Expired activity
Please go to the PowerPak homepage and select a course.

» Download PDF

Introduction: Pharmacist’s Expanding Role in HIV Prevention

Human immunodeficiency virus (HIV) preexposure prophylaxis (PrEP) remains underutilized in the United States.1 As 85% to 90% of PrEP medications are filled at community pharmacies, pharmacists have the capability to join others in ending the HIV epidemic by 2030.2,3 Pharmacists have legal authority in most states to prescribe and dispense PrEP in collaboration with other clinicians.4 Recently, some states (including California, Colorado, Nevada, Oregon, and Virginia) have granted pharmacists legal authority to prescribe and dispense PrEP independently.5 Pharmacists could facilitate PrEP uptake and adherence through consultations with and HIV screening for interested patients, point-of-care testing for HIV and other sexually transmitted infections (STIs), PrEP prescriptions, and PrEP adherence counseling.6 Pharmacy-based interventions such as refill reminders and adherence counseling have improved adherence to antiretroviral therapy (ART) regimens among people living with HIV (PLWH).7-9 Because interventions that integrate pharmacists into the PrEP care continuum are increasing, pharmacists must have a thorough understanding of PrEP as well as the essential roles that they serve when caring for patients who may be at risk of acquiring HIV.10,11

HIV Epidemiology and the Burden of Disease

Approximately 1.2 million people in the United States are living with HIV, and 1 in 7 individuals infected with HIV are unaware of their infection. Latest estimates from the Centers for Disease Control and Prevention (CDC) indicate that there were 36,740 new HIV infections in the United States in 2019.12 Populations including men who have sex with men (MSM), transgender persons, and black and Hispanic/Latinx individuals continue to be disproportionally affected. While new annual infections in the United States have been reduced by more than two thirds since the mid-1980s when HIV initially emerged, black and Hispanic/ Latinx populations are among those that continue to have high rates of undiagnosed infections (Figure 1).13-15

HIV burden has often been measured by the number of individuals dying from acquired immunodeficiency syndrome (AIDS)-related complications. Although on an international level, HIV/AIDS remains among the leading fatal infectious diseases, AIDS-related deaths in the United States have dramatically dropped because of advances in HIV treatment and management.16,17 Nonetheless, the United States government continues to spend more than $20 billion annually in direct health expenditures related to HIV care and prevention. This underscores HIV as a persistent public health threat despite greater disease awareness, highly effective ART, and proven prevention strategies and tactics.3

The advent and continual evolution of ART has allowed people with HIV (PWH; or people living with HIV [PLWH]) to enjoy longer, healthier lives through reduction in HIV-related morbidity and mortality at all stages of HIV infection.18 ART has also given rise to modalities designed to limit HIV acquisition in HIV-negative individuals—notably, specific antiretroviral drug combinations used as PrEP and postexposure prophylaxis (PEP). PrEP is prescribed for HIV-negative people who are at high ongoing risk of HIV exposure, whereas PEP is prescribed for HIV-negative people who have had a single high-risk exposure.19

Despite oral PrEP demonstrating efficacy in reducing HIV transmission rates by as much as 74% to 99% when taken daily (depending on mode of transmission—sexual or injection drug use [IDU]), fewer than 1 in 4 potential candidates receive PrEP (Figure 2).20,21,23

High-risk populations with the lowest rates of PrEP uptake include young MSM, racial or ethnic minority groups, and individuals living in the South or Southeast regions of the United States. Without preventive efforts, 1 in 2 black MSM and 1 in 5 Hispanic/Latinx MSM will contract HIV in their lifetimes.24,25 The majority of PrEP uptake (75%) has been among white gay and bisexual men, predominantly those living in the Northeast or on the West Coast.26 As of 2017, the PrEP-to-Need ratio (ie, the number of PrEP users divided by new HIV diagnoses) was lowest among individuals aged ≤24 years or ≥55 years. Geographically, lowest ratios were seen throughout the South, despite 52% of new HIV diagnoses in the United States being attributable to this region (Figure 3).12

Perceived stigma, distrust of the medical system, lack of PrEP awareness among patients and clinicians, and limited access due to cost are theorized as potential reasons why PrEP uptake is currently low. Distrust of the medical system may be tied to stigma, as patients are often reluctant to speak with their health care providers about their sexual activity or behaviors of increased risk. This is especially true among MSM.27

Further, clinicians may be uncomfortable with sexual history-taking and unfamiliar with baseline laboratory testing and how to initiate and monitor patients on PrEP. In addition, they may not realize that a less costly generic PrEP formulation is now available.28 In many locales, required prior authorization has been one of the most common barriers to prescribing PrEP, according to primary care and HIV-treatment providers.24 Without adequate insurance coverage, the annual cost of continuous PrEP use (including guideline-based interval visits, laboratory testing, and prescription refills) is more than $10,000. Although there are federal and state-level programs to increase access to PrEP, cost and lack of awareness of PrEP resources may be barriers for young black and Latinx MSM and transgender women who may benefit the most from PrEP, many of whom are likely to be uninsured or underinsured.26

Building on an Established Practice Foundation

Through expansion of scopes of practice, pharmacists are demonstrating their ability to work in chronic disease management and to dispense or administer certain drugs and biologic products.25,29 Many states, in an effort to curb the soaring rates of drug overdose, now allow pharmacists to dispense naloxone without a prescription.26,30 Frequently, pharmacists in patient-centered medical homes lead medication-management services. This entails helping patients meet clinical-management goals as diverse as control of glycated hemoglobin (HbA1c), systolic blood pressure, and serum lipids as well as increasing vaccination rates and medication adherence.31 In certain states, upon meeting additional education, credentialing, or board recognition, pharmacists are authorized to independently prescribe or furnish and dispense contraceptives, tobacco cessation therapy, and travel medications.24,29 Further, community pharmacists are extremely well-positioned to address certain public health problems, since high-risk patients visit their community pharmacies more often than their primary care providers (PCPs) and may be more comfortable in a pharmacy setting.31

Within the realm of HIV disease management in an HIV clinic setting, pharmacists are part of the HIV care team. They have already demonstrated high levels of success in recommending HIV regimen starts and modifications, increasing ART adherence via identification of patient-specific barriers (eg, health literacy, cognitive factors, mental-health and substance-use comorbidities, economic factors); detecting therapy-related barriers (eg, adverse effects, drug-drug interactions); and recognizing service-delivery barriers (eg, patient-provider relationships, providers’ cultural competencies). Pharmacists also expand access to these medications by assisting in prior authorization, activating copayment cards, patient assistance programs, state AIDS Drug Assistance Programs (ADAP), or identifying other resources to assist the patient in securing access to PrEP.32

An obvious next step in expanding the pharmacist’s role in HIV care is the prescribing and management of PrEP and PEP. Experienced in the principles and practices of chronic-disease medication management, and knowledgeable about HIV pharmacology, the pharmacist can play an essential role in HIV prevention across diverse practice venues.33-35

HIV Prevention and the Evolving Scope of Pharmacy Practice35

One state that recently increased the scope of practice for pharmacists is Colorado. Despite a relatively low HIV incidence, new cases in 2019 reflected an approximate 13% increase over cases reported in 2018.36 This contrasts with overall US HIV incidence rates, which remained stable or showed slight decreases between 2014 and 2018.37

Responding to this public health imperative to reduce HIV incidence in Colorado, legislation to expand PrEP and PEP access through inclusion of pharmacists as providers of HIV prevention passed both the State Senate and House in June 2020. Table 1 shows what is mandated in Colorado and may serve as an example for others interested in advocating in their own states.29,38

Other states have also passed new laws granting community pharmacists the authority to prescribe HIV PrEP medications. For example, in 2019, California passed a landmark bill, Senate Bill 159, that allows pharmacists to initiate PrEP and PEP. Pharmacists can initiate between a minimum of a 30- to 60-day supply of PrEP, provided that the patient has been tested and is in accordance with the guidelines. Pharmacists are required to complete a 90-minute state-required training prior to initiating PrEP. Moreover, this bill eliminated prior authorization requirements for PrEP in the state of California.39 States like California and Colorado may serve as models for other states interested in authorizing pharmacists to prescribe HIV PrEP medications in the future.

HIV Basic Science for the Pharmacist

Therapeutic Interventions in the HIV Viral Life Cycle

HIV targets and replicates within CD4 T-lymphocytes (T helper cells)—cells that are critical to adaptive immune responses—leading to CD4 destruction and, thereby, decreasing the functionality of the immune system and increasing the risk of opportunistic infections.40,41

The HIV life cycle (Figure 4) consists of 7 stages, 5 of which inform the mechanisms of action of current ARTs (Table 2).42 The first stage is binding (or attachment): HIV binds to receptors on the surface of a CD4 cell. The second stage is fusion: upon attachment, the viral envelope fuses with the CD4 cell membrane, allowing HIV to enter the host cell and release viral components. Stage 3 is reverse transcription: inside the CD4 cell, HIV releases a reverse transcriptase enzyme to convert HIV RNA (ribonucleic acid) into HIV DNA (deoxyribonucleic acid). HIV DNA then enters the nucleus and proceeds to integration (stage 4), during which HIV releases integrase, an enzyme that integrates viral DNA into the DNA of the host CD4 cell. In stage 5, replication, the machinery of the host CD4 cell produces long chains of HIV proteins (building blocks for more HIV); while in stage 6, assembly, new HIV proteins and HIV RNA move to the surface of the cell and combine as immature (noninfectious) HIV. The seventh and final step in the HIV life cycle is budding, wherein newly formed immature HIV pushes itself out of the host CD4 cell. Protease, an HIV enzyme released from the new HIV, breaks up long protein chains in the immature virus, creating a new and mature (infectious) virus.42

The Immunologic Basis for HIV Testing

Following release of mature HIV from the CD4 cell, the viral RNA is initially present in the plasma without recognition of the host immune system. By approximately day 17, the HIV p24 antigen is present. The body recognizes this viral protein as foreign, thereby eliciting an immune response with production of specific anti-HIV antibodies. During this initial 17-day period, neither HIV antigen (Ag) nor anti-HIV antibodies (Ab) are detectable with HIV tests used most commonly in the community setting. Yet HIV RNA may be detectable as early as day 10 after acquiring HIV with more highly specialized nucleic acid amplification testing (NAAT). That is, during this window period, a person infected with HIV may have an HIV-negative test result (Figure 5).43,44

PrEP 101

What Is PrEP?

PrEP is the use of specific antiretroviral agents to reduce the risk of HIV infection. It is intended for HIV-negative individuals who are at risk of infection through sexual intercourse or injection drug use, including sexually active MSM, heterosexual men and women, and transgender persons. PrEP is also an alternative prevention strategy for individuals receiving PEP who continue to engage in high-risk behavior or who have received multiple courses of PEP. Sexually active adults and adolescents are defined as people engaging in anal or vaginal sex in the past 6 months.45 At the time of this publication, CDC guidelines recognize 3 drug regimens for use as PrEP.45

2021 CDC Updated PrEP Clinical Practice Guidelines

In 2021, the CDC published updated clinical practice guidelines to prevent HIV and provide PrEP services. While the basic process for initiating PrEP has not really changed, there are 3 important updates45,46:

  • It is now recommended that clinicians inform all sexually active teens and adults about PrEP

  • PrEP should be offered and provided to anyone who requests it (following a brief history), even if they don’t report specific HIV risk behaviors or appear “at high risk” of acquiring HIV

  • Three US Food & Drug Administration (FDA)-approved medications are now available for PrEP

    • Oral daily dosing tenofovir disoproxil fumarate (TDF)/emtricitabine (FTC)

    • Oral daily dosing tenofovir alafenamide (TAF)/FTC

    • Cabotegravir (CAB) long-acting injectable (LAI)

Efficacy and Safety of Available PrEP Options

Oral PrEP: TDF/FTC and TAF/FTC

Key clinical trials involving TDF/FTC used as PrEP— included:

  • iPrEx (Iniciativa Profilaxis Pre-Exposición [Preexposure Prophylaxis Initiative])

  • Partners PrEP (Partners Pre-Exposure Prophylaxis Study)

  • FEM-PrEP (Preexposure Prophylaxis Trial for HIV Prevention Among African Women)

  • TDF2 (Botswana TDF/FTC Oral HIV Prophylaxis trial)

  • IPERGAY (Intervention Préventive de l’Exposition aux Risques avec et pour les Gays)

Demonstrated safety and efficacy of the combined use of these 2 nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs) as PrEP across all at-risk populations (Figure 6), including22,47-49,50-52:

  • MSM

  • Heterosexual HIV-discordant couples

  • Heterosexual men and women

  • People who inject drugs (PWID)

As noted in Table 2 and Figure 4 above, TDF and FTC target reverse transcription, stage 3 in the HIV life cycle. Once phosphorylated intracellularly, creating diphosphate and triphosphate metabolites, respectively, these nucleotide/nucleoside analogs inhibit HIV reverse transcriptase by competing with viral substrates, thereby causing DNA chain termination and preventing viral RNA conversion.53 Whereas older NRTIs were characterized by serious toxicities (including bone marrow suppression, peripheral neuropathy, lactic acidosis, and pancreatitis) secondary to their effects on human cellular mitochondrial DNA, these newer NRTIs are weaker inhibitors of mitochondrial DNA and are less likely to result in significant toxicities.54

TDF/FTC remained the sole FDA-approved antiretroviral regimen for use as PrEP from its approval in 2012 until October 2019. The randomized, double-blind, active-controlled DISCOVER trial evaluated the efficacy and safety of TAF/FTC vs TDF/FTC in high-risk cisgender MSM and transgender women who have sex with men (in North America and Europe). Eligible patients (N=5387) had engaged in ≥2 episodes of condomless anal sex in the preceding 12 weeks or received the diagnosis of rectal gonorrhea/ chlamydia or syphilis in the preceding 24 weeks. The primary endpoint was the rate of HIV infection per 100 person-years (PY) when 50% of patients had completed 96 weeks of PrEP use.55 Longer term results, reported once all participants completed 96 weeks, showed that just 23 HIV diagnoses occurred during the study period—an infection rate of 0.16/100 PY for TAF/FTC vs 0.30/100 PY for TDF/FTC (Figure 7).56

Safety Profiles/Side Effects

TAF/FTC was associated with superior bone and renal safety profiles compared with TDF/FTC, although both combinations were well-tolerated and associated with low rates of discontinuation due to adverse events. Combination TAF/FTC was considered noninferior to TDF/FTC in preventing HIV infection, thereby leading to expanded PrEP medication options for persons at risk of HIV acquisition, except for those at risk through receptive vaginal sex or injection drug use.56,57

Both TDF and FTC are primarily excreted through the kidneys; therefore, patients with impaired renal function—based on estimated creatinine clearance (eCrCl), as calculated using the Cockcroft-Gault equation—are at risk.58 Use of TDF/FTC as PrEP is not recommended for persons with eCrCl <60 mL/min/1.73 m2. Further, reduced-dosing strategies for TDF/FTC as PrEP have not been developed.45

In contrast, TAF is metabolized by the lysosomal multifunctional enzyme cathepsin A and can be used in patients with more compromised renal function (eCrCl ≥15 mL/min/1.73 m2) or patients on hemodialysis. Note, however: the FTC component of TAF/FTC is not recommended for patients with eCrCl <30 mL/min/1.73 m2 and, therefore, TAF/FTC for PrEP is not recommended for patients with eCrCl <30 mL/min/1.73 m2).57 The serum half-life of TAF is 0.5 hours, with an intracellular half-life of 150–180 hours. In contrast, the serum half- life of TDF is 17 hours, with an intracellular half-life of >60 hours.57 The bone- and renal-related adverse effects associated with TDF are less likely to occur with TAF due to this lower plasma exposure.59 Of note, in the context of HIV treatment (ie, not PrEP), switching patients who show a decline in bone mineral density from TDF-containing to alternative ART regimens was shown to increase bone mineral density, but the clinical significance of this increase remains unknown.60

While TAF/FTC appears to have safer kidney and bone toxicity profiles than TDF/FTC,TDF/FTC is associated with lower lipid levels and weight loss.45,53,57,61 However, the CDC PrEP guidelines do not provide further parameters for consideration when choosing between these 2 PrEP regimens.19 Other, less significant or transient adverse effects of TDF/FTC include asthenia, headache, nausea, vomiting, diarrhea, flatulence, minor decreases in high- density lipoprotein (HDL), low-density lipoprotein (LDL), and total cholesterol, and minor weight loss.53 TAF/FTC may be associated with headache, nausea, diarrhea, minor increases in LDL cholesterol and triglycerides, and minor weight gain (Table 3).57

Drug-Drug Interactions

Potential drug-drug interactions (DDIs) with TDF/FTC or TAF/FTC are listed in Table 4.36,37,50 As the incidence of hepatitis C virus (HCV) is increased in populations at risk of HIV acquisition, first-line HCV direct-acting antiviral (DAA) regimens are included, even those with no potential DDIs.

The start-up syndrome of headache, nausea, and flatulence occurring in the first month of TDF/FTC use as PrEP occurs in a minority of patients and typically resolves within 3 months, sooner for many patients. Patients should be forewarned of this possibility, instructed on the use of over-the-counter medications for symptom management, and educated regarding more serious signs and symptoms—such as those of acute HIV infection—that should be reported on an urgent basis.42,67

CAB LAI

In December 2021, the US FDA approved injectable CAB as the first long-acting injectable for HIV PrEP, representing a major change in the treatment landscape.68 This agent is administered as an intramuscular (IM) injection every 2 months after 2 initial injections that are given 1 month apart.66 It is indicated for at-risk adults and adolescents weighing at least 35 kg.66 This LAI may be critical in aiding high-risk individuals for whom adherence to daily medications poses a major challenge.45

Results of the HIV Prevention Trials Network 083 (HPTN 083) study of the efficacy of the integrase inhibitor CAB as an LAI for HIV prevention yielded encouraging findings. This National Institutes of Health (NIH)-funded multisite study of 4566 cisgender MSM and trans women who have sex with men randomized participants to receive CAB as a long-acting injectable every 8 weeks or daily oral TDF/FTC. HPTN 083 was terminated in early 2020 after the CAB group was shown to have statistically superior outcomes when compared with the TDF/FTC group, with the CAB group showing 66% lower risk of HIV infection. It should be noted that both study groups received highly effective HIV-prevention regimens: incidence rates of HIV infection were 0.41% in the cabotegravir group, and 1.22% in the TDF/FTC group (Figure 8).69

HPTN 084 was a phase 3 trial designed to track roughly 3200 women of childbearing age in sub-Saharan Africa over a 4.5-year period.70 While both methods were highly effective—HIV incidence of 0.21% (95% CI 0.06%–0.54%) in the CAB group and 1.79% (95% CI 1.24%–2.51%) in the TDF/FTC group— long-acting CAB was 89% (95% CI 68%-96%) more effective than TDF/FTC.70,71 The study was terminated early (November 2020) because of this demonstration of CAB’s statistically based superiority.71 No dosage adjustment is necessary for patients on CAB who have reduced renal function.66Table 4 lists potentially significant drug interactions; note the lack of interactions between CAB and polyvalent cations, which interact with oral INSTIs.

In the HPTN 083 and 084 trials, injection-site reactions (eg, pain, tenderness, induration) were the most frequent adverse effects associated with CAB (82%). These reactions occurred most frequently after the first 2 or 3 injections. According to the CDC, patients should be instructed to take an over-the-counter pain medication within a couple of hours before or after the injection and continue as needed for 1 to 2 days. Also, the patient may apply a warm compress or heating pad to the injection site for 15 to 20 minutes after the injection.45

Determining PrEP Eligibility45

The CDC now recommends that all sexually active adult and adolescent patients be informed about HIV PrEP and that PrEP should not be withheld from HIV-uninfected patients who request it.45 In light of this expanded approach, the updated 2021 CDC Clinical Practice Guidelines provide criteria for determining how to identify sexually active teens, adults, and PWID at substantial risk of acquiring HIV infection. They also delineate who is clinically eligible for daily oral PrEP and for long-acting injection for PrEP (Table 5). Clinicians should:

  • Conduct a comprehensive behavioral risk assessment (sexual; injection drug use) to identify appropriate PrEP candidates. (See also Embracing Sexual Wellness: Sexual Health, Well-being, and Trauma-Informed Care, page 27)

    • Figure 9 and 10 are 2 algorithms to help assess whether patients should merely be told about PrEP or it should be prescribed

    • The behavioral assessment may reveal additional needs for in-depth education, risk-reduction counseling, and community-based patient-support services

  • Assess the patient for common signs and symptoms suggestive of acute HIV infection (Table 6)

  • Document evidence of HIV-negative status with use of

    • An FDA-approved blood test, or CLIA-waived rapid point-of-care fingerstick blood test performed within 7 days of therapy initiation (CLIA-waived indicates that under the Clinical Laboratory Improvement Amendments, a laboratory test can be conducted without the need for more stringent standards)

    • This is for both oral PrEP and CAB

  • Document evidence of HIV-negative status with use of an HIV-1 RNA assay

    • If initiating CAB

    • If initiating oral PrEP AND there has been a recent exposure with someone in a high incidence population, or there are signs/symptoms of acute or primary HIV

    • Guidelines state, however, that you may initiate PrEP if you are waiting for the RNA assay results, as long as you have the documented negative HIV Ag/Ab

  • Perform baseline (pretreatment) laboratory assessment

Confirming HIV-Negative Status

HIV testing is required to confirm that patients do not have HIV infection when they start taking PrEP. Based on the latest updated 2021 CDC guidelines, required testing differs for patients who have or do not have recent antiretroviral PrEP use. Recent use is defined as taking oral PrEP in the last 3 months or taking long-acting injectable (LAI) PrEP in the past 12 months. For individuals who are starting or restarting PrEP after a long period without treatment, the clinician should test using a laboratory-based HIV 1/2 Ag/Ab test. For patients who are taking or have recently taken oral or LAI PrEP, the patient should be tested using an HIV Ag/Ab test and an HIV-1 RNA test.45

A negative HIV test following a high-risk exposure should always be clinically correlated with any signs or symptoms of acute HIV infection and repeat testing after the window period is advised.43,44Figure 11 illustrates the HIV testing guidance that should be used to determine PrEP eligibility. Ag/Ab tests utilizing blood from a vein can usually detect HIV infection 18–45 days after an exposure, whereas rapid Ag/Ab tests with finger-prick blood may detect HIV 18–90 days after exposure (Figure 5).43,44,72 Other HIV tests are recognized and allowed, but it is crucial to remember the limitations of Ab-only tests. CDC-approved, CLIA-waived rapid HIV tests, suitable for use in nonclinical settings, and involving fingerstick or venous whole blood, include but are not limited to the following:

  • Chembio SURE CHECK® HIV 1/2 Assay

  • Clearview® HIV 1/2 STAT-PAK®

  • Alere Determine™ HIV-1/2 Ag/Ab Combined Test

  • INSTI® HIV-1/HIV-2 Ab Test

  • Uni-Gold™ Recombigen® HIV- 1/2 Ab Test

Only the Alere Determine™ test is an HIV 1/2 Ag/Ab assay; all others listed test only for antibodies to HIV-1 and HIV-2.73

The following may be performed using oral-fluid swab (or fingerstick or venous whole blood). Note, however, oral-fluid swabbing should not be used to document HIV status.

  • Chembio DPP® HIV-1/2 Assay (oral test; not acceptable for confirmation of HIV status)

  • OraQuick ADVANCE® Rapid HIV-1/2 Ab Test73

Rapid tests using finger-prick or venous whole blood can yield results in approximately 10 minutes. Oral HIV tests are not recommended in these clinical scenarios.42 The CDC provides a website listing FDA-approved HIV diagnostic tests including a list of CLIA-waived tests: https://www.cdc.gov/hiv/testing/laboratorytests.html.

The 2021 CDC Update requires that patients also have an HIV-1 RNA assay in the following situations as it may detect HIV infection early enough to avoid or limit integrase inhibitor resistance should CAB LAI fail to prevent infection:74

  • Prior to initiating CAB

  • Prior to initiating oral PrEP AND there has been a recent exposure with someone in a high incidence population, or there are signs/symptoms of acute or primary HIV

Guidelines state, however, that you may initiate PrEP if you are waiting for the RNA assay results, as long as you have the documented negative HIV Ag/Ab. Positive HIV tests must be confirmed with more specific conventional blood tests, with results available in <1 hour to several days.

Initiating and Managing Oral PrEP

Pre-PrEP Laboratory Testing, Screening, and Dosing

Besides confirming that the patient is HIV-negative, there are a number of other laboratory tests that should be completed prior to initiating oral PrEP (Table 7) The most critical is the estimated creatinine clearance (as calculated using the Cockcroft-Gault equation):

  • If eCrCl <60 mL/min/1.73 m2, TDF/FTC is not recommended; reduced-dosing strategies for TDF/ FTC as PrEP have not been developed45

  • If eCrCl 15 to <30 mL/min/1.73 m2, TAF/FTC is not recommended57

  • If eCrCl <15 mL/min1.73 m2 TAF/FTC is not recommended unless receiving chronic hemodialysis (on days of hemodialysis, administer the daily dose of TAF/FTC after completion of hemodialysis treatment57

Pretreatment evaluation of hepatitis B (HBV) serology is equally important. Although HBV infection is not a contraindication to the use of PrEP, HBV status must be known and documented prior to PrEP initiation, as discontinuation of PrEP may be associated with acute exacerbation of HBV. Patients positive for hepatitis B surface antigen (HBsAg) should be referred to an appropriate clinician for evaluation for HBV treatment.45

Although TDF/FTC is a safe and effective HIV-prevention strategy for women who are at substantial risk of HIV acquisition including during conception, pregnancy, and the postpartum period, a pregnancy test is recommended for any woman of childbearing potential. Note: TAF/FTC is not approved in those having receptive vaginal sex. Continuing TDF/ FTC during breastfeeding may be beneficial for some women; however, the long-term safety of infant exposure to TDF/FTC has not been determined. TDF/FTC does not reduce the effectiveness of oral contraceptives.45

All patients should be screened for STIs and those considering TAF/FTC should have a lipid screen.

Patient Counseling

Having established the patient’s eligibility for oral PrEP, the following actions should be taken45:

  • Educate patients about the PrEP medications and available regimens to maximize safe use

  • Decide which therapy/mode of administration will be prescribed

  • Provide counseling and effective contraception to women on PrEP who do not wish to become pregnant (only TDF/FTC can be used in those having vaginal sex)

  • Provide other patient-centered counseling regarding the following:

  • Medication adherence to achieve and maintain protective tissue levels

  • HIV risk reduction; including discussion/provision of prevention-services referrals

  • Review PrEP discontinuation and resumption requirements and how to switch from daily to on-demand and vice versa

Selecting and Dosing Daily Oral TDF/FTC or TAF/FTC

Once the patient’s eligibility is established, consider the following when selecting TDF/FTC or TAF/FTC45,57:

  • Renal function: TDF/FTC should not be prescribed for individuals whose eCrCl is <60 mL/min/1.73 m2, because of TDF’s renal clearance

  • Bone density (especially if the patient is over
    50 years of age)

  • Cholesterol

  • Weight change

  • Mode of sexual exposure may influence PrEP regimen selection: as mentioned previously, TAF has not been approved for those at risk through receptive vaginal sex19

  • Potential for drug-drug interactions should be assessed based on all currently taken medications

In real-world clinical practice, selection of PrEP regimen may come down to the practical concerns of insurance coverage and cost.26 A generic formulation for TDF/FTC became available as of October 2020, but TAF/FTC is available only in brand form.28

Dosing

  • TDF/FTC: one 300 mg (TDF) /200 mg (FTC) tablet once daily with or without food, in individuals weighing at least 35 kg45

  • TAF/FTC: one 25 mg (TAF)/200 mg (FTC) tablet once daily with or without food, in individuals weighing at least 35 kg57

Selecting and Dosing On-Demand/2-1-1 Oral PrEP

While not CDC-recommended or FDA-approved, evidence-based on-demand or event-driven protocols for TDF/FTC are endorsed by other organizations. For example, 2-1-1 (also known as event-driven, intermittent, or on-demand), is a PrEP dosing protocol specifically recommended for MSM engaging in anal sex without condoms (not for vaginal sex) by the International Antiviral Society–USA (IAS–USA) and the World Health Organization (WHO).75,76 Named on the basis of the number and timing of PrEP tablets taken, 2-1-1 provides the following recommendations (Figure 12)45,52:

  • A loading dose of 2 TDF/FTC tablets is taken 2–24 hours before anal sex

  • A third tablet is taken 24 hours after the first dose

  • A fourth tablet is taken 24 hours later

  • If there is another sexual exposure within 7 days of the last dose, 1 tablet is taken 2–24 hours before anal sex, 1 tablet 24 hours after the first dose, then 1 tablet 24 hours after the second dose

  • The efficacy of this dosing protocol was demonstrated in the IPERGAY study, the scope of which was limited to TDF/FTC. The preceding 2-1-1 recommendations do not apply to TAF/FTC45,52,75

Initiating and Managing CAB LAI PrEP

Pre-PrEP Laboratory Testing, Screening, and Dosing

In addition to the 2 oral PrEP therapies, an IM LAI is now available. The FDA approved CAB LAI for at-risk adults and adolescents weighing at least 35 kg66 who report sexual behaviors that place them at substantial risk of HIV exposure and acquisition.45 Any sexually active adult or adolescent can use CAB.

As with the oral treatments, the first step is confirming that the patient is HIV-negative; other laboratory
tests include STI screening prior to initiating CAB LAI (Table 8).45

Screening for PrEP Patients Receiving CAB

Patients must be tested for HIV-1 infection prior to both initial and subsequent injections of CAB using a test approved by the FDA for the diagnosis of HIV-1 infection.66 HIV-1 RNA assay is not required at baseline but is recommended at 1 month and subsequent visits when CAB is the PrEP medication. Ideally, this testing should be conducted within 1 week prior to the initiation visit.45 If clinicians want to administer the first injection at the initial PrEP evaluation visit based on findings of a rapid combined Ag/Ab assay, blood should always be subsequently drawn for laboratory confirmatory testing that encompasses an HIV RNA assay.45

Dosing

Interestingly, clinicians may initiate with CAB oral lead-in therapy prior to IM injections or may proceed directly to IM injections (600 mg [3 mL injection, gluteal only]) without oral lead-in.

Without oral lead-in: CAB is administered every
2 months after 2 initial injections that are given
1 month apart.66 That is:

  • 1st injection at the initiation visit

  • 2nd injection at the first follow-up visit
    1 month later

  • 3rd injection at the next follow-up visit 2 months later; all subsequent injections are given every
    2 months (as long as the patient is HIV-negative)

With oral lead-in: Approved dosing for the oral lead-in therapy is 30 mg once daily for approximately 1 month. At the time of this publication, oral CAB is only available through 1 specialty pharmacy (Theracom). The injection should then be administered on the last day of oral lead-in, or within 3 days after.66 That is:

  • Oral daily CAB prescription (for 4 weeks) is given at initiation visit

  • 1st injection at the first follow-up visit 1 month later on the last day or within 3 days of last oral lead-in

  • 2nd injection at the next follow-up 1 month later

  • 3rd injection at the next follow-up visit 2 months later; all subsequent injections are given every
    2 months (as long as the patient is HIV-negative)

Missed Doses With CAB

Adherence to the CAB injection dosing schedule is strongly encouraged. Patients who miss a scheduled dose of CAB should be clinically reassessed to ensure that resumption of CAB remains appropriate.66 Refer to Table 9 for dosing recommendations after missed injections.

If a patient plans to miss a scheduled every-2-month continuation injection of CAB by more than 7 days, the pharmacist should instruct the individual to take daily oral CAB for up to 2 months to replace 1 missed scheduled every-2-month injection. The recommended oral daily dose of CAB is 30 mg daily. The initial dose of oral CAB should be taken approximately 2 months after the last injection of CAB. The patient should restart injection with CAB on or within 3 days of the day oral dosing ends.66

Pharmacist-Led Administration of CAB

Community pharmacists can administer the optional oral lead-in dosing for CAB but are not able to give the intramuscular injection; adjustments are required in pharmacies and provider offices to establish workflow processes that are safe and efficient for providing these injections.77

Monitoring and Counseling Patients on PrEP

Time to Protection

Time to maximum HIV prevention varies with mode of sexual exposure and type of PrEP taken. Table 10 presents the time to maximum HIV protection when using oral PrEP. Time to protection with CAB LAI is currently unknown.78 Patients should be advised that condom use is critical while tissue concentrations remain low, and that ongoing, consistent use of condoms is necessary for the prevention of both HIV and other STIs.45

Effective HIV prevention requires a high level of daily adherence to the PrEP medication, but there is some leeway for missed doses. The STRAND trial showed that 6 to 7 doses of oral PrEP per week are necessary to maintain protective vaginal tissue concentration; fewer doses per week may be adequate to maintain colorectal tissue concentrations. Nonetheless, daily adherence should be stressed for all persons on oral PrEP. Should a patient report missing a dose, instruct the patient to take the single missed dose as soon as it is remembered. If, however, it is nearly time for the next dose, the patient should proceed with the regular dosing schedule.45

Supporting the Patient’s PrEP Adherence

Key tactics for successful medication-adherence counseling include the following:

  • Simplified education and explanations, eg, dosage schedule, impact of adherence on PrEP efficacy

  • Strategies that address patient-specific adherence barriers; eg, substance use

  • Nonjudgmental adherence monitoring; eg, normalizing occasional missed doses, reinforcing success, encouraging condom use

PrEP adherence is supported by effective HIV risk-reduction education. Counseling should include information about substance use and protection from STIs with consistent condom use.45,78 Whether recreational and/or associated with injection practices, psychotropic substances may decrease inhibitions, increase risk tolerance, and reduce self-efficacy with regard to risk- reduction practices.79 Patients also should be reminded that PrEP does not protect against other STIs. Such personal risk-reduction practices—with emphasis on consistent condom use—remain critical, even when one is using PrEP. Adherence should also include:

  • Acknowledgment of the effort required for behavior change

  • Reinforcement of success

  • Assistance with turning around any missteps along the way45

As adverse effects may impact PrEP adherence, pharmacists should counsel patients regarding the following concerns45:

  • Common side effects for oral PrEP (headache, nausea, and flatulence) typically resolve within the first month of taking PrEP (start-up syndrome) using over-the-counter medications for symptom management

  • Common side effects for CAB include mild to moderate injection-site reaction; in clinical trials 4% reported diarrhea, nausea, pyrexia, and fatigue.

  • Signs or symptoms of acute HIV infection or acute renal injury, both of which require urgent intervention

Patient Monitoring: Oral PrEP

The following table provides a comprehensive look at CDC guidelines for oral on-PrEP interval laboratory testing and patient monitoring (Table 11).45

Stopping and Restarting Oral PrEP

Patients should receive specific counseling on how to safely discontinue and restart PrEP. Discontinuation may be prompted by the individual’s personal choice, changed life circumstances, non-tolerated side effects, toxicities, chronic nonadherence, or acute HIV infection (AHI). The protection provided by PrEP wanes over the 7–10 days following discontinuation; therefore, alternative methods to reduce risk of HIV acquisition should be discussed, including the emergent use of PEP in the event of a high-risk exposure.45

Per CDC guidelines, PrEP discontinuation for any reason should be documented by the PrEP provider. This should include HIV status at the time of discontinuation, reason for discontinuation, recent PrEP adherence, and reported sexual risk behavior. Should the patient choose to restart oral PrEP, screening and preprescription evaluation are the same as for someone newly beginning oral PrEP. The PrEP provider should engage the patient in an open discussion regarding change in personal circumstances and commitment to PrEP adherence.45

Patient Monitoring: CAB LAI PrEP

The following table provides a comprehensive look at CDC guidelines for CAB LAI interval laboratory testing and patient monitoring (Table 12).45

Stopping CAB LAI

CAB has a long “tail” of gradually declining drug levels when discontinuing the injections. At some point during this “tail” phase, it’s possible CAB levels will fall below a protective threshold and persist for some time at nonprotective levels, exposing the patient to the risk of HIV acquisition. In the HPTN 077 trial, the median time to undetectable CAB plasma levels was 44 weeks for men and 67 weeks for women with a wide range for both sexes. While there is a risk of developing a drug-resistant strain if HIV infection is acquired during that time, this has not been borne out in the small number of tail-phase incident HIV infections observed. Therefore:

  • Remind patients of the importance of keeping their follow-up appointments if they decide to discontinue CAB for PrEP

  • Initiate an individualized discussion about ongoing HIV prevention if a patient no longer wants to receive CAB LAI, but remains at-risk for HIV infection

    • This may include oral PrEP options in combination with other effective HIV prevention behavioral changes, including universal condom use

    • Patients who want to switch to oral PrEP may do so within 8 weeks of last CAB injection

  • Perform all screening as if new to therapy prior to restarting if a patient wishes to resume PrEP therapy with CAB

  • The US FDA recommends quarterly HIV RNA testing over 1 year after the last injection.45

Tear-Out Inserts of the CDC Clinical Guidelines Summary Tables

For your convenience we have included the full tables provided in the CDC updated guidelines for both oral and CAB PrEP (Table 13; Table 14)

PrEP in Adolescents

In the US, TDF/FTC is approved by the FDA for use as PrEP in adolescent patients who weigh at least 35 kg.53 TAF/FTC is also approved for use as PrEP in adolescent patients who weigh at least 35 kg and do not engage in receptive vaginal sex.57 Moreover, CAB is also indicated for use as PrEP in adolescents.66 Young MSM are at particularly high risk for acquiring HIV.80,81 Among MSM aged 13 to 24 years, the rate of new infections increased by 43% from 2003 to 2014.81

The Adolescent Trials Network (ATN) studied daily TDF/FTC PrEP in 78 patients 15 to 17 years of age. TDF/FTC was well tolerated among those who took their medication; however, adherence was suboptimal for many of the youth (protective levels of tenofovir were found in approximately 50% at 12 weeks when they were being seen every 4 weeks, but only in 22% at 48 weeks when visits were quarterly).82 The findings from this study suggest that potential barriers to adherence should be identified and addressed. Furthermore, for younger MSM, more frequent pharmacy-patient contact may be important.

In addition to addressing adherence, pharmacists must weigh the potential risk of bone effects with the relative risk of acquiring HIV. The bone loss seen with TDF may pose additional risks in adolescents, however, data indicate that any effects are reversed upon discontinuation of TDF.45,83

Laws and regulations that may be relevant for PrEP-related services provided to adolescent minors by pharmacists differ by jurisdiction. Pharmacists considering providing PrEP to a patient <18 years old must be aware of applicable local laws, regulations, and policies. The following website from the CDC outlines the differences among states in the US: https://www.cdc.gov/hiv/policies/law/states/minors.html. Pharmacists should discuss any limits of confidentiality based on these local laws, regulations, and policies.45

PrEP Resistance or Failures: Transitioning From PrEP to HIV Treatment

Patients taking PrEP may become HIV-positive for various reasons. If detected at the first follow-up visit after PrEP initiation, it may be due to the patient having had undetectable acute infection at the time of pre-PrEP HIV testing (ie, within the window period). If detected at a later follow-up visit, this may indicate adherence issues.45 Rarely, some patients become HIV-positive despite good PrEP adherence: risk of HIV acquisition is not completely eliminated by high levels of adherence and may be due to drug-resistant viral strains.84

Among patients who become infected with HIV while receiving PrEP, most are unlikely to develop drug-resistant virus.47,48,50,51 In a meta-analysis that evaluated drug resistance in 6 clinical trials, drug resistance was identified in 6 of the 533 patients who became infected with HIV after enrollment, and 8 of the 44 patients who had undiagnosed acute HIV infection at study entry.85

In patients who develop drug-resistant virus while taking PrEP, the M184V mutation is most likely to emerge since the genetic barrier to emtricitabine is low.86 Exposure to study drug was believed to cause drug resistance in 4 of the 33 females in the Fem-PrEP trial and in 2 of the 51 patients in the Partners PrEP study.50,87 Among those who developed resistance, the M184V emtricitabine resistance mutation arose most frequently.50,87

Individuals receiving PrEP may be infected with drug-resistant HIV. In a case report, a patient became infected with HIV that contained drug resistance mutations for several classes of agents, including tenofovir and emtricitabine.88 Transmission occurred despite tenofovir levels that were consistent with recent administration of the drug and long-term adherence.88

Transitioning From PrEP to HIV Treatment as Prevention

US Department of Health and Human Services (DHHS) HIV/AIDS treatment guidelines recommend that upon laboratory confirmation of HIV infection in an individual on PrEP, the patient should transition from PrEP to comprehensive HIV treatment.61 Using a shared decision-making approach, an HIV provider and the patient should both be involved in determining ART selection and initiating therapy. Under guidelines for rapid ART initiation (which can be used in all patients; rapid/immediate ART; rapid/immediate start), there is no need to wait for additional laboratory results prior to starting treatment. However, baseline laboratory data should be drawn at the visit in which ART is initiated. Rapid initiation refers to initiating ART within 14 days of HIV diagnosis, while immediate initiation refers to starting ART on the same day as diagnosis.89 The goal of a rapid/immediate start strategy is to accelerate the time from HIV diagnosis to engagement in care, ART uptake, and, ultimately, viral suppression. Current guidelines call for ART initiation on the day of diagnosis (or as close as possible).61 Patients should be counseled regarding the benefits of early/continuous viral suppression for both themselves and their partners, with emphasis on undetectable=untransmittable (U=U). That is, maintenance of viral suppression (an undetectable viral load on ART), helps prevent the transmission of HIV to others. This is the basis of HIV treatment as prevention.90,91

PEP 101

PEP: Pharmacologic Considerations:
Efficacy and Safety

HIV postexposure prophylaxis (PEP) utilizes specific combinations of antiretroviral agents to prevent infection in appropriately identified and evaluated HIV-negative people who have had a single high-risk HIV exposure.19,92 Prescribing PEP should be considered an emergency intervention: PEP is not a substitute for appropriate, continuous use of PrEP in conjunction with behavioral risk reduction. It can be thought of as similar to dispensing the morning-after contraceptive pill. PEP is most effective when initiated as soon as possible—no more than 72 hours—after a potential HIV exposure.92

  • Occupational HIV exposure refers to high-risk events occurring in a health care setting

  • Nonoccupational HIV exposure refers to an
    isolated exposure to potentially infectious body fluids that occurs outside of a health care setting, most commonly through sexual activity or injection drug use

This discussion addresses nonoccupational PEP (nPEP) only.92

Although TDF and FTC are used in PEP, the clinical/ pharmacologic concerns—and intensity of treatment— are more similar to treatment of established HIV infection than to PrEP. PEP requires the use of either an integrase strand transfer inhibitor (INSTI) or a boosted protease inhibitor (PI) in combination with both TDF and FTC. Current CDC PEP guidelines do not include use of TAF or CAB as a PEP-regimen component. 92

PEP is usually prescribed as a 28-day course of treatment. Currently recommended (preferred) ART regimens for use as PEP in HIV-negative adults and adolescents include the following92:

  • TDF 300 mg with FTC 200 mg once daily plus

  • Raltegravir (RAL) 400 mg twice daily

OR

  • TDF 300 mg with FTC 200 mg once daily plus

  • Dolutegravir (DTG) 50 mg once daily

The following regimen is recommended as an alternative:

  • TDF 300 mg with FTC 200 mg once daily plus

  • Darunavir (DRV) 800 mg and ritonavir (RTV) 100 mg once daily

The CDC PEP guidelines do not in general recommend one INSTI over the other (ie, DTG vs RAL), however, DTG may be preferred for its once-daily dosing.92-94 Note that use of DTG should be avoided in the periconception period because of the risk of neural tube defects linked to drug exposure during the first 28 days of gestation. There are no data to suggest that DTG has any teratogenic signal after the periconception period.95 Among deliveries in which the mother was taking DTG at conception, neural tube defects were found in 0.30% of cases, versus 0.10% where the mother was taking a non-DTG drug at conception, and 0.08% in HIV-uninfected mothers.95 Similarly, nonpregnant women of childbearing potential who are sexually active or have been sexually assaulted and are not using effective birth control should not receive DTG.94 In either clinical scenario, patients should be prescribed RAL. Additional pharmacologic considerations when prescribing RAL or DTG as PEP are outlined in Table 15.61,93,94

RAL and DTG are both associated with adverse events that include headache, insomnia, and weight gain. 61,93,94 Both agents are also rarely associated with depression and suicidal ideation, although this usually occurs in patients who have preexisting psychiatric conditions. RAL may additionally cause fever, nausea, and diarrhea, and may be associated with more severe conditions, including Stevens-Johnson syndrome, toxic epidermal necrolysis, and rhabdomyolysis.93,94

DTG is associated with additional adverse effects that include hepatotoxicity, hypersensitivity reactions (including rash, constitutional symptoms, organ dysfunction, and liver injury) and, as noted, potential for increased risk of neural tube defects in infants born to patients treated with DTG surrounding the time of conception (Table 16).94

Pretreatment Determination of PEP Eligibility92

As with HIV PrEP, HIV PEP prescribing is associated with several pretreatment requirements:

  • Conduct a comprehensive behavioral-risk assessment (sexual; injection-drug use) and identify individuals at substantial risk of HIV acquisition

  • Confirm HIV-negative status: select preferred HIV tests and/or accurately interpret HIV test results

    • If a rapid test is not available and nPEP is otherwise indicated, therapy should still be initiated

  • Order appropriate laboratory tests to assess PEP-associated risks

  • Screen for drug/drug interactions

As PEP is most effective when initiated as soon as possible—no more than 72 hours—after a potential HIV exposure, these pretreatment evaluations should be performed immediately. In addition, PEP is not indicated if the patient has been consistently adherent to PrEP. 92

Comprehensive Sexual (and Injection Drug Use)
Risk Assessment92

During the initial postexposure evaluation, the pharmacist should determine the following:

  • Route/source of the potential HIV exposure

  • Timing and characteristics of the exposure, including sexual assault

  • Frequency of potential HIV exposures

  • Risk of other STIs (gonorrhea, chlamydia, syphilis), HBV, or HCV

  • Potential for current pregnancy (with pregnancy testing)

Needle-sharing during injection drug use and receptive anal intercourse are considered the highest per-act risks for nonoccupational HIV transmission. Insertive anal intercourse, insertive penile-vaginal intercourse, and oral sex carry relatively lower risks.92Figure 13 is an algorithm for determining HIV exposure and whether nPEP is recommended.

Confirmation of HIV-Negative Status92

HIV-testing considerations, as outlined for pre-PrEP assessment—Determining PrEP Eligibility, Confirming HIV-Negative Status (pg 13)—also pertain to pre-PEP assessment. Special emphasis should be placed on clinical correlation of HIV-testing results and screening for signs and symptoms suggestive of acute/primary HIV infection. Importantly, if HIV test results are unavailable during the initial evaluation, initiation of PEP should be determined on the initial assumption that the potentially exposed patient is not infected. When the HIV status of the potential source of exposure is unknown—if possible—that individual should consent to clinical evaluation and HIV testing using an HIV 1/2 Ag/Ab test. PEP may be considered and prescribed if an HIV test is not available or if the HIV status of the source is unknown.

Other Baseline Labs and Assessment for PEP-Associated Risks

As with HIV PrEP, pre-PEP laboratory assessment must be performed prior to treatment initiation. Again, critical labs include estimated creatinine clearance (eCrCl), HBV serology, and pregnancy testing. Additionally, if indicated by the comprehensive risk assessment, patients considered for PEP should be referred for timely STI testing and treatment. When appropriate, the patient should be offered routine prophylaxis for gonorrhea, chlamydia, or trichomoniasis. This is also an opportunity to address the need for vaccinations for HBV, HCV, and human papillomavirus (HPV).92

DHHS guidelines for pre-PEP (baseline) and follow-up laboratory testing largely replicate those for PrEP initiation and continuation. Differences include the addition of liver function evaluation (alanine aminotransferase [ALT] and aspartate aminotransferase [AST]) and test-specific requirements for postexposure follow-up testing (Table 17). 92

Patient-Centered PEP: Regimen Selection and Pretreatment Counseling92

Having established the patient’s PEP eligibility—or having done so to the pharmacist’s satisfaction, if HIV results are not immediately available—the PEP provider’s additional pre-PEP requirements include the following clinical activities. Table 18 provides a full summary of initial PEP evaluation, medication prescribed, and management.58

  • Prescribe safe and effective PEP regimens for eligible persons without HIV infection

  • Educate patients about the selected PEP regimen to optimize safe use

  • Provide counseling and effective contraception to women initiating PEP who do not wish to become pregnant

    • Discuss emergency contraception with women of reproductive potential

    • Refer immediately for assessment of any reported sexual assault

  • Provide other patient-centered counseling regarding the following:

    • Medication-adherence to achieve and maintain protective tissue levels

    • HIV risk reduction; including discussion and provision of prevention services referrals

    • PEP medication safety during pregnancy and breastfeeding, if indicated

Basic medication-education components include the following:

  • Which PEP medication is being prescribed? How is it taken? What is the dosing schedule?

  • How to manage missed doses

  • Common signs and symptoms suggestive of acute HIV infection (Table 6)

  • Common side effects associated with raltegravir and dolutegravir (Table 16)

  • What quantity will be dispensed?

A 28-day course of an INSTI-based 3-drug antiretroviral regimen should be prescribed for all persons requiring PEP. To optimize adherence, consideration should be given to patient-specific characteristics, number of doses per day and/or pills per dose, and minimization of side effects. Providing an entire 28-day course of the PEP regimen and scheduling an early follow-up visit may increase the likelihood of treatment adherence, especially if patients cannot return for multiple follow-up visits.

  • Follow-up visits allow pharmacists to provide further patient-centered evaluation, education, and counseling:

  • Discuss baseline blood test results (including HIV results, if a rapid test was not available at the time of initial assessment)

  • Assess for adherence and adverse effects, or change the PEP regimen, if indicated

  • Recommend medications for symptomatic relief of side effects (eg, antiemetics)

  • Recommend pill boxes or other medication adherence aids (including smartphone apps)

  • Identify strategies for dosing in line with a patient’s daily schedule

  • Ensure means for patient-pharmacist contact during PEP treatment

Patients should be counseled repeatedly on the importance of PEP-regimen adherence and treatment completion to optimize prevention of HIV infection.92

Transitioning From PEP to PrEP (PEP2PrEP)

PEP is indicated only for patients with ≥1 known or suspected high-risk, unprotected HIV exposure who are HIV-negative at initial PEP evaluation. Patients with documented HIV-negative status (preferably using an HIV 1/2 Ag/Ab test) upon completion of a 28-day course of PEP, can be transitioned from PEP to PrEP without any gap in treatment.45,92 Patients who are at frequent, recurrent risk of HIV exposure, or require sequential or near-continuous courses of PEP, should be offered one of the 3 FDA-approved PrEP regimens that best meets their needs, in conjunction with behavioral risk-reduction counseling and interventions. Patients should receive full PrEP education and be reassessed and counseled regarding medication adherence—despite their successful completion of PEP.92

PEP Failure and Transitioning From PEP to
HIV Treatment as Prevention92

If use of PEP fails to prevent HIV infection, patients may experience signs and symptoms of acute/primary, or early, HIV.44 Patients should be instructed during the initial PEP evaluation about such manifestations, as listed in Table 6. Should any signs and symptoms related to acute HIV infection occur during the 28-day PEP course or anytime within 1 month after PEP completion, patients should return for further evaluation and referral to an HIV treatment specialist, if indicated. If a patient is taking 3-agent PEP ART at the time of detected infection, ART should be continued until patient evaluation and treatment planning with an experienced HIV treatment specialist are completed.

Embracing Sexual Wellness: Sexual Health, Well-being, and Trauma-Informed Care

Trauma-Informed Care

SAMHSA (Substance Abuse and Mental Health Services Administration) defines trauma as events or circumstances experienced by an individual as physically or emotionally harmful or life-threatening, which result in adverse effects on a person’s functioning and well-being.117 Trauma is very prevalent in PLWH in the United States: 30% experience childhood physical and/or sexual abuse before age 13; 68% to 95% of women living with HIV experience intimate partner violence (IPV), as do 68% to 77% of men and 93% of people who identify as transgender. Trauma is associated with psychosocial impairment (including posttraumatic stress disorder [PTSD]); panic, phobic, or generalized anxiety disorders; depression; increases in alcohol or substance use; and an increased likelihood of factors associated with increased risk for HIV, such as inconsistent condom use and multiple sex partners. In addition, poverty, race/ethnicity, sexual orientation, and gender may increase the risk of exposure to potentially traumatic events, thereby compounding HIV risk.118 One study of young MSM (74% African American) found that childhood trauma and generalized anxiety were associated with decreased PrEP adherence.121

Being trauma-informed is an approach to care and prevention that acknowledges that traumas may have occurred or may even be an active part in a patient’s life, and that those traumas may manifest physically, mentally and/or behaviorally.124 A number of states have included the concept of a trauma-informed approach (TIA) to care, or trauma-informed care (TIC) in their board of pharmacy PrEP/PEP protocols. TIC recognizes that health care providers and organizations must have a complete picture of a patient’s life story in order to effectively provide health care services with a healing approach. This may improve adherence, patient engagement, health outcomes, and provider wellness. SAMHSA’s TIA is based on the following 4 “R” assumptions, whereby TIC seeks to117,119:

  • Realize the widespread impact of trauma and understand paths for recovery

  • Recognize the signs and symptoms of trauma in patients, families, and staff

  • Respond by fully integrating knowledge about trauma policies, procedures, and practices

  • Resist retraumatization of patients and staff

In a TIA, providers and staff at all levels of the organization have a basic understanding of trauma and how it may affect individuals, families, and communities. It is generally accepted that trauma plays a role in mental and substance use disorders (SUD) and needs to be addressed in prevention, treatment, and recovery.

In addition, there are 6 key principles of a TIA117,119, 124:

  • Physical and Emotional Safety: throughout the organization, pharmacy, and clinic, where patients and staff feel physically and psychologically safe

    • Create a safe and welcoming environment

    • Ensure privacy and confidentiality

    • Be consistent and predictable, nonshaming, nonblaming

    • Trustworthiness and Transparency: decisions are made with transparency, with the goal of building and maintaining trust

    • Maintain professional boundaries

    • Keep roles clear, maintaining transparent policies and processes (including informed consent and grievance process)

  • Peer Support: individuals with shared experiences are integrated into the organization and viewed as intrinsic to service delivery. Peer support and mutual self-help can:

    • Establish safety and hope

    • Build trust

    • Enhance collaboration

    • Promote recovery and healing

  • Collaboration: importance is placed on partnering and leveling of existing power differences between staff and patients, and among staff; it recognizes that everyone has a role to play (ie, “you don’t have to be a therapist to be therapeutic”)

    • Ensure respect, connection, and hope

    • Recognize that healing occurs in the context of the interpersonal relationship

    • Share in decision-making (ie, doing “with” rather than “to” or “for”)

  • Empowerment, Voice, and Choice: patient and staff strengths are recognized, built upon, and validated; this includes the belief in resilience and the ability to heal from trauma

    • Use strengths to build and enhance healthy coping skills

    • Understand past coping mechanisms and the normalcy of the response to a not-normal situation

    • Value and ensure individual control and autonomy

    • Frame experiences as survivorship, not victimization

  • Cultural, Historical, and Gender Issues: biases, stigma, stereotypes, and historical trauma are recognized and addressed

    • Actively move past stereotypes and biases based on race, ethnicity, sexual orientation, age, religion, gender, geography, etc

    • Provide gender-responsive services

    • Utilize the healing value of traditional cultural connections

Here are some examples of what pharmacists can do to promote TIC:

Among heterosexual women, unprotected condomless vaginal and/or anal sex with a male partner is the most common mode of HIV transmission. IPV constrains women’s access to and use of HIV prevention methods. PrEP is a positive intervention for women, as it does not require the same interpersonal negotiation with a partner that condoms do. Further, for patients who may not have control over their sexual encounters, PrEP confers control over HIV acquisition. Ensure that patients who have experienced IPV have alone clinic time to communicate freely and privately.120

When a patient presents as a victim of sexual assault, one of the most urgent needs (as well as safety and criminal justice concerns) surrounds risk of HIV and PEP initiation. It is very common for individuals who have experienced sexual assault to have delayed presentation for care. If they present within the 72-hour time frame for PEP, a risk assessment must be done quickly, and the patient given a trauma-informed explanation regarding their risk of HIV acquisition and offered PEP. Discuss the 28-day regimen, laboratory tests and follow-up care and referrals. In rural, low HIV-incidence communities, PEP may not be readily available; stock the medication (or consider stocking “starter” PEP packs until the individual can access the full supply) and let other pharmacies, clinics, and organizations know that you have it available and can be a referral for care.122 Make sure to take a nonjudgmental approach to someone who is requesting multiple courses of PEP, especially in situations involving sexual assaults, or in those who are sex workers.

A TIA to caring for adolescents will increase a teen’s safety and autonomy. This can include brief counseling about healthy relationships in general and may promote healthy adolescent sexual relationships. Make sure teens know about resources for relationship abuse and sexual violence and facilitate referrals to advocates who assist survivors of abuse.125

A substantial number of LGBTQ individuals are victims of IPV, hate crimes, and other forms of trauma. Because of the prevalence of trauma over the course of the lives of these individuals, clinicians must always use TIC with this population. Antecedent violence should be considered if the patient presents with injuries, depression, substance use, or PTSD. In those who report violence, pharmacists can provide TIC and support by expressing empathy, ensuring safety, and addressing health consequences, including HIV prevention (as well as referring to medical assistance or helping them get care for their injuries).123

The Sexual Health Assessment

Comprehensive sexual health assessment is critical to determining an individual’s need for PrEP, as HIV risk may be associated with numerous personal, partner, relationship, social, cultural, network, and community factors.45 Many health care providers do not ask about same-sex behaviors because of personal discomfort or anticipated patient discomfort. Further, patients often do not disclose sexual behaviors because of perception of stigma or fear of judgment. Frank, nonjudgmental questions about sexual behavior, alcohol use, and illicit drug use should be asked.45 The CDC’s sexually transmitted diseases treatment guidelines recommend the following risk-behavior questions, known as The 5 Ps96:

  • Partners: In the past month, with how many partners have you had sex? Are they men, women, transgender women, and/or transgender men?

  • Practices: What body parts do you use for sex? Based on what body parts you use, that’s where we test for sexually transmitted infections (STIs)

  • History of STIs: Have you ever had an STI? When? Have any of your partners had an STI?

  • Protection from STIs: How often do you use condoms? What do you do to protect yourself from HIV and STIs?

  • Pregnancy plans: Do you wish to prevent pregnancy? Are you using contraception; if so, what type?

Clinicians can enhance their history-taking skills by making patients feel more comfortable and, therefore, potentially more open: use neutral and inclusive terms; avoid assumptions based on age, appearance, marital status, or other factors; and ensure that patients share an understanding of the terms used.97-99

Before starting, consider informing the patient about the routine nature of conducting a sexual-health assessment with the following statement: These questions can make people feel uncomfortable. But I ask to get a better assessment of your risk and to work with you to reduce this risk specific to your life. Everything you tell me is confidential, as part of our patient-provider relationship. If my questions are triggering, please let me know. Is it okay if I begin your sexual health assessment? Do you have any questions before we start?” If patients want to know the reasons/importance of a sexual health assessment, consider using the following statements100:

  • Sexual health is important for overall emotional and physical health

  • We ask these questions every year because it is common for people’s sexual behaviors and partners to change over time

  • As you may know, sexual activity without protection can lead to STIs. These diseases—especially syphilis, gonorrhea, and chlamydia—are very common and often do not cause any symptoms. If we do not catch and treat these diseases, they can affect general health and well-being

  • These questions can also help guide our conversation about how you can protect yourself from STIs, unwanted pregnancy, or other issues that may concern you. It will also give you an opportunity to talk about problems with, or changes in, sexual desire and functioning

  • Our discussion may help us identify other health needs (eg, additional laboratory testing; vaccinations) or resources you may need as part of your lifestyle (sexual or otherwise)

Communicating With LGBTQIA Persons

In addition to the empathy and respect that you show all patients participating in sexual-history discussions, there may be special considerations when taking sexual histories with LGBTQIA persons. It’s important to remember that people who are transgender are high-risk and underrepresented among PrEP users and may require particular sensitivity. The following points taken from the National LGBTQIA+ Health Education Center may be helpful when taking sexual histories, especially with regard to gender pronouns.97

  • Make sure you have established a good rapport with the patient before asking about sexual practices (or doing a physical exam)

  • Be sure to use the patient’s preferred name during conversations and ask what the patient’s pronouns are

    • This will not necessarily be the name that appears on insurance and medical records, and some people change their personal pronoun preference

    • Ensure that pronouns are very visible in the patient’s chart and any documentation that the patient would read in MyChart would reflect chosen name and pronouns

    • Many people who are transgender want you to use the pronoun that matches their gender identity. For example, a transgender female may like you to use “she/her/hers”

    • Some people who are transgender or gender-nonconforming may ask you to use “ze” or “they,” or to try to avoid using any pronouns

    • When in doubt, use the patient’s name or the gender neutral "they"

    • Instead of assigning gender to partners, just ask the patient what body part(s) they use for sex, in order to guide your STI testing and your risk-reduction conversation

  • If possible, use affirming language when discussing a patient's body parts. The easiest way to accomplish this is to ask the patients what words they use to describe their genitals and body. Mirroring a patient’s language can lead to improved health goals and outcomes

Discussing Risk of HIV Acquisition Through Injection-Drug Practices

The US Preventive Services Task Force recommends that all health care providers be aware of signs and symptoms associated with injection drug use. Brief risk-behavior assessment questions that address this concern include the following45:

  • Have you ever injected drugs that were not prescribed to you by a clinician?

  • If yes, when did you last inject nonprescribed drugs?

  • In the past 6 months, have you injected using needles, syringes, or other drug-preparation equipment that had already been used by another person?

  • In the past 6 months, have you been in a methadone or other medication-assisted substance use disorder treatment program?

Answers from these questions may help you determine if additional resources are needed for patients who inject drugs or have other high-risk substance-use related concerns or behaviors. Such resources include harm-reduction (behavioral risk-reduction) programs and services, including the National Harm Reduction Coalition (https://harmreduction.org/), medication-assisted treatment (eg, buprenorphine), inpatient or residential drug-treatment programs, or relapse-prevention services (eg, 12-step programs or mental health/behavioral support programs). In addition, be able to offer naloxone or know where to refer people for it.45,101 The website for SAMHSA (https://www.samhsa.gov) is a valuable resource for locating substance use disorder treatment facilities and programs and behavioral and mental health services.102

Helping Your Patient Navigate Beyond Pharmacy-Based HIV Preventive Care: Referral, Advocacy, & Resources

When to Refer to Primary Care or
Specialist Providers

A positive HIV screen should prompt immediate linkage to the patient’s PCP or HIV specialist for confirmatory testing and subsequent initiation of comprehensive ART, if positive.

Further, if acute HIV is suspected (based on symptoms)—or a patient tests positive for HBV, becomes pregnant, or has symptoms or laboratory findings of acute renal injury—a referral to a PCP or other appropriate provider should be made and the patient should cease participation in the statewide protocol. A positive STI screen or considerations pertaining to usual care are not necessarily reasons to terminate a patient’s participation.

Developing relationships with physicians and advanced practice nurses in your area who are experienced in HIV care and prevention will expedite the referral process and ease patient concerns. The individual patient’s records, along with full documentation of pharmacist-provided PrEP services, should be transferred to the patient’s PCP, whenever a referral is necessary.

Patients who do not have a PCP experienced in PrEP provision can be referred to the American Academy of HIV Medicine database to locate a PrEP provider in their area103: https://providers.aahivm.org/referral-link-search?reload=timezone.

Keep in mind that patients who initially request PrEP or PEP from their pharmacist—not their PCP—may have a higher level of comfort with and/or do not perceive stigma/discrimination on the part of the pharmacist. This trusted patient-pharmacist relationship should be maintained to support the patient’s well-being.

Your Patient’s Advocate:

Financial-Assistance Resources

For patients who lack prescription drug coverage, several resources are available. Ready, Set, PrEP is a nationwide program launched by the CDC in December 2019 to make PrEP available at no cost to individuals who meet the following requirements21:

  • Test negative for HIV AND

  • Have a valid prescription from a health care provider AND

  • Do not have health insurance coverage for outpatient prescription drugs

Although Ready, Set, PrEP ensures access to PrEP for qualifying individuals, it also stipulates that the costs for necessary clinic visits and laboratory testing may vary based on income.21 Gilead’s Advancing Access® program is another patient assistance program available for both insured and uninsured patients that offers a copay coupon program and patient support: http://www.gileadadvancingaccess.com. There is also a program sponsored by ViiV, called ViiVConnect to help patients with financial and insurance issues and providers with tools, resources, and individual assistance with benefits verification, prior authorizations, or medication reimbursement: https://www.viivconnect.com/

In addition, some states in the US have PrEP-specific financial assistance programs that cover medication, clinical care, or both. This information is available from the National Alliance of State and Territorial AIDS Directors (NASTAD) (Table 19).104

Pharmacist Resources

The CDC has created tools to help pharmacists and other providers keep track of important patient and provider steps before PrEP is prescribed101:

Establishing a safe and supportive environment in which to discuss a patient’s personal health issues—one that also promotes confidentiality—is an important consideration for pharmacists who are starting to build a PrEP/PEP-provider practice.105 Several resources are available to help pharmacists develop a comprehensive and supportive sexual-history dialogue with patients:

Addressing Practical Challenges of PrEP/PEP Implementation

Legislation in states such as California and Colorado allow any qualified pharmacist in the state to offer PrEP/PEP services independent of physician authorization and is intended to help fill gaps in care, especially in urgent situations in which a PCP is not established.29,39 There are a number of other models/examples across the country:

  • Mission Wellness Pharmacy is a one-stop PrEP program that allows pharmacists to initiate and furnish PrEP directly to patients as part of a demonstration project with the San Francisco Department of Public Health.106

  • Other models in the US utilize a collaborative practice agreement (CPA) (also called a collaborative drug therapy agreement [CDTA]) that reflects an established and formal relationship among providers (eg, pharmacists, physicians, and advanced practice nurses) and generally requires a referral. This allows pharmacists to order labs and initiate PrEP.

    • Kelley-Ross Pharmacy in Seattle is able to offer pharmacist-driven PrEP provision and HIV-prevention services through not only CPAs, but also through Washington State legislation allowing pharmacists to bill for patient-care services, including monthly patient evaluations, without a limit on length of time the patient can remain under the care of a pharmacist.107

  • CPAs can include a variety of different elements. The key elements are ensuring that everybody who is a stakeholder has a voice within the process of developing the agreement and that there is an understanding of how the different practice types will be integrated for the best interest of the patient. Promoting mutual trust and respect is crucial for fostering communication and good team building to maintain these structures over time to have a productive relationship among pharmacists and other members of the multidisciplinary team.108,109

    • There are many CPA/CDTA templates that can be used and modified to fit the unique needs of each agency. Here are a few areas to consider when creating a CDTA:

      • Confirm that there is malpractice insurance for the pharmacist and the medical director

      • Include STI testing and not just PrEP/HIV, since these two go hand-in-hand in PrEP services

      • Review the rules about notifying the health department with STI/HIV-positive results

      • Work in consultation when needed

      • Develop a regular review process to maintain communication and improve the coordination between the medical-license provider and pharmacist(s)

      • CPAs will likely not allow administration of CAB for PrEP in pharmacies, which will probably remain oral at this time

  • Collaborative practice agreements are often embedded into existing pharmacy infrastructure, which makes it easier to implement these types of programs. Rather than creating a new independent service line, it is contained within existing services. It is intended to generate meaningful outcome measures, as it lends itself to programmatic evaluation and quality improvement efforts. Such collaborative practice agreements can reduce costs by improving efficiencies among diverse components of the care team.108,109

  • Table 20 outlines the barriers and opportunities associated with collaborative practice agreements in pharmacy for HIV PrEP.

  • Different types of technology, including electronic medical records, can be vital to supporting the efforts of a collaborative practice agreement and ensuring that anybody on the care team can confirm the status of patients who are being cared for. The language should be simple and empowering so that everybody has a clear understanding of the expectations. It often will include algorithmic guidance as well as clear delineations regarding when other parts of the team should be involved if there are specific questions.108,109

Building In-Pharmacy Infrastructure and Capacity

A commentary in the Journal of the American Pharmacists Association (2020) provides recommendations for implementing a community-based PrEP program, one reflecting the previously discussed San Francisco and Seattle models. That publication also provides recommendations for how community pharmacies can integrate PrEP/PEP implementation with their existing services infrastructure4:

  • Set up logistics for on-site laboratory testing or for sending out specimens

    • Provide designated physical space (including restrooms) within the pharmacy for laboratory specimen collection

    • Order CLIA-waived rapid HIV tests

    • Utilize mail-in lab kits

    • Train staff regarding on-site (“one-stop PrEP”) and/ or off-site specimen collection and handling

    • Consider use of an on-site phlebotomist

    • Establish a relationship with an external lab; using a single lab can help streamline staff training and administrative burden

    • Work with your lab of choice to discuss the feasibility of its providing designated staff at your collection site. Many labs with significant capacity can offer this service

    • Train staff on self-collection (patient) techniques and post instructions with visual aids in the restroom. If on-site specimen collection is not an option for your practice location, work with a single lab with several specimen collection locations

  • Manage laboratory test results

    • Adapt pharmacy workflow

    • Encourage an appointment-based model for protocol-based assessment, laboratory specimen collection, and prescription pickup

    • Establish private/confidential spaces for discussion of test results

    • Consider installing modular-ready private counseling rooms

    • Assign specific pharmacy staff to accommodate walk-in requests for lab results

  • Establish communication protocols and methods

    • Determine how confidential information will be shared among team members, patients, referring providers, or health departments in accordance with Health Insurance Portability and Accountability Act (HIPAA) requirements110

    • Set up online portals with secure access for e-mail, text messages, apps, and shared electronic medical records (EMRs); this can save both time and staff resources

    • Utilize secure lab-based online portals for accessing test results; access eligibility may vary

    • Create a standardized referral mechanism for positive screens or abnormal laboratory results

  • Provide and monitor pharmacist education and hands-on training, including training of pertinent auxiliary pharmacy staff

    • Require pharmacists to demonstrate competency in delivering PrEP and PEP per state and national guidelines, inclusive of sexual-health counseling, comprehensive risk-appropriate prevention strategies, laboratory testing and interpretation, and navigation of insurance benefits and patient-assistance programs

    • Set realistic time frames for implementation of new work processes and attainment of target number of patient visits

    • Provide ongoing pharmacist and staff feedback, training, and monitoring

    • Designate a pharmacy staff member to disseminate PrEP/PEP guideline changes

The approval of CAB LAI for PrEP has created additional implementation issues, which were discussed with Faculty Member, Melissa Badowski, PharmD, from the University of Illinois at Chicago, College of Pharmacy111:

  • In Illinois, Dr. Badowski is not able to administer the IM injection; only the nursing staff may do so. However, another faculty member from Nevada was able to receive additional training to give CAB gluteal IM injections. Therefore, the ability to obtain training to provide gluteal IM injections will vary by state and between state boards of pharmacy

  • Currently, the CPA at Dr. Badowski’s facility only includes oral PrEP (based on current insurance coverage), but it will be updated to include recommendations regarding CAB initiation. This will still follow the same baseline and routine monitoring as outlined by the 2021 CDC Updated Guidelines.

  • CAB does require refrigeration, and this is their process: “Once the patient shows up, that is when it is removed/picked up from the on-site pharmacy, and we wait approximately 15 minutes for CAB to come to room temperature. If it’s left out more than 2 hours in a syringe, the CAB injection needs to be discarded.”

  • Clinics need to make sure that they can store the medication, which may be an issue for some sites. A pharmacy may deliver the medication to a clinic (after dispensing), but a staff member needs to accept it (ie, it can’t arrive on a weekend or after hours and be left unrefrigerated). On-site pharmacies in clinical settings may not have these issues.

TelePrEP/Telehealth

Starting in the mid-20th century, telemedicine programs began to emerge as a novel option to provide remote clinical examination and maintenance health support.112,113 Since that time, the use of telemedicine, particularly video visits, has expanded to include telehealth and mobile health applications.114 Telehealth is different from telemedicine. Telemedicine refers specifically to remote clinical services. Telehealth is a broader scope of remote health care services and may include clinical services and nonclinical services, such as provider training, administrative meetings, and continuing education.115

During the SARS-CoV-2 (COVID-19) pandemic, the use of telehealth dramatically increased, and it is anticipated that telehealth will remain a frequently utilized modality for patient encounters. Telehealth is very helpful for chronic disease management (eg, patient taking HIV PrEP) by allowing continuity of care for high-risk populations while maintaining social distancing and reducing the risk for exposure to infection.116 As such, pharmacists providing HIV PrEP services to patients must gain competence in applying telehealth to their practices. The CDC guidelines list recommendations for adequately implementing the following procedures for care related to HIV PrEP45:

  • Conduct PrEP screening, initiation, and follow-up visits by phone or web-based consultation with clinicians

  • Obtain specimens for HIV and STI laboratory tests by:

    • Laboratory visits for specimen collection only

    • Order home specimen collection kits for
      specified tests

      • Specimen kits are sent to the patient’s home and contain supplies to collect blood from a fingerstick or other appropriate method

      • The kit is then mailed back to the laboratory with results returned to the clinician who acts on results accordingly and promptly

  • When an HIV-negative test is confirmed, provide a prescription for a 3-month supply of oral PrEP medication as opposed to a 1-month supply to minimize trips to the pharmacy

Concluding Comments

HIV Prevention: The Pharmacist as Educator, Provider, and Advocate

This monograph has outlined the pharmacist’s role in the provision of HIV PrEP and PEP. A comprehensive nationwide protocol allowing pharmacists to provide nationally endorsed public health preventive care is a win for patient access to care, and a strike against the spread of HIV. As such, it may bring the United States one step closer to ending the HIV epidemic.

The pharmacy setting is an ideal entry point for people at risk of HIV acquisition to initiate pre- or postexposure HIV prophylaxis. The pharmacist’s role as HIV-prevention educator, PrEP/PEP provider, and patient advocate has the potential to grow into a greater presence within the local public health sphere. This expansion of the pharmacist’s scope of practice can potentially help build strong relationships with patients, PCPs, and community-based patient-support services, while establishing the pharmacist as an essential and reliable resource. Provision of PrEP and PEP services by well- trained and well-placed pharmacists can be a driving force in attaining the goal of zero new HIV infections.

List of Abbreviations

Ab

antibody

Ag

antigen

AHI

acute HIV infection

AIDS

acquired immunodeficiency syndrome

ALT

alanine aminotransferase

AST

aspartate aminotransferase

ART

antiretroviral therapy

CAB

cabotegravir

CDC

Centers for Disease Control and Prevention

CDPHE

Colorado Department of Public Health & Environment

CI

confidence interval

CLIA

Clinical Laboratory Improvement Amendments

CT

chlamydia trachomatis

DAA

direct-acting antiviral

DDI

drug-drug interaction

DEXA

dual-energy X-ray absorptiometry

DHHS

US Department of Health and Human Services

DNA

deoxyribonucleic acid

DRV

darunavir

DTG

dolutegravir

EBR/GZR

elbasvir/grazoprevir

eCrCl

estimated creatinine clearance

eGFR

estimated glomerular filtration rate

EMR

electronic medical record

GC

gonorrhea

GLE/PIB

glecaprevir/pibrentasvir

FDA

US Food and Drug Administration

FEM-PrEP

Preexposure Prophylaxis Trial for HIV Prevention Among African Women

FTC

emtricitabine

Gen

generation

HAV

hepatitis A virus

HBV

hepatitis B virus

HCV

hepatitis C virus

HDL

high-density lipoprotein

HIPAA

Health Insurance Portability and Accountability Act

HIV

human immunodeficiency virus

HPV

human papillomavirus

IA

immunoassay

IgG

immunoglobulin G

IgM

immunoglobulin M

IAS–USA

International Antiviral Society–USA

IDU

injection drug use

iPrEx

Iniciativa Profilaxis Pre-Exposición [Preexposure Prophylaxis Initiative]

INSTI

integrase strand transfer inhibitor

IPV

intimate partner violence

IRR

incidence rate ratio

LAI

long-acting injection

LDL

low-density lipoprotein

LGBTQIA

lesbian, gay, bisexual, transgender, queer, intersex, asexual

MSM

men who have sex with men

NAAT

nucleic acid amplification testing

NNRTI

nonnucleoside reverse transcriptase inhibitor

NRTI

nucleoside reverse transcriptase inhibitor

NSAID

nonsteroidal anti-inflammatory drug

NI

noninferiority

NIH

National Institutes of Health

nPEP

nonoccupational PEP

PCP

primary care provider

PCR

polymerase chain reaction

PEP

postexposure prophylaxis

PI

protease inhibitor

PWH/PLWH

people with HIV/people living with HIV

PrEP

preexposure prophylaxis

PTSD

posttraumatic stress disorder

PY

person-years

PWID

people who inject drugs

RAL

raltegravir

RNA

ribonucleic acid

RTV

ritonavir

SAMHSA

Substance Abuse and Mental Health Services Administration

SBOP

State Board of Pharmacy

SOF/LDV

sofosbuvir/ledipasvir

SOF/VEL

sofosbuvir/velpatasvir

STI

sexually transmitted infection

SUD

substance use disorder

TAF

tenofovir alafenamide

TDF

tenofovir disoproxil fumarate

TDF2

Botswana TDF/FTC Oral HIV Prophylaxis trial

TFV-DP

tenofovir diphosphate.

TGW

transgender women

TIA

trauma-informed approach

TIC

trauma-informed care

WHO

World Health Organization

REFERENCES

  1. Smith DK, et al. MMWR Morb Mortal Wkly Rep. 2015;64(46):1291-1295.
  2. Aidsmap. PrEP use growing in US, but not reaching all those in need. https://www.aidsmap.com/news/mar-2018/prep-use-growing-us-not-reaching-all-those-need. Accessed May 24, 2022.
  3. US Department of Health & Human Services (DHHS). What is ending the HIV epidemic: a plan for America? June 2021. https://www.hiv.gov/federal-response/ending-the-hiv-epidemic/overview. Accessed May 24, 2022.
  4. Lopez MI, et al. J Am Pharm Assoc (2003). 2020;60(4):e18-e24.
  5. TheBodyPro. Oregon Poised to Be Third State to Allow Pharmacist-Prescribed PrEP. https://www.thebodypro.com/article/oregon-third-state-allow-pharmacist-prescribed-prep. Accessed May 24, 2022.
  6. Khosropour CM, et al. AIDS Patient Care STDS. 2020;34(1):1-6.
  7. Henderson KC, et al. AIDS Patient Care STDS. 2011;25(4):221-228.
  8. Ma A, et al. AIDS Care. 2010;22(10):1189-1194.
  9. Saberi P, et al. Patient Prefer Adherence. 2012;6:297-322.
  10. Siegler AJ, et al. Ann Epidemiol. 2018;28(12):841-849.
  11. Sullivan PS, Siegler AJ. Sex Health. 2018;15(6):522-527.
  12. CDC. HIV Surveillance Report: Diagnoses of HIV Infection in the United States and Dependent Areas, 2019. https://www.cdc.gov/hiv/pdf/library/reports/surveillance/cdc-hiv-surveillance-report-2018-updated-vol-32.pdf. Accessed March 20, 2022.
  13. CDC. The state of the HIV epidemic in the US. Updated October 2021. https://www.cdc.gov/nchhstp/newsroom/fact-sheets/hiv/state-of-the-hiv-epidemic-factsheet.html . Accessed June 1, 2022.
  14. Centers for Disease Control and Prevention (CDC). HIV basics: US statistics. Updated June 30, 2020. https://www.hiv.gov/hiv-basics/overview/data-and-trends/statistics. Accessed May 24, 2022.
  15. CDC. Policy, Planning and Strategic Communication. Dear colleagues: information from CDC’s Division of HIV/AIDS Prevention. https://www.cdc.gov/hiv/policies/dear-colleague/dcl/120821.html. Accessed May 24, 2022.
  16. Roser M, Richie H. HIV/AIDS. Updated November 2019. https://ourworldindata.org/hiv-aids. Accessed May 24, 2022.
  17. Kochanek KD, et al. Natl Vital Stat Rep. 2019;68(9):1-77.
  18. Oguntibeju OO. HIV AIDS (Auckl). 2012;4:117-124.
  19. CDC. Pre-exposure prophylaxis (PrEP) and post-exposure prophylaxis (PEP). Updated September 2020. https://www.cdc.gov/hiv/clinicians/prevention/prep-and-pep.html. Accessed May 24, 2022.
  20. CDC. PrEP for HIV prevention in the U.S. https://www.cdc.gov/nchhstp/newsroom/fact-sheets/hiv/PrEP-for-hiv-prevention-in-the-US-factsheet.html#section2; Accessed June 15, 2022.
  21. DHHS. Ending the HIV epidemic: Ready, Set, PrEP. Updated March 2022. https://www.hiv.gov/federal-response/ending-the-hiv-epidemic/prep-program. Accessed May 24, 2022.
  22. Choopanya K, et al. Lancet. 2013;381(9883):2083-2090.
  23. Anderson PL, et al. Sci Transl Med. 2012;4(151):151ra125.
  24. Kazi DS, et al. N Engl J Med. 2019;381(26):2489-2491.
  25. Hess KL, et al. Ann Epidemiol. 2017;27(4):238-243.
  26. Goldstein RH, et al. N Engl J Med. 2018;379(14):1293-1295.
  27. Cahill S, et al. AIDS Care. 2017;29(11):1351-1358.
  28. Highleyman L. First generic Truvada now available in the United States. POZ Online. October 2, 2020. https://www.poz.com/article/first-generic-truvada-now-available-united-states. Accessed May 24, 2022.
  29. State of Colorado. House Bill. 20-1061. July 13, 2020. https://leg.colorado.gov/bills/hb20-1061. Accessed May 24, 2022.
  30. CDC. Life-saving naloxone from pharmacies. Updated August 6, 2019. https://www.cdc.gov/vitalsigns/naloxone/index.html. Accessed May 24, 2022.
  31. Newman TV, et al. J Manag Care Spec Pharm. 2019;25(9):995-1000.
  32. Kibicho J, Owczarzak J. J Am Pharm Assoc (2003). 2011;51(6):746-755.
  33. Hill LA, et al. AIDS Rev. 2019;21(4):195-210.
  34. Tseng A, et al. Can J Hosp Pharm. 2012;65(2):125-145.
  35. Brown J. As HIV infection rates rise in Colorado, pharmacies can now prescribe preventative pills. The Colorado Sun. November 18, 2020. . Accessed January 21, 2021.
  36. Colorado Department of Public Health and Environment (CDPHE). HIV surveillance quarterly reports. https://cdphe.colorado.gov/sexually-transmitted-infections-and-hiv/sti-hiv-data/hiv-surveillance-quarterly-reports. Accessed January 21, 2021.
  37. DHHS. U.S. statistics: fast facts. Updated June 2021. https://www.hiv.gov/hiv-basics/overview/data-and-trends/statistics. Accessed May 24, 2022.
  38. State of Colorado. Colorado revised statutes 2019. Title 12; 12 280 103. https://leg.colorado.gov/sites/default/files/images/olls/crs2019-title-12.pdf. Accessed January 22, 2021.
  39. ASHP. California Readies for Implementation of Historic Law Aimed at Curbing HIV. https://www.ashp.org/News/2020/02/13/California-Readies-for-Implementation-of-Historic-Law-Aimed-at-Curbing-HIV?loginreturnUrl=SSOCheckOnly. Accessed May 24, 2022.
  40. Avert. The science of HIV and AIDS–overview. Last updated April 2022. https://www.beintheknow.org/hiv-and-stis/understanding-hiv-and-aids. Accessed May 24, 2022.
  41. Cummins NW, Badley AD. Mol Cell Ther. 2014;2:20.
  42. HIVinfo.NIH.gov. HIV Overview: The HIV life cycle. Updated August 2021. https://hivinfo.nih.gov/understanding-hiv/fact-sheets/hiv-life-cycle. Accessed May 24, 2022.
  43. Liu Y, Tran N. Best practices for HIV-1/2 screening: when to test and what to test. https://blog.ucdmc.ucdavis.edu/labbestpractice/?s=best+practices+for+HIV-1%2F2+screening.
    Accessed May 24, 2022.
  44. Patel P, et al. J Clin Virol. 2012;54(1):42-47.
  45. US Public Health Service. Preexposure Prophylaxis for the Prevention of HIV Infection in the United States—2021 Update (A Clinical Practice Guideline). https://www.cdc.gov/hiv/pdf/risk/prep/cdc-hiv-prep-guidelines-2021.pdf. Accessed May 24, 2022.
  46. HIV.gov. CDC Dear colleague message. https://www.hiv.gov/blog/cdc-dear-colleague-message-updated-prep-guidelines. Accessed April 1, 2022
  47. Grant RM, et al. N Engl J Med. 2010;363(27):2587-2599.
  48. Baeten JM, et al. N Engl J Med. 2012;367(5):399-410.
  49. Murnane PM, et al. AIDS. 2013;27(13):2155-2160.
  50. Van Damme L, et al. N Engl J Med. 2012;367(5):411-422.
  51. Thigpen MC, et al. N Engl J Med. 2012;367(5):423-434.
  52. Molina J-M, et al. N Engl J Med. 2015;373(23):2237-2246.
  53. US Food and Drug Administration (FDA). Drugs@FDA: FDA- Approved Drugs. Emtricitabine and tenofovir disoproxil fumarate (Truvada®; package insert). Last updated June 2020. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/021752s061lbl.pdf. Accessed May 24, 2022.
  54. Pau AK, George JM. Infect Dis Clin North Am. 2014;28(3):371-402.
  55. Mayer KH, et al. Lancet. 2020;396(10246):239-254.
  56. Ogbuagu O, et al. Lancet HIV. 2021;8(7):e397-e407.
  57. FDA. Drugs@FDA: FDA-Approved Drugs. Emtricitabine and tenofovir alafenamide (Descovy®; package insert). Last updated January 2022. https://www.gilead.com/~/media/files/pdfs/medicines/hiv/descovy/descovy_pi.pdf?la=en. Accessed June 16, 2022.
  58. Cockcroft DW, Gault MH. Nephron. 1976;16(1):31-41.
  59. Chan L, et al. Curr Opin Nephrol Hypertens. 2018;27(2):102-112.
  60. Tao X, et al. Int J Infect Dis. 2020;93:108-117.
  61. DHHS. Guidelines for the Use of Antiretroviral Agents in Adults and Adolescents Living with HIV. https://clinicalinfo.hiv.gov/en/guidelines/hiv-clinical-guidelines-adult-and-adolescent-arv/whats-new-guidelines. Accessed June 16, 2022.
  62. Gandhi M, et al. Lancet HIV. 2016;3(11):e521-e528.
  63. Shah S, et al. AIDS. 2021:35(suppl 2):S189-S195.
  64. Wood BR, Huhn GD. Open Forum Infect Dis. 2021;8(12):ofab542. 
  65. University of Liverpool. HEP Drug Interactions. https://www.hep-druginteractions.org/checker. Accessed May 24, 2022.
  66. Cabotegravir. Package insert. ViiV Healthcare; 2021. https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/215499s000lbl.pdf. Accessed July 6, 2022.
  67. Glidden DV, et al. Clin Infect Dis. 2016;62(9):1172-1177.
  68. FDA. FDA approves first injectable treatment for HIV pre-exposure prevention. https://www.fda.gov/news-events/press-announcements/fda-approves-first-injectable-treatment-hiv-pre-exposure-prevention. Accessed May 24, 2022.
  69. Landovitz RJ, et al. N Engl J Med. 2021;385(7):595-608.
  70. Delany-Moretlwe S, et al. Lancet. 2022;399(10337):1779-1789.
  71. World Health Organization (WHO). Trial results reveal that long-acting injectable cabotegravir as PrEP is highly effective in preventing HIV acquisition in women. November 9, 2020. https://www.who.int/news/item/09-11-2020-trial-results-reveal-that-long-acting-injectable-cabotegravir-as-prep-is-highly-effective-in-preventing-hiv-acquisition-in-women. Accessed May 24, 2022.
  72. CDC. HIV Testing. Last reviewed June 2022. https://www.cdc.gov/hiv/testing/index.html. Accessed May 24, 2022.
  73. CDC. Rapid HIV tests suitable for use in non-clinical settings (CLIA- waived). Updated June 11, 2018. https://www.cdc.gov/hiv/pdf/testing/rapid-hiv-tests-non-clinical.pdf. Accessed May 24, 2022.
  74. Mascolini M. RNA assay spots HIV earlier in CAB-LA PrEP users, may avert resistance. Conference on retroviruses and opportunistic infections virtual (CROI). 2022. February 12-16, 2022. https://www.natap.org/2022/CROI/croi_58.htm; Accessed June 15, 2022.
  75. Saag MS, et al. JAMA. 2018;320(4):379-396.
  76. WHO. What is the 2+1+1? Event- driven oral pre-exposure prophylaxis to prevent HIV for men who have sex with men: update to WHO’s recommendation on oral PrEP. July 2019. https://www.who.int/publications/i/item/what-s-the-2-1-1-event-driven-oral-pre-exposure-prophylaxis-to-prevent-hiv-for-men-who-have-sex-with-men. Accessed June 16, 2022.
  77. LaPreze J, Nowicki B. US Pharm. 2022;47(3):38-42.
  78. CDC. PrEP effectiveness. Last reviewed June 2022. https://www.cdc.gov/hiv/basics/prep/prep-effectiveness.html. Accessed June 15, 2022.
  79. Lambers F, et al. BMC Infect Dis. 2018;18(1):678.
  80. Stein R, et al. MMWR Morb Mortal Wkly Rep. 2017;66(24):629-635.
  81. Frieden TR, Foti KE, Mermin J. N Engl J Med. 2015;373(23):2281-2287.
  82. Hosek SG, et al. JAMA Pediatr. 2017;171(11):1063-1071.
  83. Havens PL, et al. Clin Infect Dis. 2020;70(4):687-691.
  84. Cairns G. Does PrEP cause more HIV drug resistance? Yes, some. NAM AIDSmap. February 2020. https://www.aidsmap.com/news/feb-2020/does-prep-cause-more-hiv-drug-resistance-yes-some. Accessed May 24, 2022.
  85. Fonner VA, et al. AIDS. 2016;30(12):1973-1983.
  86. Gibas KM, et al. Drugs. 2019;79(6):609-619.
  87. Lehman DA, et al. J Infect Dis. 2015;211(8):1211-1218.
  88. Knox DC, et al. N Engl J Med. 2017;376(5):501-502.
  89. Michienzi SM, et al. Curr Infect Dis Rep. 2021;23(5):7.
  90. CDC. HIV treatment as prevention. Last reviewed June 2022. https://www.cdc.gov/hiv/risk/art/index.html. Accessed May 24, 2022.
  91. Yombi JC, Mertes H. AIDS Rev. 2018;20(3):131-140.
  92. CDC. Updated Guidelines for Antiretroviral Postexposure Prophylaxis After Sexual, Injection Drug Use, or Other Nonoccupational Exposure to HIV—United States, 2016. https://www.cdc.gov/hiv/pdf/programresources/cdc-hiv-npep-guidelines.pdf. Accessed May 24, 2022.
  93. FDA. Drugs@FDA: FDA-Approved Drugs. Raltegravir package insert. Last updated August 2013. https://www.accessdata.fda.gov/drugsatfda_docs/label/2013/022145s029lbl.pdf.
    Accessed May 24, 2022.
  94. FDA. Drugs@FDA: FDA-Approved Drugs. Dolutegravir package insert. June 2020. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/204790s025lbl.pdf. Accessed May 24, 2022.
  95. Zash R, et al. N Engl J Med. 2019;381(9):827-840.
  96. Workowski J, et al. MMWR Recomm Rep. 2021;70(4):1-192.
  97. National Association of Community Health Centers, National LGBT Health Education Center. Taking Routine Histories of Sexual Health: A System-Wide Approach for Health Centers. February 2016. https://www.lgbtqiahealtheducation.org/publication/taking-routine-histories-of-sexual-health-a-system-wide-approach-for-health-centers/. Accessed June 16, 2022.
  98. TargetHIV. Sexual History-Taking Toolkit. July 2020. https://targethiv.org/library/sexual-history-taking-toolkit. Accessed May 24, 2022.
  99. CDC. Discussing sexual health with your patients. Last reviewed October 2019. https://www.cdc.gov/hiv/clinicians/screening/discussing-sexual-health.html. Accessed May 24, 2022.
  100. Pakpreo P. Virtual Mentor. 2005;7(10):677-682.
  101. US Public Health Service. Preexposure Prophylaxis for the Prevention of HIV Infection in the United States – 2021 Update. Clinical Providers’ Supplement. https://www.cdc.gov/hiv/pdf/risk/prep/cdc-hiv-prep-provider-supplement-2021.pdf. Accessed May 24, 2022.
  102. Substance Abuse and Mental Health Services Administration. https://www.samhsa.gov. Accessed May 24, 2022.
  103. American Academy of HIV Medicine (AAHIVM). Referral Link. https://providers.aahivm.org/referral-link-search?reload=timezone. Accessed May 24, 2022.
  104. NASTAD. PrEP/PEP Access: State PrEP Assistance Programs. https://nastad.org/prepcost-resources/prep-assistance-programs. Accessed May 24, 2022.
  105. CDC. A guide to taking a sexual history. https://www.cdc.gov/std/treatment/sexualhistory.pdf. Accessed May 24, 2022.
  106. Miller J. Increasing access to HIV prevention medicine: Healthforce Center at UCSF. November 19, 2019. https://healthforce.ucsf.edu/blog-article/healthcare-policy/increasing-access-hiv-prevention-medication. Accessed May 24, 2022.
  107. Tung EL, et al. Sex Health. 2018;15(6):556-561.
  108. CDC. Advancing Team-Based Care Through Collaborative Practice Agreements: A Resource and Implementation Guide for Adding Pharmacists to the Care Team. https://www.cdc.gov/dhdsp/pubs/docs/cpa-team-based-care.pdf. Accessed May 24, 2022.
  109. CDC. Collaborative Practice Agreements and Pharmacists’ Patient Care Services: A Resource for Pharmacists. https://www.cdc.gov/dhdsp/pubs/docs/translational_tools_pharmacists.pdf.
    Accessed May 24, 2022.
  110. DHHS. HIPAA for Professionals. Last reviewed May 2021. https://www.hhs.gov/hipaa/for-professionals/index.html. Accessed May 24, 2022.
  111. Badowski M, PharmD. University of Illinois at Chicago, College of Pharmacy. Personal communication. June 16, 2022.
  112. Dwyer TF. Am J Psychiatry. 1973;130(8):865-869.
  113. Mermelstein H, et al. J Technol Behav Sci. 2017;2:5.
  114. Weinstein RS, et al. Am J Med. 2014;127(3):183-187.
  115. Health.IT.gov. What is telehealth? Reviewed October 2019. https://www.healthit.gov/faq/what-telehealth-how-telehealth-different-telemedicine. Accessed June 16, 2022.
  116. Lakkireddy DR, et al. Circulation. 2020;141(21):e823-e831.
  117. SAMHSA (Substance Abuse and Mental Health Services Administration). Concept of trauma and guidance for a trauma-informed approach. July 2014. https://ncsacw.acf.hhs.gov/userfiles/files/SAMHSA_Trauma.pdf. Accessed July 12, 2022.
  118. Sales JM, et al. Curr HIV/AIDS Rep. 2016;13(6):374-382.
  119. Trauma-informed care implementation resource center. What is trauma-informed care? https://www.traumainformedcare.chcs.org/what-is-trauma-informed-care/. Accessed July 12, 2022.
  120. Willie TC, et al. Womens Health Issues. 2021;31(5):415-419.
  121. Young LB, et al. Int J STD AIDS. 2020;31(5):440-446.
  122. Daiber D. Sexual assault: PEP is an urgent health need. 2018. https://aidsetc.org/blog/sexual-assault-pep-urgent-health-need. Accessed July 12, 2022.
  123. Makadon HJ, et al. Fenway guide to lesbian, gay, bisexual, and transgender health. 2nd ed. 2015. American College of Physicians.
  124. NASTAD. Trauma-informed approaches toolkit. 2018. https://nastad.org/sites/default/files/2021-12/PDF-Trauma-Informed-Approaches.pdf. Accessed July 18, 2022.
  125. Miller E. Pediatr Ann. 2019;48(7):274-279.