User login
TP53 mutations predict inferior outcomes in newly diagnosed aggressive BCL
Key clinical point: TP53 mutations detected by clinical laboratory mutation analysis (CLMA) can independently predict poor outcomes in patients with newly diagnosed diffuse large B cell lymphoma (DLBCL) or high-grade B cell lymphoma (HGBL) treated with first-line immunochemotherapy.
Major finding: TP53 mutations significantly predicted disease progression at 2 years (adjusted hazard ratio 2.3; P = .03). Patients with vs without TP53 mutations had significantly lower overall response (71% vs 90%; P = .009), complete response (55% vs 77%, P = .01), estimated 2-year progression-free survival (57% vs 77%; P = .006), and estimated 2-year overall survival (70% vs 91%; P = .001) rates.
Study details: This study included 122 patients with newly diagnosed DLBCL or HGBL receiving first-line immunochemotherapy whose diagnostic biopsies underwent CLMA, of whom 42 patients had TP53 mutations.
Disclosures: This study did not disclose any funding source. All authors, except A Bagg, declared receiving honoraria, travel grants, or research funding from or having other ties with various sources.
Source: Landsburg DJ et al. TP53 mutations predict for poor outcomes in patients with newly-diagnosed aggressive B cell lymphomas in the current era. Blood Adv. 2023 (Oct 18). doi: 10.1182/bloodadvances.2023011384
Key clinical point: TP53 mutations detected by clinical laboratory mutation analysis (CLMA) can independently predict poor outcomes in patients with newly diagnosed diffuse large B cell lymphoma (DLBCL) or high-grade B cell lymphoma (HGBL) treated with first-line immunochemotherapy.
Major finding: TP53 mutations significantly predicted disease progression at 2 years (adjusted hazard ratio 2.3; P = .03). Patients with vs without TP53 mutations had significantly lower overall response (71% vs 90%; P = .009), complete response (55% vs 77%, P = .01), estimated 2-year progression-free survival (57% vs 77%; P = .006), and estimated 2-year overall survival (70% vs 91%; P = .001) rates.
Study details: This study included 122 patients with newly diagnosed DLBCL or HGBL receiving first-line immunochemotherapy whose diagnostic biopsies underwent CLMA, of whom 42 patients had TP53 mutations.
Disclosures: This study did not disclose any funding source. All authors, except A Bagg, declared receiving honoraria, travel grants, or research funding from or having other ties with various sources.
Source: Landsburg DJ et al. TP53 mutations predict for poor outcomes in patients with newly-diagnosed aggressive B cell lymphomas in the current era. Blood Adv. 2023 (Oct 18). doi: 10.1182/bloodadvances.2023011384
Key clinical point: TP53 mutations detected by clinical laboratory mutation analysis (CLMA) can independently predict poor outcomes in patients with newly diagnosed diffuse large B cell lymphoma (DLBCL) or high-grade B cell lymphoma (HGBL) treated with first-line immunochemotherapy.
Major finding: TP53 mutations significantly predicted disease progression at 2 years (adjusted hazard ratio 2.3; P = .03). Patients with vs without TP53 mutations had significantly lower overall response (71% vs 90%; P = .009), complete response (55% vs 77%, P = .01), estimated 2-year progression-free survival (57% vs 77%; P = .006), and estimated 2-year overall survival (70% vs 91%; P = .001) rates.
Study details: This study included 122 patients with newly diagnosed DLBCL or HGBL receiving first-line immunochemotherapy whose diagnostic biopsies underwent CLMA, of whom 42 patients had TP53 mutations.
Disclosures: This study did not disclose any funding source. All authors, except A Bagg, declared receiving honoraria, travel grants, or research funding from or having other ties with various sources.
Source: Landsburg DJ et al. TP53 mutations predict for poor outcomes in patients with newly-diagnosed aggressive B cell lymphomas in the current era. Blood Adv. 2023 (Oct 18). doi: 10.1182/bloodadvances.2023011384
Allogeneic HSCT offers a salvage treatment option for chemo-susceptible relapsed or refractory DLBCL
Key clinical point: Allogeneic hematopoietic stem cell transplantation (allo-HSCT) led to acceptable survival outcomes in patients with relapsed or refractory diffuse large B cell lymphoma (DLBCL), particularly in those achieving a partial or complete response to chemotherapy before allo-HSCT.
Major finding: At a median follow-up of 38.3 months, the estimated 5-year overall survival and event-free survival rates were 38.4% (95% CI 24.7%-51.8%) and 30.6% (95% CI 18.8%-43.3%), respectively, with patients who achieved a partial or complete response before allo-HSCT having overall survival and event-free survival rates of 54.1% (95% CI 34.2%-70.3%) and 46.4% (95% CI 28.1%-62.9%), respectively.
Study details: This retrospective study included 52 adult patients with relapsed or refractory DLBCL who either had an active disease status or achieved a partial or complete response before transplantation and underwent allo-HSCT.
Disclosures: This study did not disclose any funding source. The authors declared no conflicts of interest.
Source: Min GJ et al. The salvage role of allogeneic hematopoietic stem-cell transplantation in relapsed/refractory diffuse large B cell lymphoma. Sci Rep. 2023;13:17496 (Oct 15). doi: 10.1038/s41598-023-44241-0
Key clinical point: Allogeneic hematopoietic stem cell transplantation (allo-HSCT) led to acceptable survival outcomes in patients with relapsed or refractory diffuse large B cell lymphoma (DLBCL), particularly in those achieving a partial or complete response to chemotherapy before allo-HSCT.
Major finding: At a median follow-up of 38.3 months, the estimated 5-year overall survival and event-free survival rates were 38.4% (95% CI 24.7%-51.8%) and 30.6% (95% CI 18.8%-43.3%), respectively, with patients who achieved a partial or complete response before allo-HSCT having overall survival and event-free survival rates of 54.1% (95% CI 34.2%-70.3%) and 46.4% (95% CI 28.1%-62.9%), respectively.
Study details: This retrospective study included 52 adult patients with relapsed or refractory DLBCL who either had an active disease status or achieved a partial or complete response before transplantation and underwent allo-HSCT.
Disclosures: This study did not disclose any funding source. The authors declared no conflicts of interest.
Source: Min GJ et al. The salvage role of allogeneic hematopoietic stem-cell transplantation in relapsed/refractory diffuse large B cell lymphoma. Sci Rep. 2023;13:17496 (Oct 15). doi: 10.1038/s41598-023-44241-0
Key clinical point: Allogeneic hematopoietic stem cell transplantation (allo-HSCT) led to acceptable survival outcomes in patients with relapsed or refractory diffuse large B cell lymphoma (DLBCL), particularly in those achieving a partial or complete response to chemotherapy before allo-HSCT.
Major finding: At a median follow-up of 38.3 months, the estimated 5-year overall survival and event-free survival rates were 38.4% (95% CI 24.7%-51.8%) and 30.6% (95% CI 18.8%-43.3%), respectively, with patients who achieved a partial or complete response before allo-HSCT having overall survival and event-free survival rates of 54.1% (95% CI 34.2%-70.3%) and 46.4% (95% CI 28.1%-62.9%), respectively.
Study details: This retrospective study included 52 adult patients with relapsed or refractory DLBCL who either had an active disease status or achieved a partial or complete response before transplantation and underwent allo-HSCT.
Disclosures: This study did not disclose any funding source. The authors declared no conflicts of interest.
Source: Min GJ et al. The salvage role of allogeneic hematopoietic stem-cell transplantation in relapsed/refractory diffuse large B cell lymphoma. Sci Rep. 2023;13:17496 (Oct 15). doi: 10.1038/s41598-023-44241-0
Ibrutinib-venetoclax tops chlorambucil-obinutuzumab in treatment-naïve CLL over a 4-year follow-up
Key clinical point: Fixed-duration ibrutinib-venetoclax vs chlorambucil-obinutuzumab continues to extend progression-free survival and leads to overall survival advantage at a 4-year follow-up in patients with previously untreated chronic lymphocytic leukemia (CLL).
Major finding: At a median follow-up of 46 months, the ibrutinib-venetoclax vs chlorambucil-obinutuzumab group continued to show better progression-free survival (hazard ratio [HR] 0.214; P < .0001) while also demonstrating overall survival benefit (HR 0.487; P = .021). One treatment-related death was reported in each group.
Study details: Findings are from a 4-year follow-up of the phase 3 GLOW trial including 211 patients with previously untreated CLL who were randomly assigned to receive ibrutinib-venetoclax or chlorambucil-obinutuzumab.
Disclosures: This study was funded by Janssen Research & Development and Pharmacyclics. Several authors declared serving on the boards of directors or advisory committees of or receiving consultancy fees, research funding, or honoraria from various sources, including Janssen. Nine authors declared being employees or equity holders of Janssen.
Source: Niemann CU et al. Fixed-duration ibrutinib–venetoclax versus chlorambucil–obinutuzumab in previously untreated chronic lymphocytic leukaemia (GLOW): 4-Year follow-up from a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2023 (Nov 6). doi: 10.1016/S1470-2045(23)00452-7
Key clinical point: Fixed-duration ibrutinib-venetoclax vs chlorambucil-obinutuzumab continues to extend progression-free survival and leads to overall survival advantage at a 4-year follow-up in patients with previously untreated chronic lymphocytic leukemia (CLL).
Major finding: At a median follow-up of 46 months, the ibrutinib-venetoclax vs chlorambucil-obinutuzumab group continued to show better progression-free survival (hazard ratio [HR] 0.214; P < .0001) while also demonstrating overall survival benefit (HR 0.487; P = .021). One treatment-related death was reported in each group.
Study details: Findings are from a 4-year follow-up of the phase 3 GLOW trial including 211 patients with previously untreated CLL who were randomly assigned to receive ibrutinib-venetoclax or chlorambucil-obinutuzumab.
Disclosures: This study was funded by Janssen Research & Development and Pharmacyclics. Several authors declared serving on the boards of directors or advisory committees of or receiving consultancy fees, research funding, or honoraria from various sources, including Janssen. Nine authors declared being employees or equity holders of Janssen.
Source: Niemann CU et al. Fixed-duration ibrutinib–venetoclax versus chlorambucil–obinutuzumab in previously untreated chronic lymphocytic leukaemia (GLOW): 4-Year follow-up from a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2023 (Nov 6). doi: 10.1016/S1470-2045(23)00452-7
Key clinical point: Fixed-duration ibrutinib-venetoclax vs chlorambucil-obinutuzumab continues to extend progression-free survival and leads to overall survival advantage at a 4-year follow-up in patients with previously untreated chronic lymphocytic leukemia (CLL).
Major finding: At a median follow-up of 46 months, the ibrutinib-venetoclax vs chlorambucil-obinutuzumab group continued to show better progression-free survival (hazard ratio [HR] 0.214; P < .0001) while also demonstrating overall survival benefit (HR 0.487; P = .021). One treatment-related death was reported in each group.
Study details: Findings are from a 4-year follow-up of the phase 3 GLOW trial including 211 patients with previously untreated CLL who were randomly assigned to receive ibrutinib-venetoclax or chlorambucil-obinutuzumab.
Disclosures: This study was funded by Janssen Research & Development and Pharmacyclics. Several authors declared serving on the boards of directors or advisory committees of or receiving consultancy fees, research funding, or honoraria from various sources, including Janssen. Nine authors declared being employees or equity holders of Janssen.
Source: Niemann CU et al. Fixed-duration ibrutinib–venetoclax versus chlorambucil–obinutuzumab in previously untreated chronic lymphocytic leukaemia (GLOW): 4-Year follow-up from a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2023 (Nov 6). doi: 10.1016/S1470-2045(23)00452-7
Brexu-cel vs SOC improved survival in relapsed or refractory MCL
Key clinical point: Brexucabtagene autoleucel (brexu-cel) provides survival benefit over non-chimeric antigen receptor (CAR) T-cell standard of care (SOC) in patients with relapsed or refractory mantle cell lymphoma (MCL) treated with covalent Bruton tyrosine kinase inhibitors (BTKi).
Major finding: Inverse probability weighting showed that brexu-cel vs SOC led to a significantly reduced risk for death (adjusted hazard ratio 0.38; P < .001), with the findings being similar for other adjusted comparisons.
Study details: This indirect comparison study analyzed the individual patient data of BTKi-treated patients with relapsed or refractory MCL who received brexu-cel in ZUMA-2 (n = 68) and non-CAR T-cell SOC in SCHOLAR-2 (n = 149).
Disclosures: This study was sponsored by Kite, a Gilead Company. Some authors declared participating in the data safety monitoring or advisory boards of or receiving grants, consulting fees, travel support, or honoraria for lectures, etc., from Kite, Gilead, and others. Five authors declared being employees or stockowners of Kite, Gilead, or PRECISIONheor.
Source: Hess G et al. Indirect treatment comparison of brexucabtagene autoleucel (ZUMA-2) versus standard of care (SCHOLAR-2) in relapsed/refractory mantle cell lymphoma. Leuk Lymphoma. 2023 (Oct 16). doi: 10.1080/10428194.2023.2268228
Key clinical point: Brexucabtagene autoleucel (brexu-cel) provides survival benefit over non-chimeric antigen receptor (CAR) T-cell standard of care (SOC) in patients with relapsed or refractory mantle cell lymphoma (MCL) treated with covalent Bruton tyrosine kinase inhibitors (BTKi).
Major finding: Inverse probability weighting showed that brexu-cel vs SOC led to a significantly reduced risk for death (adjusted hazard ratio 0.38; P < .001), with the findings being similar for other adjusted comparisons.
Study details: This indirect comparison study analyzed the individual patient data of BTKi-treated patients with relapsed or refractory MCL who received brexu-cel in ZUMA-2 (n = 68) and non-CAR T-cell SOC in SCHOLAR-2 (n = 149).
Disclosures: This study was sponsored by Kite, a Gilead Company. Some authors declared participating in the data safety monitoring or advisory boards of or receiving grants, consulting fees, travel support, or honoraria for lectures, etc., from Kite, Gilead, and others. Five authors declared being employees or stockowners of Kite, Gilead, or PRECISIONheor.
Source: Hess G et al. Indirect treatment comparison of brexucabtagene autoleucel (ZUMA-2) versus standard of care (SCHOLAR-2) in relapsed/refractory mantle cell lymphoma. Leuk Lymphoma. 2023 (Oct 16). doi: 10.1080/10428194.2023.2268228
Key clinical point: Brexucabtagene autoleucel (brexu-cel) provides survival benefit over non-chimeric antigen receptor (CAR) T-cell standard of care (SOC) in patients with relapsed or refractory mantle cell lymphoma (MCL) treated with covalent Bruton tyrosine kinase inhibitors (BTKi).
Major finding: Inverse probability weighting showed that brexu-cel vs SOC led to a significantly reduced risk for death (adjusted hazard ratio 0.38; P < .001), with the findings being similar for other adjusted comparisons.
Study details: This indirect comparison study analyzed the individual patient data of BTKi-treated patients with relapsed or refractory MCL who received brexu-cel in ZUMA-2 (n = 68) and non-CAR T-cell SOC in SCHOLAR-2 (n = 149).
Disclosures: This study was sponsored by Kite, a Gilead Company. Some authors declared participating in the data safety monitoring or advisory boards of or receiving grants, consulting fees, travel support, or honoraria for lectures, etc., from Kite, Gilead, and others. Five authors declared being employees or stockowners of Kite, Gilead, or PRECISIONheor.
Source: Hess G et al. Indirect treatment comparison of brexucabtagene autoleucel (ZUMA-2) versus standard of care (SCHOLAR-2) in relapsed/refractory mantle cell lymphoma. Leuk Lymphoma. 2023 (Oct 16). doi: 10.1080/10428194.2023.2268228
Preapheresis bendamustine worsens CAR T-cell therapy outcomes in relapsed or refractory LBCL
Key clinical point: Patients with relapsed or refractory large B-cell lymphoma (LBCL) who were recently exposed to preapheresis bendamustine showed negative treatment outcomes, hematologic toxicity, and severe infections after CD19-targeted chimeric antigen receptor (CAR) T-cell therapy.
Major finding: Patients recently exposed to bendamustine (<9 months) vs those naive to it before apheresis had a significantly lower overall response rate (40% vs 66%; P = .01) and shorter overall survival (adjusted hazard ratio [aHR] 2.11; P < .01) and progression-free survival (aHR 1.82; P < .01) after CAR T-cell infusion.
Study details: This retrospective multicenter study included 439 patients with relapsed or refractory LBCL who received CD19-targeted commercial CAR T-cell therapy after ≥2 prior treatment lines, of whom 80 patients had received bendamustine before apheresis.
Disclosures: This study was supported by the Carlos III Health Institute, Spain, and others. Some authors declared serving in consulting or advisory roles for or as members of speakers’ bureaus of or receiving honoraria, research funding, or travel or accommodation expenses from various sources.
Source: Iacoboni G et al. Recent bendamustine treatment before apheresis has a negative impact on outcomes in patients with large B-cell lymphoma receiving chimeric antigen receptor T-cell therapy. J Clin Oncol. 2023 (Oct 24). doi: 10.1200/JCO.23.01097
Key clinical point: Patients with relapsed or refractory large B-cell lymphoma (LBCL) who were recently exposed to preapheresis bendamustine showed negative treatment outcomes, hematologic toxicity, and severe infections after CD19-targeted chimeric antigen receptor (CAR) T-cell therapy.
Major finding: Patients recently exposed to bendamustine (<9 months) vs those naive to it before apheresis had a significantly lower overall response rate (40% vs 66%; P = .01) and shorter overall survival (adjusted hazard ratio [aHR] 2.11; P < .01) and progression-free survival (aHR 1.82; P < .01) after CAR T-cell infusion.
Study details: This retrospective multicenter study included 439 patients with relapsed or refractory LBCL who received CD19-targeted commercial CAR T-cell therapy after ≥2 prior treatment lines, of whom 80 patients had received bendamustine before apheresis.
Disclosures: This study was supported by the Carlos III Health Institute, Spain, and others. Some authors declared serving in consulting or advisory roles for or as members of speakers’ bureaus of or receiving honoraria, research funding, or travel or accommodation expenses from various sources.
Source: Iacoboni G et al. Recent bendamustine treatment before apheresis has a negative impact on outcomes in patients with large B-cell lymphoma receiving chimeric antigen receptor T-cell therapy. J Clin Oncol. 2023 (Oct 24). doi: 10.1200/JCO.23.01097
Key clinical point: Patients with relapsed or refractory large B-cell lymphoma (LBCL) who were recently exposed to preapheresis bendamustine showed negative treatment outcomes, hematologic toxicity, and severe infections after CD19-targeted chimeric antigen receptor (CAR) T-cell therapy.
Major finding: Patients recently exposed to bendamustine (<9 months) vs those naive to it before apheresis had a significantly lower overall response rate (40% vs 66%; P = .01) and shorter overall survival (adjusted hazard ratio [aHR] 2.11; P < .01) and progression-free survival (aHR 1.82; P < .01) after CAR T-cell infusion.
Study details: This retrospective multicenter study included 439 patients with relapsed or refractory LBCL who received CD19-targeted commercial CAR T-cell therapy after ≥2 prior treatment lines, of whom 80 patients had received bendamustine before apheresis.
Disclosures: This study was supported by the Carlos III Health Institute, Spain, and others. Some authors declared serving in consulting or advisory roles for or as members of speakers’ bureaus of or receiving honoraria, research funding, or travel or accommodation expenses from various sources.
Source: Iacoboni G et al. Recent bendamustine treatment before apheresis has a negative impact on outcomes in patients with large B-cell lymphoma receiving chimeric antigen receptor T-cell therapy. J Clin Oncol. 2023 (Oct 24). doi: 10.1200/JCO.23.01097
Clinical Consult in NASH: Are Your Patients at Risk?
Nonalcoholic steatohepatitis, or NASH,a is the most severe form of nonalcoholic fatty liver disease (NAFLD).
Read More
Nonalcoholic steatohepatitis, or NASH,a is the most severe form of nonalcoholic fatty liver disease (NAFLD).
Read More
Nonalcoholic steatohepatitis, or NASH,a is the most severe form of nonalcoholic fatty liver disease (NAFLD).
Read More
Preventing RSV in children and adults: A vaccine update
In the past year, there has been significant progress in the availability of interventions to prevent respiratory syncytial virus (RSV) and its complications. Four products have been approved by the US Food and Drug Administration (FDA) and recommended by the Centers for Disease Control and Prevention’s (CDC) Advisory Committee on Immunization Practices (ACIP). They include 2 vaccines for adults ages 60 years and older, a monoclonal antibody for infants and high-risk children, and a maternal vaccine to prevent RSV infection in newborns.
RSV in adults
While there is some uncertainty about the total burden of RSV in adults in the United States, the CDC estimates that each year it causes 0.9 to 1.4 million medical encounters, 60,000 to 160,000 hospitalizations, and 6000 to 10,000 deaths.1 The rate of RSV-caused hospitalization increases with age,2 and the infection is more severe in those with certain chronic medical conditions (TABLE 11). The FIGURE2 demonstrates the outcomes of adults who are hospitalized for RSV. Adults older than 65 years have a 5% mortality rate if hospitalized for RSV infection.2
Vaccine options for adults
Two vaccines were recently approved for the prevention of RSV-associated lower respiratory tract disease (LRTD) in those ages 60 years and older: RSVPreF3 (Arexvy, GSK), which is an adjuvanted recombinant F protein vaccine, and RSVpreF (Abrysvo, Pfizer), which is a recombinant stabilized vaccine. Both require only a single dose (0.5 mL IM), which provides protection for 2 years.
The efficacy of the GSK vaccine in preventing laboratory-confirmed, RSV-associated LRTD was 82.6% during the first RSV season and 56.1% during the second season. The efficacy of the Pfizer vaccine in preventing symptomatic, laboratory-confirmed LRTD was 88.9% during the first RSV season and 78.6% during the second season.1 However, the trials leading to licensure of both vaccines were underpowered to show efficacy in the oldest adults and those who are frail or to show efficacy against RSV-caused hospitalization.
Safety of the adult RSV vaccines. The safety trials for both vaccines had a total of 38,177 participants. There were a total of 6 neurologic inflammatory conditions that developed within 42 days of vaccination, including 2 cases of suspected Guillain-Barré syndrome (GBS), 2 cases of possible acute disseminated encephalomyelitis, and 1 case each of chronic inflammatory demyelinating polyneuropathy and undifferentiated motor-sensory axonal polyneuropathy.1 That is a rate of 1 case of a neurologic inflammatory condition for every 6363 people vaccinated. Since the trials were not powered to determine whether the small number of cases were due to chance, postmarketing surveillance will be needed to clarify the true risk for GBS or other neurologic inflammatory events from RSV vaccination.
The lack of efficacy data for the most vulnerable older adults and the lingering questions about safety prompted the ACIP to recommend that adults ages 60 years and older may receive a single dose of RSV vaccine, using shared clinical decision-making—which is different from a routine or risk-based vaccine recommendation. For RSV vaccination, the decision to vaccinate should be based on a risk/benefit discussion between the clinician and the patient. Those most likely to benefit from the vaccine are listed in TABLE 1.1
While data on coadministration of RSV vaccines with other adult vaccines are sparse, the ACIP states that co-administration with other vaccines is acceptable.1 It is not known yet whether boosters will be needed after 2 years.
Continue to: RSV in infants and children
RSV in infants and children
RSV is the most common cause of hospitalization among infants and children in the United States. The CDC estimates that each year in children younger than 5 years, RSV is responsible for 1.5 million outpatient clinic visits, 520,000 emergency department visits, 58,000 to 80,000 hospitalizations, and 100 to 200 deaths.3 The risk for hospitalization from RSV is highest in the second and third months of life and decreases with increasing age.3
There are racial disparities in RSV severity: Intensive care unit admission rates are 1.2 to 1.6 times higher among non-Hispanic Black infants younger than 6 months than among non-Hispanic White infants, and hospitalization rates are up to 5 times higher in American Indian and Alaska Native populations.3
The months of highest RSV transmission in most locations are December through February, but this can vary. For practical purposes, RSV season runs from October through March.
Prevention in infants and children
The monoclonal antibody nirsevimab is now available for use in infants younger than 8 months born during or entering their first RSV season and children ages 8 to 19 months who are at increased risk for severe RSV disease and entering their second RSV season. Details regarding the use of this product were described in a recent Practice Alert Brief.4
Early studies on nirsevimab demonstrated 79% effectiveness in preventing medical-attended LRTD, 80.6% effectiveness in preventing hospitalization, and 90% effectiveness in preventing ICU admission. The number needed to immunize with nirsevimab to prevent an outpatient visit is estimated to be 17; to prevent an ED visit, 48; and to prevent an inpatient admission, 128. Due to the low RSV death rate, the studies were not able to demonstrate reduced mortality.5
Continue to: RSV vaccine in pregnancy
RSV vaccine in pregnancy
In August, the FDA approved Pfizer’s RSVpreF vaccine for use during pregnancy—as a single dose given at 32 to 36 weeks’ gestation—for the prevention of RSV LRTD in infants in the first 6 months of life. In the clinical trials, the vaccine was given at 24 to 36 weeks’ gestation. However, there was a statistically nonsignificant increase in preterm births in the RSVpreF group compared to the placebo group.6 While there were insufficient data to prove or rule out a causal relationship, the FDA advisory committee was more comfortable approving the vaccine for use only later in pregnancy, to avoid the possibility of very early preterm births after vaccination. The ACIP agreed.
From time of maternal vaccination, at least 14 days are needed to develop and transfer maternal antibodies across the placenta to protect the infant. Therefore, infants born less than 14 days after maternal vaccination should be considered unprotected.
Both maternal vaccination with RSVpreF and infant injection with nirsevimab are now options to protect newborns and infants from RSV. However, use of both products is not needed, since combined they do not offer significant added protection compared to either product alone (exceptions to be discussed shortly).6 When the estimated due date will occur in the RSV season, maternity clinicians should provide information on both products and assist the mother in deciding whether to be vaccinated or rely on administration of nirsevimab to the infant after birth. The benefits and risks of these 2 options are listed in TABLE 2.6
There are some rare situations in which use of both products is recommended, and they include6:
- When the baby is born less than 14 days from the time of maternal vaccination
- When the mother has a condition that could produce an inadequate response to the vaccine
- When the infant has had cardiopulmonary bypass, which would lead to loss of maternal antibodies
- When the infant has severe disease placing them at increased risk for severe RSV.
Conclusion
All of these new RSV preventive products should soon be widely available and covered with no out-of-pocket expense by commercial and government payers. The exception might be nirsevimab—because of the time needed to produce it, it might not be universally available in the 2023-2024 season.
1. Melgar M, Britton A, Roper LE, et al. Use of respiratory syncytial virus vaccine in older adults: recommendation of the Advisory Committee on Immunization Practices—United States, 2023. MMWR Morb Mortal Wkly Rep. 2023;72:793-801.
2. Melgar M. Evidence to recommendation framework. RSV in adults. Presented to the ACIP on February 23, 2023. Accessed November 7, 2023. www.cdc.gov/vaccines/acip/meetings/downloads/slides-2023-02/slides-02-23/RSV-Adults-04-Melgar-508.pdf
3. Jones JM, Fleming-Dutra KE, Prill MM, et al. Use of nirsevimab for the prevention of respiratory syncytial virus disease among infants and young children: recommendation of the Advisory Committee on Immunization Practices—United States, 2023. MMWR Morb Mortal Wkly Rep. 2023;72:90-925.
4. Campos-Outcalt D. Are you ready for RSV season? There’s a new preventive option. J Fam Pract. 2023;72. doi: 10.12788/jfp.0663
5. Jones J. Evidence to recommendation framework: nirsevimab updates. Presented to the ACIP on August 3, 2023. Accessed August 23, 2023. https://stacks.cdc.gov/view/cdc/131586
6. Jones J. Clinical considerations for maternal RSVPreF vaccine and nirsevimab. Presented to the ACIP on September 25, 2023. Accessed November 8, 2023. www2.cdc.gov/vaccines/ed/ciinc/archives/23/09/ciiw_RSV2/CIIW%20RSV%20maternal%20vaccine%20mAb%209.27.23.pdf
In the past year, there has been significant progress in the availability of interventions to prevent respiratory syncytial virus (RSV) and its complications. Four products have been approved by the US Food and Drug Administration (FDA) and recommended by the Centers for Disease Control and Prevention’s (CDC) Advisory Committee on Immunization Practices (ACIP). They include 2 vaccines for adults ages 60 years and older, a monoclonal antibody for infants and high-risk children, and a maternal vaccine to prevent RSV infection in newborns.
RSV in adults
While there is some uncertainty about the total burden of RSV in adults in the United States, the CDC estimates that each year it causes 0.9 to 1.4 million medical encounters, 60,000 to 160,000 hospitalizations, and 6000 to 10,000 deaths.1 The rate of RSV-caused hospitalization increases with age,2 and the infection is more severe in those with certain chronic medical conditions (TABLE 11). The FIGURE2 demonstrates the outcomes of adults who are hospitalized for RSV. Adults older than 65 years have a 5% mortality rate if hospitalized for RSV infection.2
Vaccine options for adults
Two vaccines were recently approved for the prevention of RSV-associated lower respiratory tract disease (LRTD) in those ages 60 years and older: RSVPreF3 (Arexvy, GSK), which is an adjuvanted recombinant F protein vaccine, and RSVpreF (Abrysvo, Pfizer), which is a recombinant stabilized vaccine. Both require only a single dose (0.5 mL IM), which provides protection for 2 years.
The efficacy of the GSK vaccine in preventing laboratory-confirmed, RSV-associated LRTD was 82.6% during the first RSV season and 56.1% during the second season. The efficacy of the Pfizer vaccine in preventing symptomatic, laboratory-confirmed LRTD was 88.9% during the first RSV season and 78.6% during the second season.1 However, the trials leading to licensure of both vaccines were underpowered to show efficacy in the oldest adults and those who are frail or to show efficacy against RSV-caused hospitalization.
Safety of the adult RSV vaccines. The safety trials for both vaccines had a total of 38,177 participants. There were a total of 6 neurologic inflammatory conditions that developed within 42 days of vaccination, including 2 cases of suspected Guillain-Barré syndrome (GBS), 2 cases of possible acute disseminated encephalomyelitis, and 1 case each of chronic inflammatory demyelinating polyneuropathy and undifferentiated motor-sensory axonal polyneuropathy.1 That is a rate of 1 case of a neurologic inflammatory condition for every 6363 people vaccinated. Since the trials were not powered to determine whether the small number of cases were due to chance, postmarketing surveillance will be needed to clarify the true risk for GBS or other neurologic inflammatory events from RSV vaccination.
The lack of efficacy data for the most vulnerable older adults and the lingering questions about safety prompted the ACIP to recommend that adults ages 60 years and older may receive a single dose of RSV vaccine, using shared clinical decision-making—which is different from a routine or risk-based vaccine recommendation. For RSV vaccination, the decision to vaccinate should be based on a risk/benefit discussion between the clinician and the patient. Those most likely to benefit from the vaccine are listed in TABLE 1.1
While data on coadministration of RSV vaccines with other adult vaccines are sparse, the ACIP states that co-administration with other vaccines is acceptable.1 It is not known yet whether boosters will be needed after 2 years.
Continue to: RSV in infants and children
RSV in infants and children
RSV is the most common cause of hospitalization among infants and children in the United States. The CDC estimates that each year in children younger than 5 years, RSV is responsible for 1.5 million outpatient clinic visits, 520,000 emergency department visits, 58,000 to 80,000 hospitalizations, and 100 to 200 deaths.3 The risk for hospitalization from RSV is highest in the second and third months of life and decreases with increasing age.3
There are racial disparities in RSV severity: Intensive care unit admission rates are 1.2 to 1.6 times higher among non-Hispanic Black infants younger than 6 months than among non-Hispanic White infants, and hospitalization rates are up to 5 times higher in American Indian and Alaska Native populations.3
The months of highest RSV transmission in most locations are December through February, but this can vary. For practical purposes, RSV season runs from October through March.
Prevention in infants and children
The monoclonal antibody nirsevimab is now available for use in infants younger than 8 months born during or entering their first RSV season and children ages 8 to 19 months who are at increased risk for severe RSV disease and entering their second RSV season. Details regarding the use of this product were described in a recent Practice Alert Brief.4
Early studies on nirsevimab demonstrated 79% effectiveness in preventing medical-attended LRTD, 80.6% effectiveness in preventing hospitalization, and 90% effectiveness in preventing ICU admission. The number needed to immunize with nirsevimab to prevent an outpatient visit is estimated to be 17; to prevent an ED visit, 48; and to prevent an inpatient admission, 128. Due to the low RSV death rate, the studies were not able to demonstrate reduced mortality.5
Continue to: RSV vaccine in pregnancy
RSV vaccine in pregnancy
In August, the FDA approved Pfizer’s RSVpreF vaccine for use during pregnancy—as a single dose given at 32 to 36 weeks’ gestation—for the prevention of RSV LRTD in infants in the first 6 months of life. In the clinical trials, the vaccine was given at 24 to 36 weeks’ gestation. However, there was a statistically nonsignificant increase in preterm births in the RSVpreF group compared to the placebo group.6 While there were insufficient data to prove or rule out a causal relationship, the FDA advisory committee was more comfortable approving the vaccine for use only later in pregnancy, to avoid the possibility of very early preterm births after vaccination. The ACIP agreed.
From time of maternal vaccination, at least 14 days are needed to develop and transfer maternal antibodies across the placenta to protect the infant. Therefore, infants born less than 14 days after maternal vaccination should be considered unprotected.
Both maternal vaccination with RSVpreF and infant injection with nirsevimab are now options to protect newborns and infants from RSV. However, use of both products is not needed, since combined they do not offer significant added protection compared to either product alone (exceptions to be discussed shortly).6 When the estimated due date will occur in the RSV season, maternity clinicians should provide information on both products and assist the mother in deciding whether to be vaccinated or rely on administration of nirsevimab to the infant after birth. The benefits and risks of these 2 options are listed in TABLE 2.6
There are some rare situations in which use of both products is recommended, and they include6:
- When the baby is born less than 14 days from the time of maternal vaccination
- When the mother has a condition that could produce an inadequate response to the vaccine
- When the infant has had cardiopulmonary bypass, which would lead to loss of maternal antibodies
- When the infant has severe disease placing them at increased risk for severe RSV.
Conclusion
All of these new RSV preventive products should soon be widely available and covered with no out-of-pocket expense by commercial and government payers. The exception might be nirsevimab—because of the time needed to produce it, it might not be universally available in the 2023-2024 season.
In the past year, there has been significant progress in the availability of interventions to prevent respiratory syncytial virus (RSV) and its complications. Four products have been approved by the US Food and Drug Administration (FDA) and recommended by the Centers for Disease Control and Prevention’s (CDC) Advisory Committee on Immunization Practices (ACIP). They include 2 vaccines for adults ages 60 years and older, a monoclonal antibody for infants and high-risk children, and a maternal vaccine to prevent RSV infection in newborns.
RSV in adults
While there is some uncertainty about the total burden of RSV in adults in the United States, the CDC estimates that each year it causes 0.9 to 1.4 million medical encounters, 60,000 to 160,000 hospitalizations, and 6000 to 10,000 deaths.1 The rate of RSV-caused hospitalization increases with age,2 and the infection is more severe in those with certain chronic medical conditions (TABLE 11). The FIGURE2 demonstrates the outcomes of adults who are hospitalized for RSV. Adults older than 65 years have a 5% mortality rate if hospitalized for RSV infection.2
Vaccine options for adults
Two vaccines were recently approved for the prevention of RSV-associated lower respiratory tract disease (LRTD) in those ages 60 years and older: RSVPreF3 (Arexvy, GSK), which is an adjuvanted recombinant F protein vaccine, and RSVpreF (Abrysvo, Pfizer), which is a recombinant stabilized vaccine. Both require only a single dose (0.5 mL IM), which provides protection for 2 years.
The efficacy of the GSK vaccine in preventing laboratory-confirmed, RSV-associated LRTD was 82.6% during the first RSV season and 56.1% during the second season. The efficacy of the Pfizer vaccine in preventing symptomatic, laboratory-confirmed LRTD was 88.9% during the first RSV season and 78.6% during the second season.1 However, the trials leading to licensure of both vaccines were underpowered to show efficacy in the oldest adults and those who are frail or to show efficacy against RSV-caused hospitalization.
Safety of the adult RSV vaccines. The safety trials for both vaccines had a total of 38,177 participants. There were a total of 6 neurologic inflammatory conditions that developed within 42 days of vaccination, including 2 cases of suspected Guillain-Barré syndrome (GBS), 2 cases of possible acute disseminated encephalomyelitis, and 1 case each of chronic inflammatory demyelinating polyneuropathy and undifferentiated motor-sensory axonal polyneuropathy.1 That is a rate of 1 case of a neurologic inflammatory condition for every 6363 people vaccinated. Since the trials were not powered to determine whether the small number of cases were due to chance, postmarketing surveillance will be needed to clarify the true risk for GBS or other neurologic inflammatory events from RSV vaccination.
The lack of efficacy data for the most vulnerable older adults and the lingering questions about safety prompted the ACIP to recommend that adults ages 60 years and older may receive a single dose of RSV vaccine, using shared clinical decision-making—which is different from a routine or risk-based vaccine recommendation. For RSV vaccination, the decision to vaccinate should be based on a risk/benefit discussion between the clinician and the patient. Those most likely to benefit from the vaccine are listed in TABLE 1.1
While data on coadministration of RSV vaccines with other adult vaccines are sparse, the ACIP states that co-administration with other vaccines is acceptable.1 It is not known yet whether boosters will be needed after 2 years.
Continue to: RSV in infants and children
RSV in infants and children
RSV is the most common cause of hospitalization among infants and children in the United States. The CDC estimates that each year in children younger than 5 years, RSV is responsible for 1.5 million outpatient clinic visits, 520,000 emergency department visits, 58,000 to 80,000 hospitalizations, and 100 to 200 deaths.3 The risk for hospitalization from RSV is highest in the second and third months of life and decreases with increasing age.3
There are racial disparities in RSV severity: Intensive care unit admission rates are 1.2 to 1.6 times higher among non-Hispanic Black infants younger than 6 months than among non-Hispanic White infants, and hospitalization rates are up to 5 times higher in American Indian and Alaska Native populations.3
The months of highest RSV transmission in most locations are December through February, but this can vary. For practical purposes, RSV season runs from October through March.
Prevention in infants and children
The monoclonal antibody nirsevimab is now available for use in infants younger than 8 months born during or entering their first RSV season and children ages 8 to 19 months who are at increased risk for severe RSV disease and entering their second RSV season. Details regarding the use of this product were described in a recent Practice Alert Brief.4
Early studies on nirsevimab demonstrated 79% effectiveness in preventing medical-attended LRTD, 80.6% effectiveness in preventing hospitalization, and 90% effectiveness in preventing ICU admission. The number needed to immunize with nirsevimab to prevent an outpatient visit is estimated to be 17; to prevent an ED visit, 48; and to prevent an inpatient admission, 128. Due to the low RSV death rate, the studies were not able to demonstrate reduced mortality.5
Continue to: RSV vaccine in pregnancy
RSV vaccine in pregnancy
In August, the FDA approved Pfizer’s RSVpreF vaccine for use during pregnancy—as a single dose given at 32 to 36 weeks’ gestation—for the prevention of RSV LRTD in infants in the first 6 months of life. In the clinical trials, the vaccine was given at 24 to 36 weeks’ gestation. However, there was a statistically nonsignificant increase in preterm births in the RSVpreF group compared to the placebo group.6 While there were insufficient data to prove or rule out a causal relationship, the FDA advisory committee was more comfortable approving the vaccine for use only later in pregnancy, to avoid the possibility of very early preterm births after vaccination. The ACIP agreed.
From time of maternal vaccination, at least 14 days are needed to develop and transfer maternal antibodies across the placenta to protect the infant. Therefore, infants born less than 14 days after maternal vaccination should be considered unprotected.
Both maternal vaccination with RSVpreF and infant injection with nirsevimab are now options to protect newborns and infants from RSV. However, use of both products is not needed, since combined they do not offer significant added protection compared to either product alone (exceptions to be discussed shortly).6 When the estimated due date will occur in the RSV season, maternity clinicians should provide information on both products and assist the mother in deciding whether to be vaccinated or rely on administration of nirsevimab to the infant after birth. The benefits and risks of these 2 options are listed in TABLE 2.6
There are some rare situations in which use of both products is recommended, and they include6:
- When the baby is born less than 14 days from the time of maternal vaccination
- When the mother has a condition that could produce an inadequate response to the vaccine
- When the infant has had cardiopulmonary bypass, which would lead to loss of maternal antibodies
- When the infant has severe disease placing them at increased risk for severe RSV.
Conclusion
All of these new RSV preventive products should soon be widely available and covered with no out-of-pocket expense by commercial and government payers. The exception might be nirsevimab—because of the time needed to produce it, it might not be universally available in the 2023-2024 season.
1. Melgar M, Britton A, Roper LE, et al. Use of respiratory syncytial virus vaccine in older adults: recommendation of the Advisory Committee on Immunization Practices—United States, 2023. MMWR Morb Mortal Wkly Rep. 2023;72:793-801.
2. Melgar M. Evidence to recommendation framework. RSV in adults. Presented to the ACIP on February 23, 2023. Accessed November 7, 2023. www.cdc.gov/vaccines/acip/meetings/downloads/slides-2023-02/slides-02-23/RSV-Adults-04-Melgar-508.pdf
3. Jones JM, Fleming-Dutra KE, Prill MM, et al. Use of nirsevimab for the prevention of respiratory syncytial virus disease among infants and young children: recommendation of the Advisory Committee on Immunization Practices—United States, 2023. MMWR Morb Mortal Wkly Rep. 2023;72:90-925.
4. Campos-Outcalt D. Are you ready for RSV season? There’s a new preventive option. J Fam Pract. 2023;72. doi: 10.12788/jfp.0663
5. Jones J. Evidence to recommendation framework: nirsevimab updates. Presented to the ACIP on August 3, 2023. Accessed August 23, 2023. https://stacks.cdc.gov/view/cdc/131586
6. Jones J. Clinical considerations for maternal RSVPreF vaccine and nirsevimab. Presented to the ACIP on September 25, 2023. Accessed November 8, 2023. www2.cdc.gov/vaccines/ed/ciinc/archives/23/09/ciiw_RSV2/CIIW%20RSV%20maternal%20vaccine%20mAb%209.27.23.pdf
1. Melgar M, Britton A, Roper LE, et al. Use of respiratory syncytial virus vaccine in older adults: recommendation of the Advisory Committee on Immunization Practices—United States, 2023. MMWR Morb Mortal Wkly Rep. 2023;72:793-801.
2. Melgar M. Evidence to recommendation framework. RSV in adults. Presented to the ACIP on February 23, 2023. Accessed November 7, 2023. www.cdc.gov/vaccines/acip/meetings/downloads/slides-2023-02/slides-02-23/RSV-Adults-04-Melgar-508.pdf
3. Jones JM, Fleming-Dutra KE, Prill MM, et al. Use of nirsevimab for the prevention of respiratory syncytial virus disease among infants and young children: recommendation of the Advisory Committee on Immunization Practices—United States, 2023. MMWR Morb Mortal Wkly Rep. 2023;72:90-925.
4. Campos-Outcalt D. Are you ready for RSV season? There’s a new preventive option. J Fam Pract. 2023;72. doi: 10.12788/jfp.0663
5. Jones J. Evidence to recommendation framework: nirsevimab updates. Presented to the ACIP on August 3, 2023. Accessed August 23, 2023. https://stacks.cdc.gov/view/cdc/131586
6. Jones J. Clinical considerations for maternal RSVPreF vaccine and nirsevimab. Presented to the ACIP on September 25, 2023. Accessed November 8, 2023. www2.cdc.gov/vaccines/ed/ciinc/archives/23/09/ciiw_RSV2/CIIW%20RSV%20maternal%20vaccine%20mAb%209.27.23.pdf
Is low-molecular-weight heparin superior to aspirin for VTE prophylaxis?
ILLUSTRATIVE CASE
A 72-year-old man with well-controlled hypertension and chronic obstructive pulmonary disease is scheduled for right total hip arthroplasty (THA) due to severe arthritis. He will be admitted to the hospital overnight, and his orthopedic surgeon anticipates 2 to 3 days of inpatient recovery time. In addition to medical management of the patient’s comorbid conditions, the surgeon asks if you have any insight regarding VTE prophylaxis for this patient. Specifically, do you think aspirin is equal to LMWH for VTE prophylaxis?
All adults undergoing major orthopedic surgery are considered to be at high risk for postoperative VTE development, with those having lower-limb procedures at highest risk.2 Of the more than 2.2 million THAs and total knee arthroplasties (TKAs) performed in the United States between 2012 and 2020, 55% were primary TKAs and 39% primary THAs.3 The American College of Chest Physicians (ACCP) estimated a baseline 35-day risk for VTE of 4.3% in patients undergoing major orthopedic surgery.4 The highest VTE risk occurs during the first 7 to 14 days post surgery (1.8% for symptomatic deep vein thrombosis [DVT] and 1% for pulmonary embolism [PE]), with a slightly lower risk during the subsequent 15 to 35 days (1% for symptomatic DVT and 0.5% for PE).4
Aspirin’s low cost, availability, and ease of administration make it an attractive choice for VTE prevention in patients post THA and TKA surgery. The Pulmonary Embolism Prevention (PEP) trial evaluated 13,356 patients undergoing hip fracture repair and 4088 patients undergoing arthroplasty and found aspirin to be safe and effective in prevention of VTEs compared with placebo. The investigators concluded that “there is now good evidence for considering aspirin routinely in a wide range of surgical and medical groups at high risk of venous thromboembolism.”5 The PEP study, along with others, led to the emergence of aspirin monotherapy for VTE prophylaxis.
Current guidelines for perioperative VTE prophylaxis are based on American Society of Hematology (ASH) and ACCP recommendations. For patients undergoing THA or TKA, ASH suggests using aspirin or anticoagulants for VTE prophylaxis; when anticoagulants are used, they suggest using a direct oral anticoagulant (DOAC) over LMWH.6 The ASH guidelines are conditional recommendations based on very low certainty of effects, and the ASH panel recognized the need for further investigation with large, high-quality clinical trials.
The ACCP guidelines are clearer in recommending VTE prophylaxis vs no prophylaxis for major orthopedic surgeries and recommend the use of LMWH over other agents, including aspirin, DOACs, warfarin, and intermittent pneumatic compression (IPC) devices.4
Although prophylaxis is widely recommended to mitigate the elevated risk for VTE among patients undergoing orthopedic surgery, aspirin as monotherapy remains controversial.7 Many orthopedic surgeons prescribe aspirin as a sole VTE prophylaxis agent; however, this practice is not well supported by data from large, well-conducted, randomized trials or inferiority trials.2
STUDY SUMMARY
Aspirin did not meet the noninferiority criterion for postoperative VTE
The CRISTAL trial compared the use of aspirin vs LMWH (enoxaparin) for VTE prophylaxis in patients ages 18 years or older undergoing primary THA or TKA for osteoarthritis.1 This Australian study used a cluster-randomized, crossover, registry-nested, noninferiority trial design. Of note, in Australia, aspirin is formulated in 100-mg tablets, equivalent to the standard 81-mg low-dose tablet in the United States.
Continue to: Patients taking prescribed antiplatelet...
Patients taking prescribed antiplatelet medication for preexisting conditions (~20% of patients in each group) were allowed to continue antiplatelet therapy during the trial. Patients were excluded if they were receiving an anticoagulant prior to their procedure or had a medical contraindication to aspirin or enoxaparin.
Thirty-one hospital sites were randomly assigned a treatment protocol using either aspirin or enoxaparin. Once target patient enrollment was met with the initial assigned medication, the site switched to the second/other agent. This resulted in 5675 patients in the aspirin group and 4036 in the enoxaparin group enrolled between April 2019 and December 2020, with final follow-up in August 2021; of these, 259 in the aspirin group and 249 in the enoxaparin group were lost to follow-up, opted out, or died.
The aspirin group was given 100 mg PO daily and the enoxaparin group was given 40 mg SC daily (20 mg daily for patients weighing < 50 kg or with an estimated glomerular filtration rate < 30 mL/min/1.73 m2) for 35 days after THA and 14 days after TKA. Both treatment groups received IPC calf devices intraoperatively and postoperatively, and mobilization was offered on postoperative Day 0 or 1.
The primary outcome—development of symptomatic VTE within 90 days of the procedure—occurred in 187 (3.5%) patients in the aspirin group and 69 (1.8%) patients in the enoxaparin group (estimated difference = 1.97%; 95% CI, 0.54%-3.41%). This did not meet the noninferiority criterion for aspirin, based on an estimated assumed rate of 2% and a noninferiority margin of 1%, and in fact was statistically superior for enoxaparin (P = .007). There were no significant differences between the 2 groups in major bleeding or death within 90 days.1
WHAT’S NEW
Enoxaparin was significantly superior to aspirin for VTE prophylaxis
Although this study was designed as a noninferiority trial, analysis showed enoxaparin to be significantly superior for postoperative VTE prophylaxis compared with aspirin.
Continue to: CAVEATS
CAVEATS
Study aspirin dosing differed from US standard
This study showed significantly lower rates of symptomatic VTE in the enoxaparin group compared with the aspirin group; however, the majority of this difference was driven by rates of below-the-knee DVTs, which are clinically less relevant.8 Also, this trial used a 100-mg aspirin formulation, which is not available in the United States.
CHALLENGES TO IMPLEMENTATION
Aspirin is far cheaper and administered orally
Aspirin is significantly cheaper than enoxaparin, costing about $0.13 per dose (~$4 for 30 tablets at the 81-mg dose) vs roughly $9 per 40 mg/0.4 mL dose for enoxaparin.9 However, a cost-effectiveness analysis may be useful to determine (for example) whether the higher cost of enoxaparin may be offset by fewer DVTs and other sequelae. Lastly, LMWH is an injection, which some patients may refuse.
1. CRISTAL Study Group; Sidhu VS, Kelly TL, Pratt N, et al. Effect of aspirin vs enoxaparin on symptomatic venous thromboembolism in patients undergoing hip or knee arthroplasty: the CRISTAL randomized trial. JAMA. 2022;328:719-727. doi: 10.1001/jama.2022.13416
2. Douketis JD, Mithoowani S. Prevention of venous thromboembolism in adults undergoing hip fracture repair or hip or knee replacement. UpToDate. Updated January 25, 2023. Accessed May 24, 2023. www.uptodate.com/contents/prevention-of-venous-thromboembolism-in-adults-undergoing-hip-fracture-repair-or-hip-or-knee-replacement
3. Siddiqi A, Levine BR, Springer BD. Highlights of the 2021 American Joint Replacement Registry annual report. Arthroplast Today. 2022;13:205-207. doi: 10.1016/j.artd.2022.01.020
4. Falck-Ytter Y, Francis CW, Johanson NA, et al. Prevention of VTE in orthopedic surgery patients: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 2012;141(2 suppl):e278S-e325S. doi: 10.1378/chest.11-2404
5. Pulmonary Embolism Prevention (PEP) trial Collaborative Group. Prevention of pulmonary embolism and deep vein thrombosis with low dose aspirin: Pulmonary Embolism Prevention (PEP) trial. Lancet. 2000;355:1295-1302. doi: 10.1016/S0140-6736(00)02110-3
6. Anderson DR, Morgano GP, Bennett C, et al. American Society of Hematology 2019 guidelines for management of venous thromboembolism: prevention of venous thromboembolism in surgical hospitalized patients. Blood Adv. 2019;3:3898-3944. doi: 10.1182/bloodadvances.2019000975
7. Matharu GS, Kunutsor SK, Judge A, et al. Clinical effectiveness and safety of aspirin for venous thromboembolism prophylaxis after total hip and knee replacement: a systematic review and meta-analysis of randomized clinical trials. JAMA Intern Med. 2020;180:376-384. doi: 10.1001/jamainternmed.2019.6108
8. Brett AS, Friedman RJ. Aspirin vs. enoxaparin for prophylaxis after hip or knee replacement. NEJM Journal Watch. September 15, 2022. Accessed May 24, 2023. www.jwatch.org/na55272/2022/09/15/aspirin-vs-enoxaparin-prophylaxis-after-hip-or-knee
9. Enoxaparin. GoodRx. Accessed August 7, 2023. www.goodrx.com/enoxaparin
ILLUSTRATIVE CASE
A 72-year-old man with well-controlled hypertension and chronic obstructive pulmonary disease is scheduled for right total hip arthroplasty (THA) due to severe arthritis. He will be admitted to the hospital overnight, and his orthopedic surgeon anticipates 2 to 3 days of inpatient recovery time. In addition to medical management of the patient’s comorbid conditions, the surgeon asks if you have any insight regarding VTE prophylaxis for this patient. Specifically, do you think aspirin is equal to LMWH for VTE prophylaxis?
All adults undergoing major orthopedic surgery are considered to be at high risk for postoperative VTE development, with those having lower-limb procedures at highest risk.2 Of the more than 2.2 million THAs and total knee arthroplasties (TKAs) performed in the United States between 2012 and 2020, 55% were primary TKAs and 39% primary THAs.3 The American College of Chest Physicians (ACCP) estimated a baseline 35-day risk for VTE of 4.3% in patients undergoing major orthopedic surgery.4 The highest VTE risk occurs during the first 7 to 14 days post surgery (1.8% for symptomatic deep vein thrombosis [DVT] and 1% for pulmonary embolism [PE]), with a slightly lower risk during the subsequent 15 to 35 days (1% for symptomatic DVT and 0.5% for PE).4
Aspirin’s low cost, availability, and ease of administration make it an attractive choice for VTE prevention in patients post THA and TKA surgery. The Pulmonary Embolism Prevention (PEP) trial evaluated 13,356 patients undergoing hip fracture repair and 4088 patients undergoing arthroplasty and found aspirin to be safe and effective in prevention of VTEs compared with placebo. The investigators concluded that “there is now good evidence for considering aspirin routinely in a wide range of surgical and medical groups at high risk of venous thromboembolism.”5 The PEP study, along with others, led to the emergence of aspirin monotherapy for VTE prophylaxis.
Current guidelines for perioperative VTE prophylaxis are based on American Society of Hematology (ASH) and ACCP recommendations. For patients undergoing THA or TKA, ASH suggests using aspirin or anticoagulants for VTE prophylaxis; when anticoagulants are used, they suggest using a direct oral anticoagulant (DOAC) over LMWH.6 The ASH guidelines are conditional recommendations based on very low certainty of effects, and the ASH panel recognized the need for further investigation with large, high-quality clinical trials.
The ACCP guidelines are clearer in recommending VTE prophylaxis vs no prophylaxis for major orthopedic surgeries and recommend the use of LMWH over other agents, including aspirin, DOACs, warfarin, and intermittent pneumatic compression (IPC) devices.4
Although prophylaxis is widely recommended to mitigate the elevated risk for VTE among patients undergoing orthopedic surgery, aspirin as monotherapy remains controversial.7 Many orthopedic surgeons prescribe aspirin as a sole VTE prophylaxis agent; however, this practice is not well supported by data from large, well-conducted, randomized trials or inferiority trials.2
STUDY SUMMARY
Aspirin did not meet the noninferiority criterion for postoperative VTE
The CRISTAL trial compared the use of aspirin vs LMWH (enoxaparin) for VTE prophylaxis in patients ages 18 years or older undergoing primary THA or TKA for osteoarthritis.1 This Australian study used a cluster-randomized, crossover, registry-nested, noninferiority trial design. Of note, in Australia, aspirin is formulated in 100-mg tablets, equivalent to the standard 81-mg low-dose tablet in the United States.
Continue to: Patients taking prescribed antiplatelet...
Patients taking prescribed antiplatelet medication for preexisting conditions (~20% of patients in each group) were allowed to continue antiplatelet therapy during the trial. Patients were excluded if they were receiving an anticoagulant prior to their procedure or had a medical contraindication to aspirin or enoxaparin.
Thirty-one hospital sites were randomly assigned a treatment protocol using either aspirin or enoxaparin. Once target patient enrollment was met with the initial assigned medication, the site switched to the second/other agent. This resulted in 5675 patients in the aspirin group and 4036 in the enoxaparin group enrolled between April 2019 and December 2020, with final follow-up in August 2021; of these, 259 in the aspirin group and 249 in the enoxaparin group were lost to follow-up, opted out, or died.
The aspirin group was given 100 mg PO daily and the enoxaparin group was given 40 mg SC daily (20 mg daily for patients weighing < 50 kg or with an estimated glomerular filtration rate < 30 mL/min/1.73 m2) for 35 days after THA and 14 days after TKA. Both treatment groups received IPC calf devices intraoperatively and postoperatively, and mobilization was offered on postoperative Day 0 or 1.
The primary outcome—development of symptomatic VTE within 90 days of the procedure—occurred in 187 (3.5%) patients in the aspirin group and 69 (1.8%) patients in the enoxaparin group (estimated difference = 1.97%; 95% CI, 0.54%-3.41%). This did not meet the noninferiority criterion for aspirin, based on an estimated assumed rate of 2% and a noninferiority margin of 1%, and in fact was statistically superior for enoxaparin (P = .007). There were no significant differences between the 2 groups in major bleeding or death within 90 days.1
WHAT’S NEW
Enoxaparin was significantly superior to aspirin for VTE prophylaxis
Although this study was designed as a noninferiority trial, analysis showed enoxaparin to be significantly superior for postoperative VTE prophylaxis compared with aspirin.
Continue to: CAVEATS
CAVEATS
Study aspirin dosing differed from US standard
This study showed significantly lower rates of symptomatic VTE in the enoxaparin group compared with the aspirin group; however, the majority of this difference was driven by rates of below-the-knee DVTs, which are clinically less relevant.8 Also, this trial used a 100-mg aspirin formulation, which is not available in the United States.
CHALLENGES TO IMPLEMENTATION
Aspirin is far cheaper and administered orally
Aspirin is significantly cheaper than enoxaparin, costing about $0.13 per dose (~$4 for 30 tablets at the 81-mg dose) vs roughly $9 per 40 mg/0.4 mL dose for enoxaparin.9 However, a cost-effectiveness analysis may be useful to determine (for example) whether the higher cost of enoxaparin may be offset by fewer DVTs and other sequelae. Lastly, LMWH is an injection, which some patients may refuse.
ILLUSTRATIVE CASE
A 72-year-old man with well-controlled hypertension and chronic obstructive pulmonary disease is scheduled for right total hip arthroplasty (THA) due to severe arthritis. He will be admitted to the hospital overnight, and his orthopedic surgeon anticipates 2 to 3 days of inpatient recovery time. In addition to medical management of the patient’s comorbid conditions, the surgeon asks if you have any insight regarding VTE prophylaxis for this patient. Specifically, do you think aspirin is equal to LMWH for VTE prophylaxis?
All adults undergoing major orthopedic surgery are considered to be at high risk for postoperative VTE development, with those having lower-limb procedures at highest risk.2 Of the more than 2.2 million THAs and total knee arthroplasties (TKAs) performed in the United States between 2012 and 2020, 55% were primary TKAs and 39% primary THAs.3 The American College of Chest Physicians (ACCP) estimated a baseline 35-day risk for VTE of 4.3% in patients undergoing major orthopedic surgery.4 The highest VTE risk occurs during the first 7 to 14 days post surgery (1.8% for symptomatic deep vein thrombosis [DVT] and 1% for pulmonary embolism [PE]), with a slightly lower risk during the subsequent 15 to 35 days (1% for symptomatic DVT and 0.5% for PE).4
Aspirin’s low cost, availability, and ease of administration make it an attractive choice for VTE prevention in patients post THA and TKA surgery. The Pulmonary Embolism Prevention (PEP) trial evaluated 13,356 patients undergoing hip fracture repair and 4088 patients undergoing arthroplasty and found aspirin to be safe and effective in prevention of VTEs compared with placebo. The investigators concluded that “there is now good evidence for considering aspirin routinely in a wide range of surgical and medical groups at high risk of venous thromboembolism.”5 The PEP study, along with others, led to the emergence of aspirin monotherapy for VTE prophylaxis.
Current guidelines for perioperative VTE prophylaxis are based on American Society of Hematology (ASH) and ACCP recommendations. For patients undergoing THA or TKA, ASH suggests using aspirin or anticoagulants for VTE prophylaxis; when anticoagulants are used, they suggest using a direct oral anticoagulant (DOAC) over LMWH.6 The ASH guidelines are conditional recommendations based on very low certainty of effects, and the ASH panel recognized the need for further investigation with large, high-quality clinical trials.
The ACCP guidelines are clearer in recommending VTE prophylaxis vs no prophylaxis for major orthopedic surgeries and recommend the use of LMWH over other agents, including aspirin, DOACs, warfarin, and intermittent pneumatic compression (IPC) devices.4
Although prophylaxis is widely recommended to mitigate the elevated risk for VTE among patients undergoing orthopedic surgery, aspirin as monotherapy remains controversial.7 Many orthopedic surgeons prescribe aspirin as a sole VTE prophylaxis agent; however, this practice is not well supported by data from large, well-conducted, randomized trials or inferiority trials.2
STUDY SUMMARY
Aspirin did not meet the noninferiority criterion for postoperative VTE
The CRISTAL trial compared the use of aspirin vs LMWH (enoxaparin) for VTE prophylaxis in patients ages 18 years or older undergoing primary THA or TKA for osteoarthritis.1 This Australian study used a cluster-randomized, crossover, registry-nested, noninferiority trial design. Of note, in Australia, aspirin is formulated in 100-mg tablets, equivalent to the standard 81-mg low-dose tablet in the United States.
Continue to: Patients taking prescribed antiplatelet...
Patients taking prescribed antiplatelet medication for preexisting conditions (~20% of patients in each group) were allowed to continue antiplatelet therapy during the trial. Patients were excluded if they were receiving an anticoagulant prior to their procedure or had a medical contraindication to aspirin or enoxaparin.
Thirty-one hospital sites were randomly assigned a treatment protocol using either aspirin or enoxaparin. Once target patient enrollment was met with the initial assigned medication, the site switched to the second/other agent. This resulted in 5675 patients in the aspirin group and 4036 in the enoxaparin group enrolled between April 2019 and December 2020, with final follow-up in August 2021; of these, 259 in the aspirin group and 249 in the enoxaparin group were lost to follow-up, opted out, or died.
The aspirin group was given 100 mg PO daily and the enoxaparin group was given 40 mg SC daily (20 mg daily for patients weighing < 50 kg or with an estimated glomerular filtration rate < 30 mL/min/1.73 m2) for 35 days after THA and 14 days after TKA. Both treatment groups received IPC calf devices intraoperatively and postoperatively, and mobilization was offered on postoperative Day 0 or 1.
The primary outcome—development of symptomatic VTE within 90 days of the procedure—occurred in 187 (3.5%) patients in the aspirin group and 69 (1.8%) patients in the enoxaparin group (estimated difference = 1.97%; 95% CI, 0.54%-3.41%). This did not meet the noninferiority criterion for aspirin, based on an estimated assumed rate of 2% and a noninferiority margin of 1%, and in fact was statistically superior for enoxaparin (P = .007). There were no significant differences between the 2 groups in major bleeding or death within 90 days.1
WHAT’S NEW
Enoxaparin was significantly superior to aspirin for VTE prophylaxis
Although this study was designed as a noninferiority trial, analysis showed enoxaparin to be significantly superior for postoperative VTE prophylaxis compared with aspirin.
Continue to: CAVEATS
CAVEATS
Study aspirin dosing differed from US standard
This study showed significantly lower rates of symptomatic VTE in the enoxaparin group compared with the aspirin group; however, the majority of this difference was driven by rates of below-the-knee DVTs, which are clinically less relevant.8 Also, this trial used a 100-mg aspirin formulation, which is not available in the United States.
CHALLENGES TO IMPLEMENTATION
Aspirin is far cheaper and administered orally
Aspirin is significantly cheaper than enoxaparin, costing about $0.13 per dose (~$4 for 30 tablets at the 81-mg dose) vs roughly $9 per 40 mg/0.4 mL dose for enoxaparin.9 However, a cost-effectiveness analysis may be useful to determine (for example) whether the higher cost of enoxaparin may be offset by fewer DVTs and other sequelae. Lastly, LMWH is an injection, which some patients may refuse.
1. CRISTAL Study Group; Sidhu VS, Kelly TL, Pratt N, et al. Effect of aspirin vs enoxaparin on symptomatic venous thromboembolism in patients undergoing hip or knee arthroplasty: the CRISTAL randomized trial. JAMA. 2022;328:719-727. doi: 10.1001/jama.2022.13416
2. Douketis JD, Mithoowani S. Prevention of venous thromboembolism in adults undergoing hip fracture repair or hip or knee replacement. UpToDate. Updated January 25, 2023. Accessed May 24, 2023. www.uptodate.com/contents/prevention-of-venous-thromboembolism-in-adults-undergoing-hip-fracture-repair-or-hip-or-knee-replacement
3. Siddiqi A, Levine BR, Springer BD. Highlights of the 2021 American Joint Replacement Registry annual report. Arthroplast Today. 2022;13:205-207. doi: 10.1016/j.artd.2022.01.020
4. Falck-Ytter Y, Francis CW, Johanson NA, et al. Prevention of VTE in orthopedic surgery patients: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 2012;141(2 suppl):e278S-e325S. doi: 10.1378/chest.11-2404
5. Pulmonary Embolism Prevention (PEP) trial Collaborative Group. Prevention of pulmonary embolism and deep vein thrombosis with low dose aspirin: Pulmonary Embolism Prevention (PEP) trial. Lancet. 2000;355:1295-1302. doi: 10.1016/S0140-6736(00)02110-3
6. Anderson DR, Morgano GP, Bennett C, et al. American Society of Hematology 2019 guidelines for management of venous thromboembolism: prevention of venous thromboembolism in surgical hospitalized patients. Blood Adv. 2019;3:3898-3944. doi: 10.1182/bloodadvances.2019000975
7. Matharu GS, Kunutsor SK, Judge A, et al. Clinical effectiveness and safety of aspirin for venous thromboembolism prophylaxis after total hip and knee replacement: a systematic review and meta-analysis of randomized clinical trials. JAMA Intern Med. 2020;180:376-384. doi: 10.1001/jamainternmed.2019.6108
8. Brett AS, Friedman RJ. Aspirin vs. enoxaparin for prophylaxis after hip or knee replacement. NEJM Journal Watch. September 15, 2022. Accessed May 24, 2023. www.jwatch.org/na55272/2022/09/15/aspirin-vs-enoxaparin-prophylaxis-after-hip-or-knee
9. Enoxaparin. GoodRx. Accessed August 7, 2023. www.goodrx.com/enoxaparin
1. CRISTAL Study Group; Sidhu VS, Kelly TL, Pratt N, et al. Effect of aspirin vs enoxaparin on symptomatic venous thromboembolism in patients undergoing hip or knee arthroplasty: the CRISTAL randomized trial. JAMA. 2022;328:719-727. doi: 10.1001/jama.2022.13416
2. Douketis JD, Mithoowani S. Prevention of venous thromboembolism in adults undergoing hip fracture repair or hip or knee replacement. UpToDate. Updated January 25, 2023. Accessed May 24, 2023. www.uptodate.com/contents/prevention-of-venous-thromboembolism-in-adults-undergoing-hip-fracture-repair-or-hip-or-knee-replacement
3. Siddiqi A, Levine BR, Springer BD. Highlights of the 2021 American Joint Replacement Registry annual report. Arthroplast Today. 2022;13:205-207. doi: 10.1016/j.artd.2022.01.020
4. Falck-Ytter Y, Francis CW, Johanson NA, et al. Prevention of VTE in orthopedic surgery patients: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 2012;141(2 suppl):e278S-e325S. doi: 10.1378/chest.11-2404
5. Pulmonary Embolism Prevention (PEP) trial Collaborative Group. Prevention of pulmonary embolism and deep vein thrombosis with low dose aspirin: Pulmonary Embolism Prevention (PEP) trial. Lancet. 2000;355:1295-1302. doi: 10.1016/S0140-6736(00)02110-3
6. Anderson DR, Morgano GP, Bennett C, et al. American Society of Hematology 2019 guidelines for management of venous thromboembolism: prevention of venous thromboembolism in surgical hospitalized patients. Blood Adv. 2019;3:3898-3944. doi: 10.1182/bloodadvances.2019000975
7. Matharu GS, Kunutsor SK, Judge A, et al. Clinical effectiveness and safety of aspirin for venous thromboembolism prophylaxis after total hip and knee replacement: a systematic review and meta-analysis of randomized clinical trials. JAMA Intern Med. 2020;180:376-384. doi: 10.1001/jamainternmed.2019.6108
8. Brett AS, Friedman RJ. Aspirin vs. enoxaparin for prophylaxis after hip or knee replacement. NEJM Journal Watch. September 15, 2022. Accessed May 24, 2023. www.jwatch.org/na55272/2022/09/15/aspirin-vs-enoxaparin-prophylaxis-after-hip-or-knee
9. Enoxaparin. GoodRx. Accessed August 7, 2023. www.goodrx.com/enoxaparin
PRACTICE CHANGER
Consider low-molecular-weight heparin (LMWH) rather than aspirin to prevent postoperative venous thromboembolism (VTE) in patients undergoing total hip or knee arthroplasty for osteoarthritis.
STRENGTH OF RECOMMENDATION
B: Based on a single cluster-randomized crossover trial.1
Does taking BP medicine at night (vs morning) result in fewer cardiovascular events?
Evidence summary
Recent UK study shows no difference by timing
A 2022 UK prospective, randomized, multicenter trial assigned 21,104 predominantly White adults (58% men) with hypertension to take their usual antihypertensive medication either in the morning (6
All patient baseline characteristics were equivalent between groups. If troubled by nocturia, patients in the evening group taking diuretics were told to take only the diuretic earlier (6
The median follow-up was 5.2 years. Data were collected at regular intervals through patient completion of online questionnaires and researcher analysis of National Health Service data on hospitalization and death. The intention-to-treat analysis showed no difference in the primary outcome (a composite of vascular death, nonfatal myocardial infarction, or nonfatal stroke) between the evening and morning administration groups (0.69 events vs 0.72 events per 100 person-years; hazard ratio [HR] = 0.95; 95% CI, 0.83-1.10; P = .53).
The controversial Hygia Project favored evening
Prior to the UK study was the Hygia Chronotherapy Trial, a prospective, controlled, multicenter study conducted within the primary care setting in Spain. Caucasian Spanish adults (N = 19,168; mean age, 61 years; 56% men) with hypertension were randomly assigned to take all prescribed antihypertensive medication either at bedtime or upon waking.2
The Hygia Project initially sought to establish the value of ambulatory blood pressure monitoring (ABPM) compared to office blood pressure (BP) monitoring and to explore the prognostic value of sleeping BP.3 The study objectives evolved over time. The randomization process was not clearly described,2,3 but multiple randomizations were alluded to. The authors stated that “for any of these chronotherapy trials” randomizations were done separately for “each participating center” and “randomization of participants to treatment-time regimen is done separately for each hypertension medication or combination being tested.”
The baseline characteristics of patients in the evening and morning administration groups were similar, but statistically significant differences existed in BMI (29.6 vs 29.7; P = .030) and sleep-time systolic BP percent decline (9.3 vs 9.0; P < .001). Mean baseline 48-hour BP was 132/77 mm Hg. Hypertension was defined as an awake systolic BP ≥
Prescribers were free to prescribe medicines from 5 classes (diuretic, angiotensin-converting enzyme inhibitor, angiotensin receptor blocker, calcium channel blocker, or beta-blocker) as they thought appropriate, were encouraged to use fixed-dose combination pills, and were told not to use split (eg, twice per day) dosing. Annual 48-hour ABPM was completed, and patients’ electronic health records were analyzed by blinded investigators. Median follow-up was 6.3 years, and only 84 participants failed to complete the minimum 1-year participation requirement.
Continue to: The primary outcome...
The primary outcome—a composite of cardiovascular death, myocardial infarction, coronary revascularization, heart failure, or stroke—occurred in 1752 patients, favoring the bedtime group (HR = 0.55; 95% CI, 0.50-0.61; P < .001). The calculated number of events was 1130 in the morning administration group and 622 in the evening administration group; the authors did not explicitly report the event numbers in each group. Each component of the composite outcome also favored evening administration (P < .001 for all): cardiovascular death (HR = 0.44; 95% CI, 0.34-0.56), myocardial infarction (HR = 0.66; 95% CI, 0.52-0.84), coronary revascularization (HR = 0.60; 95% CI, 0.47-0.75), heart failure (HR = 0.58; 95% CI, 0.49-0.70), and stroke (HR = 0.51; 95% CI, 0.41-0.63).
The complicated, layered study design and randomization methods limit the ability to critically appraise the study.
Smaller Spanish study also supported evening administration
A prior, smaller, prospective randomized trial conducted by the same researchers as the Hygia Project found even greater benefits to evening BP medication administration.4 The 2156 Spanish patients (52% men; average age, 55 years) from multiple primary care offices were randomized 1:1 to BP medication administration either upon awakening or at bedtime. Dozens of baseline characteristics were evenly distributed except for age (55.0 vs 56.3; P = .021) and creatinine (0.96 vs 0.98; P = .028), both of which were lower in the evening group.
After a median follow-up of 5.6 years, the bedtime group had significantly lower total events (187 events in the morning group vs 68 in the evening group; relative risk [RR] = 0.39; 95% CI, 0.29-0.51; P < .001). Individual cardiovascular outcomes also dramatically favored the evening group: total deaths (12 vs 28; P = .008), cardiovascular deaths (3 vs 14; P = .006), cardiovascular disease events (30 vs 74; P < .001), stroke (7 vs 24; P = .001), and heart failure (8 vs 33; P < .001).
Limits of both the UK trial and the Hygia Project trial included single countries of study with a lack of racial and ethnic diversity, and greater nonadherence to the evening administration of the medications.
Recommendations from others
A 2022 consensus statement from the International Society of Hypertension, published before the UK trial, recommended against bedtime dosing until more high-quality data became available. They pointed to evidence showing higher medication adherence with morning dosing, risk for asleep BP dropping, and worsening daytime BP control as reasons to continue morning administration.5 Other reviewers have questioned the Hygia Project results due to their reported implausibly large effects on cardiovascular outcomes, noting that independent attempts to verify the methods and the data have proven challenging and are not completed.6
Editor’s takeaway
I confess that I was swayed by the results of the Hygia Project; for a year or so, I advised my patients to take at least 1 BP pill at night. But after the UK study came out, I needed to reconsider. I began to worry that the great outcomes of nocturnal therapy may have been a mirage. I have returned to counseling patients to take their BP medications in whichever way fosters consistency while minimizing adverse effects for them.
1. Mackenzie IS, Rogers A, Poulter NR, et al; TIME Study Group. Cardiovascular outcomes in adults with hypertension with evening versus morning dosing of usual antihypertensives in the UK (TIME study): a prospective, randomised, open-label, blinded-endpoint clinical trial. Lancet. 2022;400:1417-1425. doi: 10.1016/S0140-6736(22)01786-X
2. Hermida RC, Crespo JJ, Domínguez-Sardiña M, et al; Hygia Project Investigators. Bedtime hypertension treatment improves cardiovascular risk reduction: the Hygia Chronotherapy Trial. Eur Heart J. 2020;41:4565-4576. doi: 10.1093/eurheartj/ehz754
3. Hermida RC. Sleep-time ambulatory blood pressure as a prognostic marker of vascular and other risks and therapeutic target for prevention by hypertension chronotherapy: rationale and design of the Hygia Project. Chronobiol Int. 2016;33:906-936. doi: 10.1080/07420528.2016.1181078
4. Hermida RC, Ayala DE, Mojón A, et al. Influence of circadian time of hypertension treatment on cardiovascular risk: results of the MAPEC study. Chronobiol Int. 2010;27:1629-1651. doi: 10.3109/07420528.2010.510230
5. Stergiou G, Brunström M, MacDonald T, et al. Bedtime dosing of antihypertensive medications: systematic review and consensus statement: International Society of Hypertension position paper endorsed by World Hypertension League and European Society of Hypertension. J Hypertens. 2022;40:1847-1858. doi: 10.1097/HJH.0000000000003240
6. Brunström M, Kjeldsen SE, Kreutz R, et al. Missing verification of source data in hypertension research: The HYGIA PROJECT in Perspective. Hypertension. 2021;78:555-558. doi: 10.1161/HYPERTENSIONAHA.121.17356
Evidence summary
Recent UK study shows no difference by timing
A 2022 UK prospective, randomized, multicenter trial assigned 21,104 predominantly White adults (58% men) with hypertension to take their usual antihypertensive medication either in the morning (6
All patient baseline characteristics were equivalent between groups. If troubled by nocturia, patients in the evening group taking diuretics were told to take only the diuretic earlier (6
The median follow-up was 5.2 years. Data were collected at regular intervals through patient completion of online questionnaires and researcher analysis of National Health Service data on hospitalization and death. The intention-to-treat analysis showed no difference in the primary outcome (a composite of vascular death, nonfatal myocardial infarction, or nonfatal stroke) between the evening and morning administration groups (0.69 events vs 0.72 events per 100 person-years; hazard ratio [HR] = 0.95; 95% CI, 0.83-1.10; P = .53).
The controversial Hygia Project favored evening
Prior to the UK study was the Hygia Chronotherapy Trial, a prospective, controlled, multicenter study conducted within the primary care setting in Spain. Caucasian Spanish adults (N = 19,168; mean age, 61 years; 56% men) with hypertension were randomly assigned to take all prescribed antihypertensive medication either at bedtime or upon waking.2
The Hygia Project initially sought to establish the value of ambulatory blood pressure monitoring (ABPM) compared to office blood pressure (BP) monitoring and to explore the prognostic value of sleeping BP.3 The study objectives evolved over time. The randomization process was not clearly described,2,3 but multiple randomizations were alluded to. The authors stated that “for any of these chronotherapy trials” randomizations were done separately for “each participating center” and “randomization of participants to treatment-time regimen is done separately for each hypertension medication or combination being tested.”
The baseline characteristics of patients in the evening and morning administration groups were similar, but statistically significant differences existed in BMI (29.6 vs 29.7; P = .030) and sleep-time systolic BP percent decline (9.3 vs 9.0; P < .001). Mean baseline 48-hour BP was 132/77 mm Hg. Hypertension was defined as an awake systolic BP ≥
Prescribers were free to prescribe medicines from 5 classes (diuretic, angiotensin-converting enzyme inhibitor, angiotensin receptor blocker, calcium channel blocker, or beta-blocker) as they thought appropriate, were encouraged to use fixed-dose combination pills, and were told not to use split (eg, twice per day) dosing. Annual 48-hour ABPM was completed, and patients’ electronic health records were analyzed by blinded investigators. Median follow-up was 6.3 years, and only 84 participants failed to complete the minimum 1-year participation requirement.
Continue to: The primary outcome...
The primary outcome—a composite of cardiovascular death, myocardial infarction, coronary revascularization, heart failure, or stroke—occurred in 1752 patients, favoring the bedtime group (HR = 0.55; 95% CI, 0.50-0.61; P < .001). The calculated number of events was 1130 in the morning administration group and 622 in the evening administration group; the authors did not explicitly report the event numbers in each group. Each component of the composite outcome also favored evening administration (P < .001 for all): cardiovascular death (HR = 0.44; 95% CI, 0.34-0.56), myocardial infarction (HR = 0.66; 95% CI, 0.52-0.84), coronary revascularization (HR = 0.60; 95% CI, 0.47-0.75), heart failure (HR = 0.58; 95% CI, 0.49-0.70), and stroke (HR = 0.51; 95% CI, 0.41-0.63).
The complicated, layered study design and randomization methods limit the ability to critically appraise the study.
Smaller Spanish study also supported evening administration
A prior, smaller, prospective randomized trial conducted by the same researchers as the Hygia Project found even greater benefits to evening BP medication administration.4 The 2156 Spanish patients (52% men; average age, 55 years) from multiple primary care offices were randomized 1:1 to BP medication administration either upon awakening or at bedtime. Dozens of baseline characteristics were evenly distributed except for age (55.0 vs 56.3; P = .021) and creatinine (0.96 vs 0.98; P = .028), both of which were lower in the evening group.
After a median follow-up of 5.6 years, the bedtime group had significantly lower total events (187 events in the morning group vs 68 in the evening group; relative risk [RR] = 0.39; 95% CI, 0.29-0.51; P < .001). Individual cardiovascular outcomes also dramatically favored the evening group: total deaths (12 vs 28; P = .008), cardiovascular deaths (3 vs 14; P = .006), cardiovascular disease events (30 vs 74; P < .001), stroke (7 vs 24; P = .001), and heart failure (8 vs 33; P < .001).
Limits of both the UK trial and the Hygia Project trial included single countries of study with a lack of racial and ethnic diversity, and greater nonadherence to the evening administration of the medications.
Recommendations from others
A 2022 consensus statement from the International Society of Hypertension, published before the UK trial, recommended against bedtime dosing until more high-quality data became available. They pointed to evidence showing higher medication adherence with morning dosing, risk for asleep BP dropping, and worsening daytime BP control as reasons to continue morning administration.5 Other reviewers have questioned the Hygia Project results due to their reported implausibly large effects on cardiovascular outcomes, noting that independent attempts to verify the methods and the data have proven challenging and are not completed.6
Editor’s takeaway
I confess that I was swayed by the results of the Hygia Project; for a year or so, I advised my patients to take at least 1 BP pill at night. But after the UK study came out, I needed to reconsider. I began to worry that the great outcomes of nocturnal therapy may have been a mirage. I have returned to counseling patients to take their BP medications in whichever way fosters consistency while minimizing adverse effects for them.
Evidence summary
Recent UK study shows no difference by timing
A 2022 UK prospective, randomized, multicenter trial assigned 21,104 predominantly White adults (58% men) with hypertension to take their usual antihypertensive medication either in the morning (6
All patient baseline characteristics were equivalent between groups. If troubled by nocturia, patients in the evening group taking diuretics were told to take only the diuretic earlier (6
The median follow-up was 5.2 years. Data were collected at regular intervals through patient completion of online questionnaires and researcher analysis of National Health Service data on hospitalization and death. The intention-to-treat analysis showed no difference in the primary outcome (a composite of vascular death, nonfatal myocardial infarction, or nonfatal stroke) between the evening and morning administration groups (0.69 events vs 0.72 events per 100 person-years; hazard ratio [HR] = 0.95; 95% CI, 0.83-1.10; P = .53).
The controversial Hygia Project favored evening
Prior to the UK study was the Hygia Chronotherapy Trial, a prospective, controlled, multicenter study conducted within the primary care setting in Spain. Caucasian Spanish adults (N = 19,168; mean age, 61 years; 56% men) with hypertension were randomly assigned to take all prescribed antihypertensive medication either at bedtime or upon waking.2
The Hygia Project initially sought to establish the value of ambulatory blood pressure monitoring (ABPM) compared to office blood pressure (BP) monitoring and to explore the prognostic value of sleeping BP.3 The study objectives evolved over time. The randomization process was not clearly described,2,3 but multiple randomizations were alluded to. The authors stated that “for any of these chronotherapy trials” randomizations were done separately for “each participating center” and “randomization of participants to treatment-time regimen is done separately for each hypertension medication or combination being tested.”
The baseline characteristics of patients in the evening and morning administration groups were similar, but statistically significant differences existed in BMI (29.6 vs 29.7; P = .030) and sleep-time systolic BP percent decline (9.3 vs 9.0; P < .001). Mean baseline 48-hour BP was 132/77 mm Hg. Hypertension was defined as an awake systolic BP ≥
Prescribers were free to prescribe medicines from 5 classes (diuretic, angiotensin-converting enzyme inhibitor, angiotensin receptor blocker, calcium channel blocker, or beta-blocker) as they thought appropriate, were encouraged to use fixed-dose combination pills, and were told not to use split (eg, twice per day) dosing. Annual 48-hour ABPM was completed, and patients’ electronic health records were analyzed by blinded investigators. Median follow-up was 6.3 years, and only 84 participants failed to complete the minimum 1-year participation requirement.
Continue to: The primary outcome...
The primary outcome—a composite of cardiovascular death, myocardial infarction, coronary revascularization, heart failure, or stroke—occurred in 1752 patients, favoring the bedtime group (HR = 0.55; 95% CI, 0.50-0.61; P < .001). The calculated number of events was 1130 in the morning administration group and 622 in the evening administration group; the authors did not explicitly report the event numbers in each group. Each component of the composite outcome also favored evening administration (P < .001 for all): cardiovascular death (HR = 0.44; 95% CI, 0.34-0.56), myocardial infarction (HR = 0.66; 95% CI, 0.52-0.84), coronary revascularization (HR = 0.60; 95% CI, 0.47-0.75), heart failure (HR = 0.58; 95% CI, 0.49-0.70), and stroke (HR = 0.51; 95% CI, 0.41-0.63).
The complicated, layered study design and randomization methods limit the ability to critically appraise the study.
Smaller Spanish study also supported evening administration
A prior, smaller, prospective randomized trial conducted by the same researchers as the Hygia Project found even greater benefits to evening BP medication administration.4 The 2156 Spanish patients (52% men; average age, 55 years) from multiple primary care offices were randomized 1:1 to BP medication administration either upon awakening or at bedtime. Dozens of baseline characteristics were evenly distributed except for age (55.0 vs 56.3; P = .021) and creatinine (0.96 vs 0.98; P = .028), both of which were lower in the evening group.
After a median follow-up of 5.6 years, the bedtime group had significantly lower total events (187 events in the morning group vs 68 in the evening group; relative risk [RR] = 0.39; 95% CI, 0.29-0.51; P < .001). Individual cardiovascular outcomes also dramatically favored the evening group: total deaths (12 vs 28; P = .008), cardiovascular deaths (3 vs 14; P = .006), cardiovascular disease events (30 vs 74; P < .001), stroke (7 vs 24; P = .001), and heart failure (8 vs 33; P < .001).
Limits of both the UK trial and the Hygia Project trial included single countries of study with a lack of racial and ethnic diversity, and greater nonadherence to the evening administration of the medications.
Recommendations from others
A 2022 consensus statement from the International Society of Hypertension, published before the UK trial, recommended against bedtime dosing until more high-quality data became available. They pointed to evidence showing higher medication adherence with morning dosing, risk for asleep BP dropping, and worsening daytime BP control as reasons to continue morning administration.5 Other reviewers have questioned the Hygia Project results due to their reported implausibly large effects on cardiovascular outcomes, noting that independent attempts to verify the methods and the data have proven challenging and are not completed.6
Editor’s takeaway
I confess that I was swayed by the results of the Hygia Project; for a year or so, I advised my patients to take at least 1 BP pill at night. But after the UK study came out, I needed to reconsider. I began to worry that the great outcomes of nocturnal therapy may have been a mirage. I have returned to counseling patients to take their BP medications in whichever way fosters consistency while minimizing adverse effects for them.
1. Mackenzie IS, Rogers A, Poulter NR, et al; TIME Study Group. Cardiovascular outcomes in adults with hypertension with evening versus morning dosing of usual antihypertensives in the UK (TIME study): a prospective, randomised, open-label, blinded-endpoint clinical trial. Lancet. 2022;400:1417-1425. doi: 10.1016/S0140-6736(22)01786-X
2. Hermida RC, Crespo JJ, Domínguez-Sardiña M, et al; Hygia Project Investigators. Bedtime hypertension treatment improves cardiovascular risk reduction: the Hygia Chronotherapy Trial. Eur Heart J. 2020;41:4565-4576. doi: 10.1093/eurheartj/ehz754
3. Hermida RC. Sleep-time ambulatory blood pressure as a prognostic marker of vascular and other risks and therapeutic target for prevention by hypertension chronotherapy: rationale and design of the Hygia Project. Chronobiol Int. 2016;33:906-936. doi: 10.1080/07420528.2016.1181078
4. Hermida RC, Ayala DE, Mojón A, et al. Influence of circadian time of hypertension treatment on cardiovascular risk: results of the MAPEC study. Chronobiol Int. 2010;27:1629-1651. doi: 10.3109/07420528.2010.510230
5. Stergiou G, Brunström M, MacDonald T, et al. Bedtime dosing of antihypertensive medications: systematic review and consensus statement: International Society of Hypertension position paper endorsed by World Hypertension League and European Society of Hypertension. J Hypertens. 2022;40:1847-1858. doi: 10.1097/HJH.0000000000003240
6. Brunström M, Kjeldsen SE, Kreutz R, et al. Missing verification of source data in hypertension research: The HYGIA PROJECT in Perspective. Hypertension. 2021;78:555-558. doi: 10.1161/HYPERTENSIONAHA.121.17356
1. Mackenzie IS, Rogers A, Poulter NR, et al; TIME Study Group. Cardiovascular outcomes in adults with hypertension with evening versus morning dosing of usual antihypertensives in the UK (TIME study): a prospective, randomised, open-label, blinded-endpoint clinical trial. Lancet. 2022;400:1417-1425. doi: 10.1016/S0140-6736(22)01786-X
2. Hermida RC, Crespo JJ, Domínguez-Sardiña M, et al; Hygia Project Investigators. Bedtime hypertension treatment improves cardiovascular risk reduction: the Hygia Chronotherapy Trial. Eur Heart J. 2020;41:4565-4576. doi: 10.1093/eurheartj/ehz754
3. Hermida RC. Sleep-time ambulatory blood pressure as a prognostic marker of vascular and other risks and therapeutic target for prevention by hypertension chronotherapy: rationale and design of the Hygia Project. Chronobiol Int. 2016;33:906-936. doi: 10.1080/07420528.2016.1181078
4. Hermida RC, Ayala DE, Mojón A, et al. Influence of circadian time of hypertension treatment on cardiovascular risk: results of the MAPEC study. Chronobiol Int. 2010;27:1629-1651. doi: 10.3109/07420528.2010.510230
5. Stergiou G, Brunström M, MacDonald T, et al. Bedtime dosing of antihypertensive medications: systematic review and consensus statement: International Society of Hypertension position paper endorsed by World Hypertension League and European Society of Hypertension. J Hypertens. 2022;40:1847-1858. doi: 10.1097/HJH.0000000000003240
6. Brunström M, Kjeldsen SE, Kreutz R, et al. Missing verification of source data in hypertension research: The HYGIA PROJECT in Perspective. Hypertension. 2021;78:555-558. doi: 10.1161/HYPERTENSIONAHA.121.17356
EVIDENCE-BASED ANSWER:
51-year-old woman • history of Graves disease • general fatigue, palpitations, and hand tremors • Dx?
THE CASE
A 51-year-old Japanese woman presented with fever, sore throat, and dyspnea of less than 1 day’s duration. Although she had developed general fatigue, palpitations, and tremors of the hands 2 months earlier, she had not sought medical care.
Her medical history included Graves disease, which had been diagnosed 13 years earlier. She reported that her only medication was methimazole 10 mg/d. She did not have any family history of endocrinopathies or hematologic diseases.
Physical examination revealed a body temperature of 99.7 °F; heart rate, 130 beats/min; blood pressure, 182/62 mm Hg; respiratory rate, 46 breaths/min; and oxygen saturation, 100% on room air. Pharyngeal erythema was seen. Lung sounds were clear. The patient had tremors in her hands, tenderness of the thyroid gland, and exophthalmos. No leg edema or jugular vein distension was seen.
Laboratory tests indicated hyperthyroidism, with a thyroid-stimulating hormone level < 0.01 µIU/mL (normal range, 0.5-5 µIU/mL); free T3 level, 4.87 pg/mL (normal range, 2.3-4.3 pg/mL); and free T4 level, 2.97 ng/dL (normal range, 0.9-1.7 ng/dL). The patient also had a white blood cell (WBC) count of 1020 cells/µL (normal range, 3500-9000 cells/µL) and neutrophil count of 5 cells/µL (normal range, 1500-6500 cells/µL).
Other blood cell counts were normal, and a chest x-ray did not reveal any abnormal findings. In addition, there was no evidence to suggest hematologic malignancies or congenital neutropenia.
THE DIAGNOSIS
Based on the patient’s low WBC and neutrophil counts, agranulocytosis due to antithyroid drug therapy was suspected; however, this diagnosis would be highly unusual in the context of a 13-year history of therapy. Further history taking revealed that, because of her lack of financial means, unstable living conditions, and lack of understanding of the necessity for medication adherence, the patient had not taken methimazole
In consideration of these factors, a diagnosis of exacerbation of hyperthyroidism and agranulocytosis (due to methimazole restart and upper respiratory infection) was made.
Continue to: DISCUSSION
DISCUSSION
Agranulocytosis is a severe adverse event of antithyroid agents and requires prompt diagnosis and treatment. In a 26-year study at one clinic, it occurred in approximately 0.4% of patients taking antithyroid agents.1 The possible mechanisms of agranulocytosis are the direct toxicity of drugs and immune-mediated responses.2 Older age, female sex, and some HLA genotypes are reported to be associated with susceptibility to agranulocytosis.2
Although the development of agranulocytosis tends to be dose related, a small dose of antithyroid agent can sometimes cause the condition.3,4 It usually occurs within the first 3 months of treatment initiation, but occasionally patients develop agranulocytosis after long-term therapy.5 Interruption and subsequent resumption of the same antithyroid drug treatment also can be a risk factor for agranulocytosis, as in this case.5
Treatment includes drug cessation, administration of broad-spectrum antibiotics if infection is suspected, and granulocyte-colony stimulating factor (G-CSF) therapy.5
Our patient was hospitalized, and methimazole was stopped immediately. Administration of potassium iodide 50 mg/d and G-CSF was started. Meropenem 3 g/d also was administered for neutropenic fever.
The patient’s condition improved, and her WBC count increased to 1640 cells/µL on Day 8 and 10,890 cells/µL on Day 9. G-CSF was stopped on Day 12 and meropenem on Day 13. Bone marrow aspiration was not performed because of improvement in lab values and her overall condition. Although monitoring of WBC count during methimazole therapy is controversial,5 we decided to routinely monitor this patient due to the possibility of drug cross-reactivity.
Continue to: Despite repeated explanations...
Despite repeated explanations that it was dangerous for a patient who had developed agranulocytosis to take another antithyroid medication, the patient refused surgical treatment or radioiodine ablation because of her financial situation. (While all Japanese citizens are covered by a national health insurance program, patients ages 6 to 70 years are required to pay approximately 30% of medical and pharmaceutical costs.) On Day 21, potassium iodide was stopped, and propylthiouracil 300 mg/d was administered with careful follow-up. Agranulocytosis did not recur.
Immediate problem solved, but what about the future?
During her hospital stay, the medical team spoke with the patient many times, during which she expressed anxiety about her health conditions and the difficulties that she had experienced in her life. The clinicians acknowledged her concerns and assured the patient of their continuing commitment to her well-being even after discharge. The patient also was advised that she should take her medication as prescribed and that if she had a fever or sore throat, she should stop the medication and seek medical care as soon as possible. The patient accepted the medical team’s advice and expressed hope for the future.
Conversations about medication adherence. In 1 survey, about 60% of patients taking antithyroid drugs were unfamiliar with the symptoms of agranulocytosis.6 To deliver safe and effective treatment and detect conditions such as agranulocytosis at an early stage, clinicians must communicate clearly with patients who have hyperthyroidism, providing sufficient explanation and ensuring understanding on the patient’s part.
Patients may be reluctant to provide the details of medication adherence.7 Although it is common for patients to need services for socioeconomic issues,8 health care professionals sometimes fail to adequately discuss these issues with patients, especially if the patients are marginalized and/or have lower economic status.9 Cases such as ours underscore the importance of improving clinicians’ awareness and sensitivity to patients’ socioeconomic challenges.10,11
Our patient received information about welfare and other government services from a medical social worker during her hospital stay. She also was informed that she could seek assistance from medical social workers in the future if needed.
Continue to: The patient was discharged...
The patient was discharged on Day 28. After discharge, she took propylthiouracil as prescribed (300 mg/d), and her Graves disease was well controlled. Outpatient follow-up visits were performed every 1 or 2 months. No adverse events of propylthiouracil were seen in the ensuing time.
THE TAKEAWAY
Patients with chronic conditions sometimes discontinue medications, and they may not talk about it with their medical team, especially if they have socioeconomic or other difficulties in their lives. Clinicians should consider medication nonadherence and its risk factors when patients with chronic conditions develop unexpected adverse events.
We thank Jane Charbonneau, DVM, from Edanz for doing an English-language review of a draft of this manuscript.
CORRESPONDENCE
Takuya Maejima, MD, Department of General Medicine and Primary Care, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki 305-8576 Japan; [email protected]
1. Tajiri J, Noguchi S. Antithyroid drug-induced agranulocytosis: special reference to normal white blood cell count agranulocytosis. Thyroid. 2004;14:459-462. doi: 10.1089/105072504323150787
2. Vicente N, Cardoso L, Barros L, et al. Antithyroid drug-induced agranulocytosis: state of the art on diagnosis and management. Drugs R D. 2017;17:91-96. doi: 10.1007/s40268-017-0172-1
3. Takata K, Kubota S, Fukata S, et al. Methimazole-induced agranulocytosis in patients with Graves’ disease is more frequent with an initial dose of 30 mg daily than with 15 mg daily. Thyroid. 2009;19:559-563. doi: 10.1089/thy.2008.0364
4. Tsuboi K, Ueshiba H, Shimojo M, et al. The relation of initial methimazole dose to the incidence of methimazole-induced agranulocytosis in patients with Graves’ disease. Endocr J. 2007;54:39-43. doi: 10.1507/endocrj.k05-068
5. Burch HB, Cooper DS. Management of Graves disease: a review. J Am Med Assoc. 2015;314:2544-2554. doi: 10.1001/jama.2015.16535
6. Robinson J, Richardson M, Hickey J, et al. Patient knowledge of antithyroid drug-induced agranulocytosis. Eur Thyroid J. 2014;3:245-251. doi: https://doi.org/10.1159/000367990
7. Kini V, Ho PM. Interventions to improve medication adherence: a review. J Am Med Assoc. 2018;320:2461-2473. doi: 10.1001/jama.2018.19271
8. Vest JR, Grannis SJ, Haut DP, et al. Using structured and unstructured data to identify patients’ need for services that address the social determinants of health. Int J Med Inform. 2017;107:101-106. doi: 10.1016/j.ijmedinf.2017.09.008
9. Willems S, De Maesschalck S, Deveugele M, et al. Socio-economic status of the patient and doctor-patient communication: does it make a difference? Patient Educ Couns. 2005;56:139-146. doi: 10.1016/j.pec.2004.02.011
10. The College of Family Physicians of Canada. Best advice: social determinants of health. Accessed September 15, 2023. https://patientsmedicalhome.ca/resources/best-advice-guides/best-advice-guide-social-determinants-health/
11. Hunter K, Thomson B. A scoping review of social determinants of health curricula in post-graduate medical education. Can Med Educ J. 2019;10:e61-e71. doi: 10.36834/cmej.61709
THE CASE
A 51-year-old Japanese woman presented with fever, sore throat, and dyspnea of less than 1 day’s duration. Although she had developed general fatigue, palpitations, and tremors of the hands 2 months earlier, she had not sought medical care.
Her medical history included Graves disease, which had been diagnosed 13 years earlier. She reported that her only medication was methimazole 10 mg/d. She did not have any family history of endocrinopathies or hematologic diseases.
Physical examination revealed a body temperature of 99.7 °F; heart rate, 130 beats/min; blood pressure, 182/62 mm Hg; respiratory rate, 46 breaths/min; and oxygen saturation, 100% on room air. Pharyngeal erythema was seen. Lung sounds were clear. The patient had tremors in her hands, tenderness of the thyroid gland, and exophthalmos. No leg edema or jugular vein distension was seen.
Laboratory tests indicated hyperthyroidism, with a thyroid-stimulating hormone level < 0.01 µIU/mL (normal range, 0.5-5 µIU/mL); free T3 level, 4.87 pg/mL (normal range, 2.3-4.3 pg/mL); and free T4 level, 2.97 ng/dL (normal range, 0.9-1.7 ng/dL). The patient also had a white blood cell (WBC) count of 1020 cells/µL (normal range, 3500-9000 cells/µL) and neutrophil count of 5 cells/µL (normal range, 1500-6500 cells/µL).
Other blood cell counts were normal, and a chest x-ray did not reveal any abnormal findings. In addition, there was no evidence to suggest hematologic malignancies or congenital neutropenia.
THE DIAGNOSIS
Based on the patient’s low WBC and neutrophil counts, agranulocytosis due to antithyroid drug therapy was suspected; however, this diagnosis would be highly unusual in the context of a 13-year history of therapy. Further history taking revealed that, because of her lack of financial means, unstable living conditions, and lack of understanding of the necessity for medication adherence, the patient had not taken methimazole
In consideration of these factors, a diagnosis of exacerbation of hyperthyroidism and agranulocytosis (due to methimazole restart and upper respiratory infection) was made.
Continue to: DISCUSSION
DISCUSSION
Agranulocytosis is a severe adverse event of antithyroid agents and requires prompt diagnosis and treatment. In a 26-year study at one clinic, it occurred in approximately 0.4% of patients taking antithyroid agents.1 The possible mechanisms of agranulocytosis are the direct toxicity of drugs and immune-mediated responses.2 Older age, female sex, and some HLA genotypes are reported to be associated with susceptibility to agranulocytosis.2
Although the development of agranulocytosis tends to be dose related, a small dose of antithyroid agent can sometimes cause the condition.3,4 It usually occurs within the first 3 months of treatment initiation, but occasionally patients develop agranulocytosis after long-term therapy.5 Interruption and subsequent resumption of the same antithyroid drug treatment also can be a risk factor for agranulocytosis, as in this case.5
Treatment includes drug cessation, administration of broad-spectrum antibiotics if infection is suspected, and granulocyte-colony stimulating factor (G-CSF) therapy.5
Our patient was hospitalized, and methimazole was stopped immediately. Administration of potassium iodide 50 mg/d and G-CSF was started. Meropenem 3 g/d also was administered for neutropenic fever.
The patient’s condition improved, and her WBC count increased to 1640 cells/µL on Day 8 and 10,890 cells/µL on Day 9. G-CSF was stopped on Day 12 and meropenem on Day 13. Bone marrow aspiration was not performed because of improvement in lab values and her overall condition. Although monitoring of WBC count during methimazole therapy is controversial,5 we decided to routinely monitor this patient due to the possibility of drug cross-reactivity.
Continue to: Despite repeated explanations...
Despite repeated explanations that it was dangerous for a patient who had developed agranulocytosis to take another antithyroid medication, the patient refused surgical treatment or radioiodine ablation because of her financial situation. (While all Japanese citizens are covered by a national health insurance program, patients ages 6 to 70 years are required to pay approximately 30% of medical and pharmaceutical costs.) On Day 21, potassium iodide was stopped, and propylthiouracil 300 mg/d was administered with careful follow-up. Agranulocytosis did not recur.
Immediate problem solved, but what about the future?
During her hospital stay, the medical team spoke with the patient many times, during which she expressed anxiety about her health conditions and the difficulties that she had experienced in her life. The clinicians acknowledged her concerns and assured the patient of their continuing commitment to her well-being even after discharge. The patient also was advised that she should take her medication as prescribed and that if she had a fever or sore throat, she should stop the medication and seek medical care as soon as possible. The patient accepted the medical team’s advice and expressed hope for the future.
Conversations about medication adherence. In 1 survey, about 60% of patients taking antithyroid drugs were unfamiliar with the symptoms of agranulocytosis.6 To deliver safe and effective treatment and detect conditions such as agranulocytosis at an early stage, clinicians must communicate clearly with patients who have hyperthyroidism, providing sufficient explanation and ensuring understanding on the patient’s part.
Patients may be reluctant to provide the details of medication adherence.7 Although it is common for patients to need services for socioeconomic issues,8 health care professionals sometimes fail to adequately discuss these issues with patients, especially if the patients are marginalized and/or have lower economic status.9 Cases such as ours underscore the importance of improving clinicians’ awareness and sensitivity to patients’ socioeconomic challenges.10,11
Our patient received information about welfare and other government services from a medical social worker during her hospital stay. She also was informed that she could seek assistance from medical social workers in the future if needed.
Continue to: The patient was discharged...
The patient was discharged on Day 28. After discharge, she took propylthiouracil as prescribed (300 mg/d), and her Graves disease was well controlled. Outpatient follow-up visits were performed every 1 or 2 months. No adverse events of propylthiouracil were seen in the ensuing time.
THE TAKEAWAY
Patients with chronic conditions sometimes discontinue medications, and they may not talk about it with their medical team, especially if they have socioeconomic or other difficulties in their lives. Clinicians should consider medication nonadherence and its risk factors when patients with chronic conditions develop unexpected adverse events.
We thank Jane Charbonneau, DVM, from Edanz for doing an English-language review of a draft of this manuscript.
CORRESPONDENCE
Takuya Maejima, MD, Department of General Medicine and Primary Care, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki 305-8576 Japan; [email protected]
THE CASE
A 51-year-old Japanese woman presented with fever, sore throat, and dyspnea of less than 1 day’s duration. Although she had developed general fatigue, palpitations, and tremors of the hands 2 months earlier, she had not sought medical care.
Her medical history included Graves disease, which had been diagnosed 13 years earlier. She reported that her only medication was methimazole 10 mg/d. She did not have any family history of endocrinopathies or hematologic diseases.
Physical examination revealed a body temperature of 99.7 °F; heart rate, 130 beats/min; blood pressure, 182/62 mm Hg; respiratory rate, 46 breaths/min; and oxygen saturation, 100% on room air. Pharyngeal erythema was seen. Lung sounds were clear. The patient had tremors in her hands, tenderness of the thyroid gland, and exophthalmos. No leg edema or jugular vein distension was seen.
Laboratory tests indicated hyperthyroidism, with a thyroid-stimulating hormone level < 0.01 µIU/mL (normal range, 0.5-5 µIU/mL); free T3 level, 4.87 pg/mL (normal range, 2.3-4.3 pg/mL); and free T4 level, 2.97 ng/dL (normal range, 0.9-1.7 ng/dL). The patient also had a white blood cell (WBC) count of 1020 cells/µL (normal range, 3500-9000 cells/µL) and neutrophil count of 5 cells/µL (normal range, 1500-6500 cells/µL).
Other blood cell counts were normal, and a chest x-ray did not reveal any abnormal findings. In addition, there was no evidence to suggest hematologic malignancies or congenital neutropenia.
THE DIAGNOSIS
Based on the patient’s low WBC and neutrophil counts, agranulocytosis due to antithyroid drug therapy was suspected; however, this diagnosis would be highly unusual in the context of a 13-year history of therapy. Further history taking revealed that, because of her lack of financial means, unstable living conditions, and lack of understanding of the necessity for medication adherence, the patient had not taken methimazole
In consideration of these factors, a diagnosis of exacerbation of hyperthyroidism and agranulocytosis (due to methimazole restart and upper respiratory infection) was made.
Continue to: DISCUSSION
DISCUSSION
Agranulocytosis is a severe adverse event of antithyroid agents and requires prompt diagnosis and treatment. In a 26-year study at one clinic, it occurred in approximately 0.4% of patients taking antithyroid agents.1 The possible mechanisms of agranulocytosis are the direct toxicity of drugs and immune-mediated responses.2 Older age, female sex, and some HLA genotypes are reported to be associated with susceptibility to agranulocytosis.2
Although the development of agranulocytosis tends to be dose related, a small dose of antithyroid agent can sometimes cause the condition.3,4 It usually occurs within the first 3 months of treatment initiation, but occasionally patients develop agranulocytosis after long-term therapy.5 Interruption and subsequent resumption of the same antithyroid drug treatment also can be a risk factor for agranulocytosis, as in this case.5
Treatment includes drug cessation, administration of broad-spectrum antibiotics if infection is suspected, and granulocyte-colony stimulating factor (G-CSF) therapy.5
Our patient was hospitalized, and methimazole was stopped immediately. Administration of potassium iodide 50 mg/d and G-CSF was started. Meropenem 3 g/d also was administered for neutropenic fever.
The patient’s condition improved, and her WBC count increased to 1640 cells/µL on Day 8 and 10,890 cells/µL on Day 9. G-CSF was stopped on Day 12 and meropenem on Day 13. Bone marrow aspiration was not performed because of improvement in lab values and her overall condition. Although monitoring of WBC count during methimazole therapy is controversial,5 we decided to routinely monitor this patient due to the possibility of drug cross-reactivity.
Continue to: Despite repeated explanations...
Despite repeated explanations that it was dangerous for a patient who had developed agranulocytosis to take another antithyroid medication, the patient refused surgical treatment or radioiodine ablation because of her financial situation. (While all Japanese citizens are covered by a national health insurance program, patients ages 6 to 70 years are required to pay approximately 30% of medical and pharmaceutical costs.) On Day 21, potassium iodide was stopped, and propylthiouracil 300 mg/d was administered with careful follow-up. Agranulocytosis did not recur.
Immediate problem solved, but what about the future?
During her hospital stay, the medical team spoke with the patient many times, during which she expressed anxiety about her health conditions and the difficulties that she had experienced in her life. The clinicians acknowledged her concerns and assured the patient of their continuing commitment to her well-being even after discharge. The patient also was advised that she should take her medication as prescribed and that if she had a fever or sore throat, she should stop the medication and seek medical care as soon as possible. The patient accepted the medical team’s advice and expressed hope for the future.
Conversations about medication adherence. In 1 survey, about 60% of patients taking antithyroid drugs were unfamiliar with the symptoms of agranulocytosis.6 To deliver safe and effective treatment and detect conditions such as agranulocytosis at an early stage, clinicians must communicate clearly with patients who have hyperthyroidism, providing sufficient explanation and ensuring understanding on the patient’s part.
Patients may be reluctant to provide the details of medication adherence.7 Although it is common for patients to need services for socioeconomic issues,8 health care professionals sometimes fail to adequately discuss these issues with patients, especially if the patients are marginalized and/or have lower economic status.9 Cases such as ours underscore the importance of improving clinicians’ awareness and sensitivity to patients’ socioeconomic challenges.10,11
Our patient received information about welfare and other government services from a medical social worker during her hospital stay. She also was informed that she could seek assistance from medical social workers in the future if needed.
Continue to: The patient was discharged...
The patient was discharged on Day 28. After discharge, she took propylthiouracil as prescribed (300 mg/d), and her Graves disease was well controlled. Outpatient follow-up visits were performed every 1 or 2 months. No adverse events of propylthiouracil were seen in the ensuing time.
THE TAKEAWAY
Patients with chronic conditions sometimes discontinue medications, and they may not talk about it with their medical team, especially if they have socioeconomic or other difficulties in their lives. Clinicians should consider medication nonadherence and its risk factors when patients with chronic conditions develop unexpected adverse events.
We thank Jane Charbonneau, DVM, from Edanz for doing an English-language review of a draft of this manuscript.
CORRESPONDENCE
Takuya Maejima, MD, Department of General Medicine and Primary Care, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki 305-8576 Japan; [email protected]
1. Tajiri J, Noguchi S. Antithyroid drug-induced agranulocytosis: special reference to normal white blood cell count agranulocytosis. Thyroid. 2004;14:459-462. doi: 10.1089/105072504323150787
2. Vicente N, Cardoso L, Barros L, et al. Antithyroid drug-induced agranulocytosis: state of the art on diagnosis and management. Drugs R D. 2017;17:91-96. doi: 10.1007/s40268-017-0172-1
3. Takata K, Kubota S, Fukata S, et al. Methimazole-induced agranulocytosis in patients with Graves’ disease is more frequent with an initial dose of 30 mg daily than with 15 mg daily. Thyroid. 2009;19:559-563. doi: 10.1089/thy.2008.0364
4. Tsuboi K, Ueshiba H, Shimojo M, et al. The relation of initial methimazole dose to the incidence of methimazole-induced agranulocytosis in patients with Graves’ disease. Endocr J. 2007;54:39-43. doi: 10.1507/endocrj.k05-068
5. Burch HB, Cooper DS. Management of Graves disease: a review. J Am Med Assoc. 2015;314:2544-2554. doi: 10.1001/jama.2015.16535
6. Robinson J, Richardson M, Hickey J, et al. Patient knowledge of antithyroid drug-induced agranulocytosis. Eur Thyroid J. 2014;3:245-251. doi: https://doi.org/10.1159/000367990
7. Kini V, Ho PM. Interventions to improve medication adherence: a review. J Am Med Assoc. 2018;320:2461-2473. doi: 10.1001/jama.2018.19271
8. Vest JR, Grannis SJ, Haut DP, et al. Using structured and unstructured data to identify patients’ need for services that address the social determinants of health. Int J Med Inform. 2017;107:101-106. doi: 10.1016/j.ijmedinf.2017.09.008
9. Willems S, De Maesschalck S, Deveugele M, et al. Socio-economic status of the patient and doctor-patient communication: does it make a difference? Patient Educ Couns. 2005;56:139-146. doi: 10.1016/j.pec.2004.02.011
10. The College of Family Physicians of Canada. Best advice: social determinants of health. Accessed September 15, 2023. https://patientsmedicalhome.ca/resources/best-advice-guides/best-advice-guide-social-determinants-health/
11. Hunter K, Thomson B. A scoping review of social determinants of health curricula in post-graduate medical education. Can Med Educ J. 2019;10:e61-e71. doi: 10.36834/cmej.61709
1. Tajiri J, Noguchi S. Antithyroid drug-induced agranulocytosis: special reference to normal white blood cell count agranulocytosis. Thyroid. 2004;14:459-462. doi: 10.1089/105072504323150787
2. Vicente N, Cardoso L, Barros L, et al. Antithyroid drug-induced agranulocytosis: state of the art on diagnosis and management. Drugs R D. 2017;17:91-96. doi: 10.1007/s40268-017-0172-1
3. Takata K, Kubota S, Fukata S, et al. Methimazole-induced agranulocytosis in patients with Graves’ disease is more frequent with an initial dose of 30 mg daily than with 15 mg daily. Thyroid. 2009;19:559-563. doi: 10.1089/thy.2008.0364
4. Tsuboi K, Ueshiba H, Shimojo M, et al. The relation of initial methimazole dose to the incidence of methimazole-induced agranulocytosis in patients with Graves’ disease. Endocr J. 2007;54:39-43. doi: 10.1507/endocrj.k05-068
5. Burch HB, Cooper DS. Management of Graves disease: a review. J Am Med Assoc. 2015;314:2544-2554. doi: 10.1001/jama.2015.16535
6. Robinson J, Richardson M, Hickey J, et al. Patient knowledge of antithyroid drug-induced agranulocytosis. Eur Thyroid J. 2014;3:245-251. doi: https://doi.org/10.1159/000367990
7. Kini V, Ho PM. Interventions to improve medication adherence: a review. J Am Med Assoc. 2018;320:2461-2473. doi: 10.1001/jama.2018.19271
8. Vest JR, Grannis SJ, Haut DP, et al. Using structured and unstructured data to identify patients’ need for services that address the social determinants of health. Int J Med Inform. 2017;107:101-106. doi: 10.1016/j.ijmedinf.2017.09.008
9. Willems S, De Maesschalck S, Deveugele M, et al. Socio-economic status of the patient and doctor-patient communication: does it make a difference? Patient Educ Couns. 2005;56:139-146. doi: 10.1016/j.pec.2004.02.011
10. The College of Family Physicians of Canada. Best advice: social determinants of health. Accessed September 15, 2023. https://patientsmedicalhome.ca/resources/best-advice-guides/best-advice-guide-social-determinants-health/
11. Hunter K, Thomson B. A scoping review of social determinants of health curricula in post-graduate medical education. Can Med Educ J. 2019;10:e61-e71. doi: 10.36834/cmej.61709
► History of Graves disease
► General fatigue, palpitations, and hand tremors