinemia, which are major drivers of non-adherence in this population. The hospital has a PGx program for all patients started on antipsychotics.

The PGx Report Highlight:
Gene: `CYP2D6` | Genotype: `*1/*2×2` | Predicted Phenotype: Ultra-rapid Metabolizer

The Pharmacist’s Tutorial: Pre-emptive Strike Against Treatment Failure

This is a perfect example of how PGx can be used proactively to prevent a negative outcome.

  1. Identify the Risk: The patient is a `CYP2D6` Ultra-rapid Metabolizer. This means he has an extra, functional copy of the `CYP2D6` gene and will clear `CYP2D6` substrates from his body at a much faster rate than normal.
  2. Analyze the Options: Many of the most common first-line SGAs are major `CYP2D6` substrates.
    • Risperidone (Risperdal): Extensively metabolized by `CYP2D6` to its active metabolite, paliperidone. In a UM, risperidone would be rapidly converted and cleared, likely leading to sub-therapeutic levels and poor efficacy.
    • Aripiprazole (Abilify): Also a major `CYP2D6` substrate. A standard dose in a UM would likely result in therapeutic failure.
  3. Consult the Guidelines: You turn to the CPIC guidelines for antipsychotics. For `CYP2D6` Ultra-rapid Metabolizers and aripiprazole, the guideline gives two options: 1) Consider an alternative drug not metabolized by `CYP2D6`, or 2) Consider increasing the starting dose by 1.5 to 2-fold, with careful monitoring.
  4. Formulate the Recommendation: You have several excellent, genetically-informed paths forward.
    • The “Dose-Increase” Strategy: You could recommend starting aripiprazole, but at a higher-than-usual dose, for example, 15 mg/day instead of the typical 10 mg. This is a reasonable option.
    • The “Alternative Drug” Strategy: This is often the safer and more straightforward approach. You need to identify SGAs that are not primarily `CYP2D6` substrates.
      • Olanzapine (Zyprexa): Metabolized by `CYP1A2` and UGT enzymes. A great choice from an efficacy standpoint, but carries a high risk of metabolic side effects (weight gain, diabetes).
      • Paliperidone (Invega): This is the active metabolite of risperidone. It is primarily cleared renally and is not dependent on `CYP2D6`. By giving paliperidone directly, you completely bypass the patient’s genetic variability. This is an elegant and highly effective solution.
      • Lurasidone (Latuda): Metabolized by `CYP3A4`. Another excellent option, with a lower metabolic risk profile than olanzapine.
  5. The Final Recommendation (SBAR): “Dr. Lee, I’m calling about the new antipsychotic order for Kevin Johnson. His PGx results are back, and he is a `CYP2D6` Ultra-rapid Metabolizer. (A) This means he will clear drugs like risperidone and aripiprazole very quickly, putting him at high risk for treatment failure at standard doses. (R) To avoid this, the CPIC guidelines suggest either a significant dose increase or an alternative agent. To simplify treatment and ensure efficacy, I recommend we start a drug that bypasses `CYP2D6` metabolism. Paliperidone 3-6 mg daily would be an excellent choice, as would lurasidone. This should give us a much better chance of achieving a therapeutic response from the outset. Shall I enter an order for paliperidone?”