When should travelers begin malaria prophylaxis?

Article Type
Changed
Mon, 01/14/2019 - 11:21
Display Headline
When should travelers begin malaria prophylaxis?
EVIDENCE-BASED ANSWER

Travelers should start on chloroquine 1 to 2 weeks before entering an area without chloroquine resistance (strength of recommendation [SOR]: C, based on expert opinion). In areas with chloroquine-resistant Plasmodium falciparum, travelers will need to take atovaquone/proguanil, doxycycline, or primaquine 1 day before entering the area, or mefloquine 2 to 7 weeks before travel (SOR: B, based on prospective patient-oriented outcomes and expert opinion).

Before prescribing medications, determine malaria risk and sensitivity of Plasmodium species by country at wwwn.cdc.gov/travel/yellowBookCh5MalariaYellowFeverTable.aspx (SOR: C, based on patient-oriented expert opinion).

Clinical commentary

5 tips to help travelers avoid malaria
Brian V. Reamy, MD
Uniformed Services University, Bethesda, Md

Despite our best efforts, more than 10,000 American and European travelers contract malaria each year. Five clinical pointers are helpful in prescribing malaria prophylaxis and preventing malaria in travelers.

1. Advise patients that they’ll need to get their antimalarials before they leave for their trip. The CDC recommends against the purchase of antimalarials while overseas because of concerns about product quality.

2. Encourage patients to plan ahead. Most local community pharmacies do not routinely stock antimalarials and must special order them. If a patient mentions an upcoming trip, advise them that they’ll need to allow an extra 2 weeks to obtain their medications.

3. Consult 1 of 2 continuously updated Web sites prior to selecting a medication for malaria prophylaxis: wwwn.cdc.gov/travel/destinationList.aspx or www.who.int/ith/en.

Start times vary from 1 day to several weeks prior to travel based on the medication selected.

4. Encourage patients to spray clothing with permethrin prior to travel. Permethrin remains effective as a repellent even after months of clothing use and multiple washes.

5. Encourage travelers to finish their medication after they return and to report unexplained fevers for up to 1 year after travel.

Evidence summary

 

Travelers to malaria-endemic areas should avoid mosquito bites by using netting and repellents, and use chemoprophylaxis to prevent infection.

Although no drug regimen guarantees protection against malaria, physicians should prescribe 1 of several options based on the location of travel, the susceptibility of indigenous P falciparum, and the side-effect profile.1

Timing and dosage of prophylactic drugs

Prophylactic medications must be started at different times before travel, but for some medications the optimal time to initiate treatment is unclear. Evidence-based recommendations2,3 with consideration for side-effect profiles are given in the TABLE.

In contrast to the pretreatment times for all other malarial prophylaxes, the generally accepted pretreatment time for mefloquine is 1 to 2 weeks before entering a risk area. However, this may still be inadequate due to the drug’s long half-life, which results in a long delay in reaching therapeutic blood levels.4 The evidence indicates that mefloquine should be started at least 2, and as many as 7, weeks before travel.

The standard recommended dose of 250 mg/week of mefloquine “produces maximum steady-state plasma concentrations of 1000 to 2000 mcg/L, which are reached only after 7 to 10 weeks.”4 One study of 293 children under the age of 5 years in Malawi found that plasma concentrations of mefloquine were below prophylactic level (500 mcg/mL) against P falciparum until the fourth to seventh week of once-weekly dosing (P<.0003).5

One way of reaching prophylactic levels earlier would be to give mefloquine 250 mg daily for 3 days followed by 250 mg weekly.4 A safety study of 157 healthy US Marine volunteers showed that preloading achieves prophylactic blood levels of mefloquine by the third day while weekly mefloquine is subprophylactic until the fifth week.4

 

 

While a study of the long-term use of mefloquine in 421 healthy Peace Corps volunteers has shown it to be safe,6 clinical trials and case reports indicate that a loading dose of mefloquine is associated with adverse drug events, which include neuropsychiatric and gastrointestinal symptoms.4,7

TABLE
Evidence-based recommendations for prevention of malaria2-3,8

DRUGUSAGEADULT DOSETREATMENT SCHEDULE
Atovaquone/proguanil Contraindicated in pregnancyProphylaxis in areas with chloroquine-resistant or mefloquine-resistant P falciparum1 tablet orally each day 250 mg atovaquone and 100 mg proguanil hydrochloride)Daily from 1 day prior to entry until 7 days after leaving
ChloroquineProphylaxis only in areas with chloroquine-sensitive P falciparum300 mg base (500 mg salt) orally, once/weekWeekly from 2 weeks prior to entry until 4 weeks after leaving (take on the same day of the week)
Doxycycline Contraindicated in children <8 years of age and pregnant womenProphylaxis in areas with chloroquine-resistant or mefloquine-resistant P falciparum100 mg orally, dailyDaily from 1 day prior to entry until 4 weeks after leaving
MefloquineProphylaxis in areas with chloroquine-resistant P falciparum228 mg base (250 mg salt) orally, once/weekWeekly from 2–7 weeks before entry until 4 weeks after leaving (take on the same day of the week)
PrimaquineAn option for prophylaxis in special circumstances30 mg base (52.6 mg salt) orally, dailyDaily from 1 day prior to entry until 7 days after leaving
 

Recommendations from others

The World Health Organization (WHO) states that “weekly mefloquine should be started at least 1 week, but preferably 2–3 weeks before departure, to achieve higher pre-travel blood levels and to allow side effects to be detected before travel so that possible alternatives can be considered.”8

Centers for Disease Control and Prevention recommendations integrate recommendations from WHO and Cochrane.

Acknowledgments

The opinions and assertions contained herein are the private views of the authors and not to be construed as official, or as reflecting the views of the US Air Force Medical Service or the US Air Force at large.

References

1. Chen LH, Keystone JS. New strategies for the prevention of malaria in travelers. Infect Dis Clin North Am 2005;19:185-210.

2. Physicians’ Desk Reference. 61st ed. Montvale, NJ: Thomson; 2007:2786.

3. Parise M, Barber A, Mali S. Prevention of specific infectious diseases—malaria. In Arguin PM, Kozarsky PE, Navin AW (eds), Health Information for International Travel 2005-2006. Atlanta, Ga: US Department of Health and Human Services, Public Health Service, Centers for Disease Control and Prevention; 2007. Available at: wwwn.cdc.gov/travel/yellowBookCh4-Malaria.aspx. Accessed on October 11, 2007.

4. Boudreau E, Schuster B, Sanchez J, et al. Tolerability of prophylactic Lariam regimens. Trop Med Parasitol 1993;44:257-265.

5. Slutsker LM, Khoromana CO, Payne D, et al. Mefloquine therapy for Plasmodium falciparum. malaria in children under 5 years of age in Malawi: in vivo/in vitro efficacy and correlation of drug concentration with parasitological outcome. Bull World Health Organ 1990;68:53-59.

6. Lobel HO, Miani M, Eng T, et al. Long-term malaria prophylaxis with weekly mefloquine. Lancet 1993;341:848-51.

7. Schlagenhauf P. Mefloquine for malaria chemoprophylaxis 1992-1998: a review. J Travel Med 1999;6:122-133.

8. International Travel and Health 2005. Chapter 7: Malaria. Geneva: World Health Organization; 2005. Available at: whqlibdoc.who.int/publications/2005/9241580364_chap7.pdf. Accessed on October 11, 2007.

Article PDF
Author and Disclosure Information

Steven L. Clark, MD
Paul Crawford, MD
Eglin Air Force Base Family Medicine Residency, Eglin Air Force Base, Fla

William Nichols, MLS
Librarian, Eglin Regional Hospital, Eglin Air Force Base, Fla

Issue
The Journal of Family Practice - 56(11)
Publications
Topics
Page Number
950-951
Legacy Keywords
Steven L. Clark;MD; Paul Crawford;MD; William Nichols;MLS; Brian V. Reamy ;MD; malaria; prophylaxis; anopheles; chloroquine; resistant; Plasmodium falciparum; travel; atovaquone; proguanil; doxycycline; primaquine; mefloquine; schedule; infections; parasite; CDC; WHO
Sections
Author and Disclosure Information

Steven L. Clark, MD
Paul Crawford, MD
Eglin Air Force Base Family Medicine Residency, Eglin Air Force Base, Fla

William Nichols, MLS
Librarian, Eglin Regional Hospital, Eglin Air Force Base, Fla

Author and Disclosure Information

Steven L. Clark, MD
Paul Crawford, MD
Eglin Air Force Base Family Medicine Residency, Eglin Air Force Base, Fla

William Nichols, MLS
Librarian, Eglin Regional Hospital, Eglin Air Force Base, Fla

Article PDF
Article PDF
EVIDENCE-BASED ANSWER

Travelers should start on chloroquine 1 to 2 weeks before entering an area without chloroquine resistance (strength of recommendation [SOR]: C, based on expert opinion). In areas with chloroquine-resistant Plasmodium falciparum, travelers will need to take atovaquone/proguanil, doxycycline, or primaquine 1 day before entering the area, or mefloquine 2 to 7 weeks before travel (SOR: B, based on prospective patient-oriented outcomes and expert opinion).

Before prescribing medications, determine malaria risk and sensitivity of Plasmodium species by country at wwwn.cdc.gov/travel/yellowBookCh5MalariaYellowFeverTable.aspx (SOR: C, based on patient-oriented expert opinion).

Clinical commentary

5 tips to help travelers avoid malaria
Brian V. Reamy, MD
Uniformed Services University, Bethesda, Md

Despite our best efforts, more than 10,000 American and European travelers contract malaria each year. Five clinical pointers are helpful in prescribing malaria prophylaxis and preventing malaria in travelers.

1. Advise patients that they’ll need to get their antimalarials before they leave for their trip. The CDC recommends against the purchase of antimalarials while overseas because of concerns about product quality.

2. Encourage patients to plan ahead. Most local community pharmacies do not routinely stock antimalarials and must special order them. If a patient mentions an upcoming trip, advise them that they’ll need to allow an extra 2 weeks to obtain their medications.

3. Consult 1 of 2 continuously updated Web sites prior to selecting a medication for malaria prophylaxis: wwwn.cdc.gov/travel/destinationList.aspx or www.who.int/ith/en.

Start times vary from 1 day to several weeks prior to travel based on the medication selected.

4. Encourage patients to spray clothing with permethrin prior to travel. Permethrin remains effective as a repellent even after months of clothing use and multiple washes.

5. Encourage travelers to finish their medication after they return and to report unexplained fevers for up to 1 year after travel.

Evidence summary

 

Travelers to malaria-endemic areas should avoid mosquito bites by using netting and repellents, and use chemoprophylaxis to prevent infection.

Although no drug regimen guarantees protection against malaria, physicians should prescribe 1 of several options based on the location of travel, the susceptibility of indigenous P falciparum, and the side-effect profile.1

Timing and dosage of prophylactic drugs

Prophylactic medications must be started at different times before travel, but for some medications the optimal time to initiate treatment is unclear. Evidence-based recommendations2,3 with consideration for side-effect profiles are given in the TABLE.

In contrast to the pretreatment times for all other malarial prophylaxes, the generally accepted pretreatment time for mefloquine is 1 to 2 weeks before entering a risk area. However, this may still be inadequate due to the drug’s long half-life, which results in a long delay in reaching therapeutic blood levels.4 The evidence indicates that mefloquine should be started at least 2, and as many as 7, weeks before travel.

The standard recommended dose of 250 mg/week of mefloquine “produces maximum steady-state plasma concentrations of 1000 to 2000 mcg/L, which are reached only after 7 to 10 weeks.”4 One study of 293 children under the age of 5 years in Malawi found that plasma concentrations of mefloquine were below prophylactic level (500 mcg/mL) against P falciparum until the fourth to seventh week of once-weekly dosing (P<.0003).5

One way of reaching prophylactic levels earlier would be to give mefloquine 250 mg daily for 3 days followed by 250 mg weekly.4 A safety study of 157 healthy US Marine volunteers showed that preloading achieves prophylactic blood levels of mefloquine by the third day while weekly mefloquine is subprophylactic until the fifth week.4

 

 

While a study of the long-term use of mefloquine in 421 healthy Peace Corps volunteers has shown it to be safe,6 clinical trials and case reports indicate that a loading dose of mefloquine is associated with adverse drug events, which include neuropsychiatric and gastrointestinal symptoms.4,7

TABLE
Evidence-based recommendations for prevention of malaria2-3,8

DRUGUSAGEADULT DOSETREATMENT SCHEDULE
Atovaquone/proguanil Contraindicated in pregnancyProphylaxis in areas with chloroquine-resistant or mefloquine-resistant P falciparum1 tablet orally each day 250 mg atovaquone and 100 mg proguanil hydrochloride)Daily from 1 day prior to entry until 7 days after leaving
ChloroquineProphylaxis only in areas with chloroquine-sensitive P falciparum300 mg base (500 mg salt) orally, once/weekWeekly from 2 weeks prior to entry until 4 weeks after leaving (take on the same day of the week)
Doxycycline Contraindicated in children <8 years of age and pregnant womenProphylaxis in areas with chloroquine-resistant or mefloquine-resistant P falciparum100 mg orally, dailyDaily from 1 day prior to entry until 4 weeks after leaving
MefloquineProphylaxis in areas with chloroquine-resistant P falciparum228 mg base (250 mg salt) orally, once/weekWeekly from 2–7 weeks before entry until 4 weeks after leaving (take on the same day of the week)
PrimaquineAn option for prophylaxis in special circumstances30 mg base (52.6 mg salt) orally, dailyDaily from 1 day prior to entry until 7 days after leaving
 

Recommendations from others

The World Health Organization (WHO) states that “weekly mefloquine should be started at least 1 week, but preferably 2–3 weeks before departure, to achieve higher pre-travel blood levels and to allow side effects to be detected before travel so that possible alternatives can be considered.”8

Centers for Disease Control and Prevention recommendations integrate recommendations from WHO and Cochrane.

Acknowledgments

The opinions and assertions contained herein are the private views of the authors and not to be construed as official, or as reflecting the views of the US Air Force Medical Service or the US Air Force at large.

EVIDENCE-BASED ANSWER

Travelers should start on chloroquine 1 to 2 weeks before entering an area without chloroquine resistance (strength of recommendation [SOR]: C, based on expert opinion). In areas with chloroquine-resistant Plasmodium falciparum, travelers will need to take atovaquone/proguanil, doxycycline, or primaquine 1 day before entering the area, or mefloquine 2 to 7 weeks before travel (SOR: B, based on prospective patient-oriented outcomes and expert opinion).

Before prescribing medications, determine malaria risk and sensitivity of Plasmodium species by country at wwwn.cdc.gov/travel/yellowBookCh5MalariaYellowFeverTable.aspx (SOR: C, based on patient-oriented expert opinion).

Clinical commentary

5 tips to help travelers avoid malaria
Brian V. Reamy, MD
Uniformed Services University, Bethesda, Md

Despite our best efforts, more than 10,000 American and European travelers contract malaria each year. Five clinical pointers are helpful in prescribing malaria prophylaxis and preventing malaria in travelers.

1. Advise patients that they’ll need to get their antimalarials before they leave for their trip. The CDC recommends against the purchase of antimalarials while overseas because of concerns about product quality.

2. Encourage patients to plan ahead. Most local community pharmacies do not routinely stock antimalarials and must special order them. If a patient mentions an upcoming trip, advise them that they’ll need to allow an extra 2 weeks to obtain their medications.

3. Consult 1 of 2 continuously updated Web sites prior to selecting a medication for malaria prophylaxis: wwwn.cdc.gov/travel/destinationList.aspx or www.who.int/ith/en.

Start times vary from 1 day to several weeks prior to travel based on the medication selected.

4. Encourage patients to spray clothing with permethrin prior to travel. Permethrin remains effective as a repellent even after months of clothing use and multiple washes.

5. Encourage travelers to finish their medication after they return and to report unexplained fevers for up to 1 year after travel.

Evidence summary

 

Travelers to malaria-endemic areas should avoid mosquito bites by using netting and repellents, and use chemoprophylaxis to prevent infection.

Although no drug regimen guarantees protection against malaria, physicians should prescribe 1 of several options based on the location of travel, the susceptibility of indigenous P falciparum, and the side-effect profile.1

Timing and dosage of prophylactic drugs

Prophylactic medications must be started at different times before travel, but for some medications the optimal time to initiate treatment is unclear. Evidence-based recommendations2,3 with consideration for side-effect profiles are given in the TABLE.

In contrast to the pretreatment times for all other malarial prophylaxes, the generally accepted pretreatment time for mefloquine is 1 to 2 weeks before entering a risk area. However, this may still be inadequate due to the drug’s long half-life, which results in a long delay in reaching therapeutic blood levels.4 The evidence indicates that mefloquine should be started at least 2, and as many as 7, weeks before travel.

The standard recommended dose of 250 mg/week of mefloquine “produces maximum steady-state plasma concentrations of 1000 to 2000 mcg/L, which are reached only after 7 to 10 weeks.”4 One study of 293 children under the age of 5 years in Malawi found that plasma concentrations of mefloquine were below prophylactic level (500 mcg/mL) against P falciparum until the fourth to seventh week of once-weekly dosing (P<.0003).5

One way of reaching prophylactic levels earlier would be to give mefloquine 250 mg daily for 3 days followed by 250 mg weekly.4 A safety study of 157 healthy US Marine volunteers showed that preloading achieves prophylactic blood levels of mefloquine by the third day while weekly mefloquine is subprophylactic until the fifth week.4

 

 

While a study of the long-term use of mefloquine in 421 healthy Peace Corps volunteers has shown it to be safe,6 clinical trials and case reports indicate that a loading dose of mefloquine is associated with adverse drug events, which include neuropsychiatric and gastrointestinal symptoms.4,7

TABLE
Evidence-based recommendations for prevention of malaria2-3,8

DRUGUSAGEADULT DOSETREATMENT SCHEDULE
Atovaquone/proguanil Contraindicated in pregnancyProphylaxis in areas with chloroquine-resistant or mefloquine-resistant P falciparum1 tablet orally each day 250 mg atovaquone and 100 mg proguanil hydrochloride)Daily from 1 day prior to entry until 7 days after leaving
ChloroquineProphylaxis only in areas with chloroquine-sensitive P falciparum300 mg base (500 mg salt) orally, once/weekWeekly from 2 weeks prior to entry until 4 weeks after leaving (take on the same day of the week)
Doxycycline Contraindicated in children <8 years of age and pregnant womenProphylaxis in areas with chloroquine-resistant or mefloquine-resistant P falciparum100 mg orally, dailyDaily from 1 day prior to entry until 4 weeks after leaving
MefloquineProphylaxis in areas with chloroquine-resistant P falciparum228 mg base (250 mg salt) orally, once/weekWeekly from 2–7 weeks before entry until 4 weeks after leaving (take on the same day of the week)
PrimaquineAn option for prophylaxis in special circumstances30 mg base (52.6 mg salt) orally, dailyDaily from 1 day prior to entry until 7 days after leaving
 

Recommendations from others

The World Health Organization (WHO) states that “weekly mefloquine should be started at least 1 week, but preferably 2–3 weeks before departure, to achieve higher pre-travel blood levels and to allow side effects to be detected before travel so that possible alternatives can be considered.”8

Centers for Disease Control and Prevention recommendations integrate recommendations from WHO and Cochrane.

Acknowledgments

The opinions and assertions contained herein are the private views of the authors and not to be construed as official, or as reflecting the views of the US Air Force Medical Service or the US Air Force at large.

References

1. Chen LH, Keystone JS. New strategies for the prevention of malaria in travelers. Infect Dis Clin North Am 2005;19:185-210.

2. Physicians’ Desk Reference. 61st ed. Montvale, NJ: Thomson; 2007:2786.

3. Parise M, Barber A, Mali S. Prevention of specific infectious diseases—malaria. In Arguin PM, Kozarsky PE, Navin AW (eds), Health Information for International Travel 2005-2006. Atlanta, Ga: US Department of Health and Human Services, Public Health Service, Centers for Disease Control and Prevention; 2007. Available at: wwwn.cdc.gov/travel/yellowBookCh4-Malaria.aspx. Accessed on October 11, 2007.

4. Boudreau E, Schuster B, Sanchez J, et al. Tolerability of prophylactic Lariam regimens. Trop Med Parasitol 1993;44:257-265.

5. Slutsker LM, Khoromana CO, Payne D, et al. Mefloquine therapy for Plasmodium falciparum. malaria in children under 5 years of age in Malawi: in vivo/in vitro efficacy and correlation of drug concentration with parasitological outcome. Bull World Health Organ 1990;68:53-59.

6. Lobel HO, Miani M, Eng T, et al. Long-term malaria prophylaxis with weekly mefloquine. Lancet 1993;341:848-51.

7. Schlagenhauf P. Mefloquine for malaria chemoprophylaxis 1992-1998: a review. J Travel Med 1999;6:122-133.

8. International Travel and Health 2005. Chapter 7: Malaria. Geneva: World Health Organization; 2005. Available at: whqlibdoc.who.int/publications/2005/9241580364_chap7.pdf. Accessed on October 11, 2007.

References

1. Chen LH, Keystone JS. New strategies for the prevention of malaria in travelers. Infect Dis Clin North Am 2005;19:185-210.

2. Physicians’ Desk Reference. 61st ed. Montvale, NJ: Thomson; 2007:2786.

3. Parise M, Barber A, Mali S. Prevention of specific infectious diseases—malaria. In Arguin PM, Kozarsky PE, Navin AW (eds), Health Information for International Travel 2005-2006. Atlanta, Ga: US Department of Health and Human Services, Public Health Service, Centers for Disease Control and Prevention; 2007. Available at: wwwn.cdc.gov/travel/yellowBookCh4-Malaria.aspx. Accessed on October 11, 2007.

4. Boudreau E, Schuster B, Sanchez J, et al. Tolerability of prophylactic Lariam regimens. Trop Med Parasitol 1993;44:257-265.

5. Slutsker LM, Khoromana CO, Payne D, et al. Mefloquine therapy for Plasmodium falciparum. malaria in children under 5 years of age in Malawi: in vivo/in vitro efficacy and correlation of drug concentration with parasitological outcome. Bull World Health Organ 1990;68:53-59.

6. Lobel HO, Miani M, Eng T, et al. Long-term malaria prophylaxis with weekly mefloquine. Lancet 1993;341:848-51.

7. Schlagenhauf P. Mefloquine for malaria chemoprophylaxis 1992-1998: a review. J Travel Med 1999;6:122-133.

8. International Travel and Health 2005. Chapter 7: Malaria. Geneva: World Health Organization; 2005. Available at: whqlibdoc.who.int/publications/2005/9241580364_chap7.pdf. Accessed on October 11, 2007.

Issue
The Journal of Family Practice - 56(11)
Issue
The Journal of Family Practice - 56(11)
Page Number
950-951
Page Number
950-951
Publications
Publications
Topics
Article Type
Display Headline
When should travelers begin malaria prophylaxis?
Display Headline
When should travelers begin malaria prophylaxis?
Legacy Keywords
Steven L. Clark;MD; Paul Crawford;MD; William Nichols;MLS; Brian V. Reamy ;MD; malaria; prophylaxis; anopheles; chloroquine; resistant; Plasmodium falciparum; travel; atovaquone; proguanil; doxycycline; primaquine; mefloquine; schedule; infections; parasite; CDC; WHO
Legacy Keywords
Steven L. Clark;MD; Paul Crawford;MD; William Nichols;MLS; Brian V. Reamy ;MD; malaria; prophylaxis; anopheles; chloroquine; resistant; Plasmodium falciparum; travel; atovaquone; proguanil; doxycycline; primaquine; mefloquine; schedule; infections; parasite; CDC; WHO
Sections
PURLs Copyright

Evidence-based answers from the Family Physicians Inquiries Network

Disallow All Ads
Article PDF Media

What is the best initial treatment for orbital cellulitis in children?

Article Type
Changed
Mon, 01/14/2019 - 11:20
Display Headline
What is the best initial treatment for orbital cellulitis in children?
EVIDENCE-BASED ANSWER

Although antibiotics are the best initial treatment, surgical intervention is warranted when a child has:

  • visual impairment, complete ophthalmoplegia, or well-defined abscess on presentation, or
  • no clearly apparent clinical improvement by 24 hours (strength of recommendation [SOR]: C, based on patient-oriented case-series studies).

Target antimicrobial therapy toward the common pathogens associated with predisposing factors for orbital cellulitis, such as sinusitis—and pay attention to local resistance patterns (SOR: C, based on patient-oriented case series).

Clinical commentary

Rare but serious risk factors
Peter C. Smith, MD
Rose Family Medicine Residency, University of Colorado Health Sciences Center, Denver

The incidence of Haemophilus influenzae–related periorbital cellulitis appears to have plummeted with the advent of Hib vaccine. And while no national data have been published, case series support my clinical observation that the overall incidence of periorbital cellulitis has dropped as well.

The arrival of heptavalent pneumococcal vaccine may further contribute to its welcome scarcity. take this changing bacteriology—in conjunction with local resistance patterns—into account when considering antibiotic coverage.

When confronted by the rare case of periorbital cellulitis, I always consider risk factors that may change my management, such as immunization status and asplenia. Also, meningitis is a rare but serious complication, so I also keep meningitis risk factors in mind, such as immunosuppression, coincident trauma, or a poor response to initial medical therapy.

Finally, any question of orbital involvement should prompt an emergent consultation.

Evidence summary

Orbital cellulitis is a serious soft-tissue infection of childhood with very different etiologies.

  • Periorbital (or preseptal) cellulitis is synonymous with stage I orbital cellulitis, in which there is induration, erythema, warmth, and tenderness of the periorbital soft tissues, usually secondary to external inoculation, but the inflammation does not extend into the bony orbit.
  • Stages II, III, and IV orbital cellulitis are progressively more invasive infections that generally arise from the sinuses; they may involve the retro-orbital area. These stages of orbital cellulitis can cause proptosis, decrease visual acuity, or appear as abscesses on computed tomography scan.1,2

Staged treatment

Many retrospective studies of stage II–IV orbital cellulitis with relatively few subjects and small prospective case series have been published with common themes for management recommendations:

  • early intravenous antibiotics (likely for an inpatient), and
  • involvement of otolaryngology and ophthalmology specialists.

No head-to-head trials have been completed to evaluate efficacy of specific antimicrobial regimens.

Oral antibiotics. First, treat stage I orbital cellulitis with oral antibiotics.

IV antibiotics. Modify treatment to intravenous antibiotics when there is no improvement within 24 hours or if you discover any characteristic of more severe orbital cellulitis.

Medical management of stage II–IV orbital cellulitis with intravenous antibiotics is the current standard of care until it is clear that one of the following is present:

  • no improvement by 24 to 48 hours
  • visual impairment
  • complete ophthalmoplegia, or
  • well-defined periosteal abscess.1,2
 

Surgery. For refractory cases, surgical decompression will likely be required.

The evidence. A small case series (n=9) found 21 children admitted to hospital for preseptal cellulitis, of whom 4 later were diagnosed with orbital cellulitis. There was a total of 9 cases of orbital cellulitis; however, only 1 required operative management of orbital cellulitis.3 In a prospective study to evaluate medical management (n=23), 87% of patients responded to intravenous antibiotics.4 No statistically significant long-term difference in subperiosteal abscesses (as a complication of orbital cellulitis) was found in another retrospective study comparing medical to surgical management.5

Target the likely pathogens

Direct antimicrobial therapy toward common pathogens for likely sources of infection, paying attention to local resistance patterns and the pathogens usually associated with sinusitis (TABLE).1,2,6-8

A retrospective case series of 94 patients of all ages in China implicated Staphylococcus aureus and streptococcal species based on cultures taken from eye purulence and abscesses.6 Another retrospective case series from Vanderbilt (n=80) found streptococci as the most common cause, based on blood and wound cultures in the Hib vaccination era; however, only 12 wounds returned positive cultures.7

TABLE
Choose antibiotic based on cause and likely pathogen1,2,6-8

ANTECEDENT EVENTLIKELY PATHOGENSBEST DRUGS
Acute sinusitisStreptococcus pneumoniae
Haemophilus influenzae
Moraxella catarrhalis
Penicillinase-resistant penicillins
TraumaStaphylococcus aureus
Group A β-hernolytic streptococci
Increasing concern for methicillin-resistant S aureus
Penicillinase-resistant penicillins
First-generation cephalosporins
Consider drugs appropriate for methicillin-resistant S aureus
Chronic sinusitisAnaerobesMetronidazole Clindamycin
 

 

 

Steroids have no proven benefit

Systemic steroids have no proven benefit in the treatment of pediatric orbital cellulitis with subperiosteal abscess.

A small retrospective cohort study of the benefit of intravenous steroids in addition to antibiotics showed no decrease in hospital stay or need for surgical decompression (n=23, P=.26 and .20, respectively).9 Without prospective data and a power analysis, lack of benefit of steroids cannot be definitively shown.

Recommendations from others

Infectious Disease Society of America. The guidelines for the management of skin and soft-tissue infections implicate β-hemolytic streptococci as the most common cellulitis pathogen, but also recommend empiric coverage against S aureus.

Periorbital and orbital cellulitis are not specifically addressed in these guidelines, but oral dicloxacillin, cephalexin, clindamycin, or erythromycin are recommended for superficial cellulitis, provided there is no known resistance to these antibiotics.

Intravenous penicillinase-resistant penicillins (nafcillin) or a first-generation cephalosporin (cefazolin) may be used for more severe infections.

For penicillin-allergic patients, the IDSA recommends clindamycin or vancomycin.10

Sanford Guide to Antimicrobial Therapy. Nafcillin plus ceftriaxone and metronidazole is the recommended treatment for orbital cellulitis.

For patients allergic to penicillin, vancomycin plus levofloxacin and metronidazole are recommended.8

Neither the American Academy of Ophthalmology nor the International Council of Ophthalmology offers clinical statements on orbital cellulitis.

Acknowledgments

The opinions and assertions contained herein are the private views of the author and not to be construed as official, or as reflecting the views of the US Air Force Medical service or the US Air Force at large.

References

1. Nageswaran S, Woods CR, Benjamin DK, Jr, Givner LB, Shetty AK. Orbital cellulitis in children. Pediatr Infect Dis J 2006;25:695-699.

2. Vayalumkal JV, Jadavji T. Children hospitalized with skin and soft tissue infections: a guide to antibacterial selection and treatment. Paediatr Drugs 2006;8:99-111.

3. Starkey CR, Steele RW. Medical management of orbital cellulitis. Pediatr Infect Dis J 2001;20:1002-1005.

4. Noel LP, Clarke WN, MacDonald N. Clinical management of orbital cellulitis in children. Can J Ophthalmol 1990;25:11-16.

5. Greenberg MF, Pollard ZF. Medical treatment of pediatric subperiosteal orbital abscess secondary to sinusitis. J AAPOS 1998;2:351-355.

6. Liu IT, Kao SC, Wang AG, Tsai CC, Liang CK, Hsu WM. Preseptal and orbital cellulitis: a 10-year review of hospitalized patients. J Chin Med Assoc 2006;69:415-422.

7. Donohue SP, Schwartz G. Preseptal and orbital cellulitis in childhood. A changing microbiologic spectrum. Ophthalmology 1998;105:1902;1905.

8. Gilbert DM, Eliopoulos GM, Moellering RC, Sande MA. The Sanford Guide to Antimicrobial Therapy 2006 36th ed. Sperryville, Va: Antimicrobial Therapy; 2006;12.-

9. Yen MT, Yen KG. Effect of corticosteroids in the acute management of pediatric orbital cellulitis with subperiosteal abscess. Ophthal Plast Reconstr Surg 2005;21:363-366.

10. Stevens DL, Bisno AL, Chambers HF, et al. Infectious Diseases Society of America. Practice guidelines for the diagnosis and management of skin and soft-tissue infections. Clin Infect Dis 2005;41:1373-1406.

Article PDF
Author and Disclosure Information

Sarah Hennemann, MD
Paul Crawford, MD
Eglin Air Force Base Family Medicine Residency, Eglin Air Force Base, Fla

Loan Nguyen, MLS
Baylor College of Medicine, Houston, Tex

Issue
The Journal of Family Practice - 56(8)
Publications
Topics
Page Number
662-663
Legacy Keywords
orbital; cellulitis; pediatrics; children; eye; antibiotics; Sarah Hennemann MD; Paul Crawford MD; Loan Nguyen MLS; Peter C. Smith MD
Sections
Author and Disclosure Information

Sarah Hennemann, MD
Paul Crawford, MD
Eglin Air Force Base Family Medicine Residency, Eglin Air Force Base, Fla

Loan Nguyen, MLS
Baylor College of Medicine, Houston, Tex

Author and Disclosure Information

Sarah Hennemann, MD
Paul Crawford, MD
Eglin Air Force Base Family Medicine Residency, Eglin Air Force Base, Fla

Loan Nguyen, MLS
Baylor College of Medicine, Houston, Tex

Article PDF
Article PDF
EVIDENCE-BASED ANSWER

Although antibiotics are the best initial treatment, surgical intervention is warranted when a child has:

  • visual impairment, complete ophthalmoplegia, or well-defined abscess on presentation, or
  • no clearly apparent clinical improvement by 24 hours (strength of recommendation [SOR]: C, based on patient-oriented case-series studies).

Target antimicrobial therapy toward the common pathogens associated with predisposing factors for orbital cellulitis, such as sinusitis—and pay attention to local resistance patterns (SOR: C, based on patient-oriented case series).

Clinical commentary

Rare but serious risk factors
Peter C. Smith, MD
Rose Family Medicine Residency, University of Colorado Health Sciences Center, Denver

The incidence of Haemophilus influenzae–related periorbital cellulitis appears to have plummeted with the advent of Hib vaccine. And while no national data have been published, case series support my clinical observation that the overall incidence of periorbital cellulitis has dropped as well.

The arrival of heptavalent pneumococcal vaccine may further contribute to its welcome scarcity. take this changing bacteriology—in conjunction with local resistance patterns—into account when considering antibiotic coverage.

When confronted by the rare case of periorbital cellulitis, I always consider risk factors that may change my management, such as immunization status and asplenia. Also, meningitis is a rare but serious complication, so I also keep meningitis risk factors in mind, such as immunosuppression, coincident trauma, or a poor response to initial medical therapy.

Finally, any question of orbital involvement should prompt an emergent consultation.

Evidence summary

Orbital cellulitis is a serious soft-tissue infection of childhood with very different etiologies.

  • Periorbital (or preseptal) cellulitis is synonymous with stage I orbital cellulitis, in which there is induration, erythema, warmth, and tenderness of the periorbital soft tissues, usually secondary to external inoculation, but the inflammation does not extend into the bony orbit.
  • Stages II, III, and IV orbital cellulitis are progressively more invasive infections that generally arise from the sinuses; they may involve the retro-orbital area. These stages of orbital cellulitis can cause proptosis, decrease visual acuity, or appear as abscesses on computed tomography scan.1,2

Staged treatment

Many retrospective studies of stage II–IV orbital cellulitis with relatively few subjects and small prospective case series have been published with common themes for management recommendations:

  • early intravenous antibiotics (likely for an inpatient), and
  • involvement of otolaryngology and ophthalmology specialists.

No head-to-head trials have been completed to evaluate efficacy of specific antimicrobial regimens.

Oral antibiotics. First, treat stage I orbital cellulitis with oral antibiotics.

IV antibiotics. Modify treatment to intravenous antibiotics when there is no improvement within 24 hours or if you discover any characteristic of more severe orbital cellulitis.

Medical management of stage II–IV orbital cellulitis with intravenous antibiotics is the current standard of care until it is clear that one of the following is present:

  • no improvement by 24 to 48 hours
  • visual impairment
  • complete ophthalmoplegia, or
  • well-defined periosteal abscess.1,2
 

Surgery. For refractory cases, surgical decompression will likely be required.

The evidence. A small case series (n=9) found 21 children admitted to hospital for preseptal cellulitis, of whom 4 later were diagnosed with orbital cellulitis. There was a total of 9 cases of orbital cellulitis; however, only 1 required operative management of orbital cellulitis.3 In a prospective study to evaluate medical management (n=23), 87% of patients responded to intravenous antibiotics.4 No statistically significant long-term difference in subperiosteal abscesses (as a complication of orbital cellulitis) was found in another retrospective study comparing medical to surgical management.5

Target the likely pathogens

Direct antimicrobial therapy toward common pathogens for likely sources of infection, paying attention to local resistance patterns and the pathogens usually associated with sinusitis (TABLE).1,2,6-8

A retrospective case series of 94 patients of all ages in China implicated Staphylococcus aureus and streptococcal species based on cultures taken from eye purulence and abscesses.6 Another retrospective case series from Vanderbilt (n=80) found streptococci as the most common cause, based on blood and wound cultures in the Hib vaccination era; however, only 12 wounds returned positive cultures.7

TABLE
Choose antibiotic based on cause and likely pathogen1,2,6-8

ANTECEDENT EVENTLIKELY PATHOGENSBEST DRUGS
Acute sinusitisStreptococcus pneumoniae
Haemophilus influenzae
Moraxella catarrhalis
Penicillinase-resistant penicillins
TraumaStaphylococcus aureus
Group A β-hernolytic streptococci
Increasing concern for methicillin-resistant S aureus
Penicillinase-resistant penicillins
First-generation cephalosporins
Consider drugs appropriate for methicillin-resistant S aureus
Chronic sinusitisAnaerobesMetronidazole Clindamycin
 

 

 

Steroids have no proven benefit

Systemic steroids have no proven benefit in the treatment of pediatric orbital cellulitis with subperiosteal abscess.

A small retrospective cohort study of the benefit of intravenous steroids in addition to antibiotics showed no decrease in hospital stay or need for surgical decompression (n=23, P=.26 and .20, respectively).9 Without prospective data and a power analysis, lack of benefit of steroids cannot be definitively shown.

Recommendations from others

Infectious Disease Society of America. The guidelines for the management of skin and soft-tissue infections implicate β-hemolytic streptococci as the most common cellulitis pathogen, but also recommend empiric coverage against S aureus.

Periorbital and orbital cellulitis are not specifically addressed in these guidelines, but oral dicloxacillin, cephalexin, clindamycin, or erythromycin are recommended for superficial cellulitis, provided there is no known resistance to these antibiotics.

Intravenous penicillinase-resistant penicillins (nafcillin) or a first-generation cephalosporin (cefazolin) may be used for more severe infections.

For penicillin-allergic patients, the IDSA recommends clindamycin or vancomycin.10

Sanford Guide to Antimicrobial Therapy. Nafcillin plus ceftriaxone and metronidazole is the recommended treatment for orbital cellulitis.

For patients allergic to penicillin, vancomycin plus levofloxacin and metronidazole are recommended.8

Neither the American Academy of Ophthalmology nor the International Council of Ophthalmology offers clinical statements on orbital cellulitis.

Acknowledgments

The opinions and assertions contained herein are the private views of the author and not to be construed as official, or as reflecting the views of the US Air Force Medical service or the US Air Force at large.

EVIDENCE-BASED ANSWER

Although antibiotics are the best initial treatment, surgical intervention is warranted when a child has:

  • visual impairment, complete ophthalmoplegia, or well-defined abscess on presentation, or
  • no clearly apparent clinical improvement by 24 hours (strength of recommendation [SOR]: C, based on patient-oriented case-series studies).

Target antimicrobial therapy toward the common pathogens associated with predisposing factors for orbital cellulitis, such as sinusitis—and pay attention to local resistance patterns (SOR: C, based on patient-oriented case series).

Clinical commentary

Rare but serious risk factors
Peter C. Smith, MD
Rose Family Medicine Residency, University of Colorado Health Sciences Center, Denver

The incidence of Haemophilus influenzae–related periorbital cellulitis appears to have plummeted with the advent of Hib vaccine. And while no national data have been published, case series support my clinical observation that the overall incidence of periorbital cellulitis has dropped as well.

The arrival of heptavalent pneumococcal vaccine may further contribute to its welcome scarcity. take this changing bacteriology—in conjunction with local resistance patterns—into account when considering antibiotic coverage.

When confronted by the rare case of periorbital cellulitis, I always consider risk factors that may change my management, such as immunization status and asplenia. Also, meningitis is a rare but serious complication, so I also keep meningitis risk factors in mind, such as immunosuppression, coincident trauma, or a poor response to initial medical therapy.

Finally, any question of orbital involvement should prompt an emergent consultation.

Evidence summary

Orbital cellulitis is a serious soft-tissue infection of childhood with very different etiologies.

  • Periorbital (or preseptal) cellulitis is synonymous with stage I orbital cellulitis, in which there is induration, erythema, warmth, and tenderness of the periorbital soft tissues, usually secondary to external inoculation, but the inflammation does not extend into the bony orbit.
  • Stages II, III, and IV orbital cellulitis are progressively more invasive infections that generally arise from the sinuses; they may involve the retro-orbital area. These stages of orbital cellulitis can cause proptosis, decrease visual acuity, or appear as abscesses on computed tomography scan.1,2

Staged treatment

Many retrospective studies of stage II–IV orbital cellulitis with relatively few subjects and small prospective case series have been published with common themes for management recommendations:

  • early intravenous antibiotics (likely for an inpatient), and
  • involvement of otolaryngology and ophthalmology specialists.

No head-to-head trials have been completed to evaluate efficacy of specific antimicrobial regimens.

Oral antibiotics. First, treat stage I orbital cellulitis with oral antibiotics.

IV antibiotics. Modify treatment to intravenous antibiotics when there is no improvement within 24 hours or if you discover any characteristic of more severe orbital cellulitis.

Medical management of stage II–IV orbital cellulitis with intravenous antibiotics is the current standard of care until it is clear that one of the following is present:

  • no improvement by 24 to 48 hours
  • visual impairment
  • complete ophthalmoplegia, or
  • well-defined periosteal abscess.1,2
 

Surgery. For refractory cases, surgical decompression will likely be required.

The evidence. A small case series (n=9) found 21 children admitted to hospital for preseptal cellulitis, of whom 4 later were diagnosed with orbital cellulitis. There was a total of 9 cases of orbital cellulitis; however, only 1 required operative management of orbital cellulitis.3 In a prospective study to evaluate medical management (n=23), 87% of patients responded to intravenous antibiotics.4 No statistically significant long-term difference in subperiosteal abscesses (as a complication of orbital cellulitis) was found in another retrospective study comparing medical to surgical management.5

Target the likely pathogens

Direct antimicrobial therapy toward common pathogens for likely sources of infection, paying attention to local resistance patterns and the pathogens usually associated with sinusitis (TABLE).1,2,6-8

A retrospective case series of 94 patients of all ages in China implicated Staphylococcus aureus and streptococcal species based on cultures taken from eye purulence and abscesses.6 Another retrospective case series from Vanderbilt (n=80) found streptococci as the most common cause, based on blood and wound cultures in the Hib vaccination era; however, only 12 wounds returned positive cultures.7

TABLE
Choose antibiotic based on cause and likely pathogen1,2,6-8

ANTECEDENT EVENTLIKELY PATHOGENSBEST DRUGS
Acute sinusitisStreptococcus pneumoniae
Haemophilus influenzae
Moraxella catarrhalis
Penicillinase-resistant penicillins
TraumaStaphylococcus aureus
Group A β-hernolytic streptococci
Increasing concern for methicillin-resistant S aureus
Penicillinase-resistant penicillins
First-generation cephalosporins
Consider drugs appropriate for methicillin-resistant S aureus
Chronic sinusitisAnaerobesMetronidazole Clindamycin
 

 

 

Steroids have no proven benefit

Systemic steroids have no proven benefit in the treatment of pediatric orbital cellulitis with subperiosteal abscess.

A small retrospective cohort study of the benefit of intravenous steroids in addition to antibiotics showed no decrease in hospital stay or need for surgical decompression (n=23, P=.26 and .20, respectively).9 Without prospective data and a power analysis, lack of benefit of steroids cannot be definitively shown.

Recommendations from others

Infectious Disease Society of America. The guidelines for the management of skin and soft-tissue infections implicate β-hemolytic streptococci as the most common cellulitis pathogen, but also recommend empiric coverage against S aureus.

Periorbital and orbital cellulitis are not specifically addressed in these guidelines, but oral dicloxacillin, cephalexin, clindamycin, or erythromycin are recommended for superficial cellulitis, provided there is no known resistance to these antibiotics.

Intravenous penicillinase-resistant penicillins (nafcillin) or a first-generation cephalosporin (cefazolin) may be used for more severe infections.

For penicillin-allergic patients, the IDSA recommends clindamycin or vancomycin.10

Sanford Guide to Antimicrobial Therapy. Nafcillin plus ceftriaxone and metronidazole is the recommended treatment for orbital cellulitis.

For patients allergic to penicillin, vancomycin plus levofloxacin and metronidazole are recommended.8

Neither the American Academy of Ophthalmology nor the International Council of Ophthalmology offers clinical statements on orbital cellulitis.

Acknowledgments

The opinions and assertions contained herein are the private views of the author and not to be construed as official, or as reflecting the views of the US Air Force Medical service or the US Air Force at large.

References

1. Nageswaran S, Woods CR, Benjamin DK, Jr, Givner LB, Shetty AK. Orbital cellulitis in children. Pediatr Infect Dis J 2006;25:695-699.

2. Vayalumkal JV, Jadavji T. Children hospitalized with skin and soft tissue infections: a guide to antibacterial selection and treatment. Paediatr Drugs 2006;8:99-111.

3. Starkey CR, Steele RW. Medical management of orbital cellulitis. Pediatr Infect Dis J 2001;20:1002-1005.

4. Noel LP, Clarke WN, MacDonald N. Clinical management of orbital cellulitis in children. Can J Ophthalmol 1990;25:11-16.

5. Greenberg MF, Pollard ZF. Medical treatment of pediatric subperiosteal orbital abscess secondary to sinusitis. J AAPOS 1998;2:351-355.

6. Liu IT, Kao SC, Wang AG, Tsai CC, Liang CK, Hsu WM. Preseptal and orbital cellulitis: a 10-year review of hospitalized patients. J Chin Med Assoc 2006;69:415-422.

7. Donohue SP, Schwartz G. Preseptal and orbital cellulitis in childhood. A changing microbiologic spectrum. Ophthalmology 1998;105:1902;1905.

8. Gilbert DM, Eliopoulos GM, Moellering RC, Sande MA. The Sanford Guide to Antimicrobial Therapy 2006 36th ed. Sperryville, Va: Antimicrobial Therapy; 2006;12.-

9. Yen MT, Yen KG. Effect of corticosteroids in the acute management of pediatric orbital cellulitis with subperiosteal abscess. Ophthal Plast Reconstr Surg 2005;21:363-366.

10. Stevens DL, Bisno AL, Chambers HF, et al. Infectious Diseases Society of America. Practice guidelines for the diagnosis and management of skin and soft-tissue infections. Clin Infect Dis 2005;41:1373-1406.

References

1. Nageswaran S, Woods CR, Benjamin DK, Jr, Givner LB, Shetty AK. Orbital cellulitis in children. Pediatr Infect Dis J 2006;25:695-699.

2. Vayalumkal JV, Jadavji T. Children hospitalized with skin and soft tissue infections: a guide to antibacterial selection and treatment. Paediatr Drugs 2006;8:99-111.

3. Starkey CR, Steele RW. Medical management of orbital cellulitis. Pediatr Infect Dis J 2001;20:1002-1005.

4. Noel LP, Clarke WN, MacDonald N. Clinical management of orbital cellulitis in children. Can J Ophthalmol 1990;25:11-16.

5. Greenberg MF, Pollard ZF. Medical treatment of pediatric subperiosteal orbital abscess secondary to sinusitis. J AAPOS 1998;2:351-355.

6. Liu IT, Kao SC, Wang AG, Tsai CC, Liang CK, Hsu WM. Preseptal and orbital cellulitis: a 10-year review of hospitalized patients. J Chin Med Assoc 2006;69:415-422.

7. Donohue SP, Schwartz G. Preseptal and orbital cellulitis in childhood. A changing microbiologic spectrum. Ophthalmology 1998;105:1902;1905.

8. Gilbert DM, Eliopoulos GM, Moellering RC, Sande MA. The Sanford Guide to Antimicrobial Therapy 2006 36th ed. Sperryville, Va: Antimicrobial Therapy; 2006;12.-

9. Yen MT, Yen KG. Effect of corticosteroids in the acute management of pediatric orbital cellulitis with subperiosteal abscess. Ophthal Plast Reconstr Surg 2005;21:363-366.

10. Stevens DL, Bisno AL, Chambers HF, et al. Infectious Diseases Society of America. Practice guidelines for the diagnosis and management of skin and soft-tissue infections. Clin Infect Dis 2005;41:1373-1406.

Issue
The Journal of Family Practice - 56(8)
Issue
The Journal of Family Practice - 56(8)
Page Number
662-663
Page Number
662-663
Publications
Publications
Topics
Article Type
Display Headline
What is the best initial treatment for orbital cellulitis in children?
Display Headline
What is the best initial treatment for orbital cellulitis in children?
Legacy Keywords
orbital; cellulitis; pediatrics; children; eye; antibiotics; Sarah Hennemann MD; Paul Crawford MD; Loan Nguyen MLS; Peter C. Smith MD
Legacy Keywords
orbital; cellulitis; pediatrics; children; eye; antibiotics; Sarah Hennemann MD; Paul Crawford MD; Loan Nguyen MLS; Peter C. Smith MD
Sections
PURLs Copyright

Evidence-based answers from the Family Physicians Inquiries Network

Disallow All Ads
Article PDF Media

What treatment approach to intrapartum maternal fever has the best fetal outcomes?

Article Type
Changed
Mon, 01/14/2019 - 11:19
Display Headline
What treatment approach to intrapartum maternal fever has the best fetal outcomes?
EVIDENCE-BASED ANSWER

A combination of beta-lactam and aminoglycoside antibiotics are the recommended empiric agents for the treatment of acute chorioamnionitis, given that no head-to-head trials exist (strength of recommendation [SOR]: C, based on expert opinion). Intrapartum antibiotic treatment is not superior to postpartum antibiotics for reducing neonatal sepsis and pneumonia (SOR: C, based on patient-oriented, underpowered randomized trials).

Clinical commentary

Carefully follow laboring patients with fever for other signs of chorioamnionitis
Jon O. Neher, MD
Valley Family Medicine, Renton, Wash

The data on the best antibiotic treatment of clinical chorioamnionitis remains as slim as ever, it appears. But since experts continue to recommend potentially toxic gentamicin as part of therapy, you should carefully monitor laboring patients at term who develop a fever for the development of other diagnostic signs of chorioamnionitis. While maternal and fetal tachycardia are frequently caused by conditions other than infection, their appearance in a febrile gravida should prompt full chorioamnionitis therapy (even in patients already on empiric antibiotics for group B streptococci). With epidural anesthesia, uterine tenderness is an unreliable sign of infection. Purulent amniotic fluid is a late sign and rarely contributes clinically.

 

Evidence summary

Acute chorioamnionitis (or intra-amniotic infection) poses a high risk of maternal and neonatal morbidity. Neonatal sepsis or pneumonia occurs in up to 24% of infants born to mothers with chorioamnionitis;1 1% to 2% of pregnancies complicated by chorioamnionitis end in neonatal death.1,2

Acute chorioamnionitis is defined as intrapartum maternal fever and maternal tachycardia, fetal tachycardia, uterine tenderness, or purulent amniotic fluid.1,3 Antibiotic treatment of acute chorioamnionitis is widely accepted, yet in vivo studies to determine the most effective empiric antibiotic regimens are lacking.

Intrapartum antibiotics probably reduce sepsis

Although few well-designed trials stand out, a Cochrane review4 summarizing 2 relevant studies is available. Gibbs et al3 performed an underpowered, randomized comparative trial of intrapartum vs postpartum treatment of chorioamnionitis, with both groups (45 patients total) receiving ampicillin 2 g IV every 6 hours plus gentamicin 1.5 mg/kg IV every 8 hours.3 Those women who underwent cesarean section also received clindamycin 900 mg IV every 8 hours starting at cord clamping. In this study, investigators reported neonatal sepsis was significantly reduced with intrapartum treatment (0 vs 21%; P=.03, number needed to treat=4.8), as were neonatal hospital stays (3.8 vs 5.7 days; P=.02), regardless of delivery method. The study had been planned for 92 patients; it was stopped early (n=48) after an interim analysis.

Because of the small sample size, other findings from the study must be viewed with caution. Intrapartum treatment with antibiotics was associated with a “significant” clinical reduction in neonatal sepsis (relative risk [RR]=0.08; 95% confidence interval [CI], 0.00–1.44) and pneumonia (RR=0.15; 95% CI, 0.01–2.92) compared with treatment given immediately postpartum; however, neither value was truly statistically significant according to the Cochrane review.4

The research suggests a potential benefit to adding clindamycin to ampicillin and gentamicin. In an effort to test this, 1 study randomized 133 women into 2 arms—treatment with ampicillin, gentamicin, and clindamycin compared with ampicillin and gentamicin alone—and found no additional benefit in regards to neonatal sepsis (RR=2.16; 95% CI, 0.20–23.21) or neonatal death (RR=0.72; 95% CI, 0.12–4.16).1 There was a trend towards a decrease in the incidence of postpartum endometritis in women who received ampicillin, gentamicin, and clindamycin, but this did not reach statistical significance (RR=0.54; 95% CI, 0.19–1.49).4

 

 

 

Recommendations from others

A 2002 bulletin from American College of Obstetricians and Gynecologists (ACOG) and the American Academy of Pediatrics5 recommended the combination of ampicillin 2 gm IV every 4 to 6 hours or penicillin 5 million units IV every 4 to 6 hours, plus an aminoglycoside (such as gentamicin 1.5 mg/kg IV every 8 hours), since this regimen provides appropriate coverage for typical organisms associated with acute chorioamnionitis. At the time the bulletin was published, the use of single daily dosing of aminoglycoside did not have sufficient studies to back its use. In addition, ACOG recommends adding clindamycin, metronidazole, or an extended-spectrum third-generation cephalosporin to the treatment regimen if cesarean section is required, to provide coverage for anaerobic organisms. They recommend clindamycin 900 mg IV every 8 hours to replace amoxicillin in penicillin-allergic patients. The Nottingham Guideline Development Group recommends amoxicillin 2 gm IV initially then 1 gm every 8 hours, and in place of gentamicin, recommends metronidazole 500 mg IV, every 8 hours (or 1 gm PR twice a day).6 Both recommendations suggest clindamycin 900 mg IV every 8 hours to replace amoxicillin in penicillin-allergic patients. For patients with nonanaphylactic reactions to penicillin, they recommend cefotaxime 1 g IV every 8 hours.

Acknowledgments

The opinions and assertions contained herein are the private views of the author and not to be construed as official, or as reflecting the views of the US Air Force Medical Service or the US Air Force at large.

References

1. Maberry MC, Gilstrap LC, 3rd. Intrapartum antibiotic therapy for suspected intraamniotic infection: impact on the fetus and neonate. Clin Obstet Gyn 1991;34:345-351.

2. Hauth JC, Gilstrap LC, Hankins GD, Conner KD. Term maternal and neonatal complications of acute chorioamnionitis. Obstet Gyn 1985;66:59-62.

3. Gibbs RS, Dinsmoor MJ, Newton ER, et al. A randomized trial of intrapartum versus immediate postpartum treatment of women with intra-amniotic infection. Obstet Gyn 1988;72:823-828.

4. Hopkins L, Smaill F. Antibiotic regimens for management of intraamniotic infection. Cochrane Database Syst Rev 2002;(3):CD003254.-

5. American College of Obstetricians and Gynecologists, American Academy of Pediatrics. Guidelines for Perinatal Care. 5th ed. Washington, DC: ACOG;2002:165-166.

6. Hayman R, Kean L. Guidelines for the Prevention of Neonatal Group B Streptococcal Infection. Nottingham: Nottingham City Hospital, National Health Service; 2002. Revised 2005. Available at: www.nuh.nhs.uk/nch/antibiotics. Accessed on March 30, 2007.

Article PDF
Author and Disclosure Information

Matthew Snyder, DO
Paul Crawford, MD
Eglin Air Force Base Family Medicine Residency, Eglin AFB, Fla

Barbara Jamieson, MLS
Medical College of Wisconsin, Milwaukee

Issue
The Journal of Family Practice - 56(5)
Publications
Topics
Page Number
401-402
Legacy Keywords
fever; women’s health; pregnancy; birth; intrapartum; intrapartum fever; neonatal; obstetric
Sections
Author and Disclosure Information

Matthew Snyder, DO
Paul Crawford, MD
Eglin Air Force Base Family Medicine Residency, Eglin AFB, Fla

Barbara Jamieson, MLS
Medical College of Wisconsin, Milwaukee

Author and Disclosure Information

Matthew Snyder, DO
Paul Crawford, MD
Eglin Air Force Base Family Medicine Residency, Eglin AFB, Fla

Barbara Jamieson, MLS
Medical College of Wisconsin, Milwaukee

Article PDF
Article PDF
EVIDENCE-BASED ANSWER

A combination of beta-lactam and aminoglycoside antibiotics are the recommended empiric agents for the treatment of acute chorioamnionitis, given that no head-to-head trials exist (strength of recommendation [SOR]: C, based on expert opinion). Intrapartum antibiotic treatment is not superior to postpartum antibiotics for reducing neonatal sepsis and pneumonia (SOR: C, based on patient-oriented, underpowered randomized trials).

Clinical commentary

Carefully follow laboring patients with fever for other signs of chorioamnionitis
Jon O. Neher, MD
Valley Family Medicine, Renton, Wash

The data on the best antibiotic treatment of clinical chorioamnionitis remains as slim as ever, it appears. But since experts continue to recommend potentially toxic gentamicin as part of therapy, you should carefully monitor laboring patients at term who develop a fever for the development of other diagnostic signs of chorioamnionitis. While maternal and fetal tachycardia are frequently caused by conditions other than infection, their appearance in a febrile gravida should prompt full chorioamnionitis therapy (even in patients already on empiric antibiotics for group B streptococci). With epidural anesthesia, uterine tenderness is an unreliable sign of infection. Purulent amniotic fluid is a late sign and rarely contributes clinically.

 

Evidence summary

Acute chorioamnionitis (or intra-amniotic infection) poses a high risk of maternal and neonatal morbidity. Neonatal sepsis or pneumonia occurs in up to 24% of infants born to mothers with chorioamnionitis;1 1% to 2% of pregnancies complicated by chorioamnionitis end in neonatal death.1,2

Acute chorioamnionitis is defined as intrapartum maternal fever and maternal tachycardia, fetal tachycardia, uterine tenderness, or purulent amniotic fluid.1,3 Antibiotic treatment of acute chorioamnionitis is widely accepted, yet in vivo studies to determine the most effective empiric antibiotic regimens are lacking.

Intrapartum antibiotics probably reduce sepsis

Although few well-designed trials stand out, a Cochrane review4 summarizing 2 relevant studies is available. Gibbs et al3 performed an underpowered, randomized comparative trial of intrapartum vs postpartum treatment of chorioamnionitis, with both groups (45 patients total) receiving ampicillin 2 g IV every 6 hours plus gentamicin 1.5 mg/kg IV every 8 hours.3 Those women who underwent cesarean section also received clindamycin 900 mg IV every 8 hours starting at cord clamping. In this study, investigators reported neonatal sepsis was significantly reduced with intrapartum treatment (0 vs 21%; P=.03, number needed to treat=4.8), as were neonatal hospital stays (3.8 vs 5.7 days; P=.02), regardless of delivery method. The study had been planned for 92 patients; it was stopped early (n=48) after an interim analysis.

Because of the small sample size, other findings from the study must be viewed with caution. Intrapartum treatment with antibiotics was associated with a “significant” clinical reduction in neonatal sepsis (relative risk [RR]=0.08; 95% confidence interval [CI], 0.00–1.44) and pneumonia (RR=0.15; 95% CI, 0.01–2.92) compared with treatment given immediately postpartum; however, neither value was truly statistically significant according to the Cochrane review.4

The research suggests a potential benefit to adding clindamycin to ampicillin and gentamicin. In an effort to test this, 1 study randomized 133 women into 2 arms—treatment with ampicillin, gentamicin, and clindamycin compared with ampicillin and gentamicin alone—and found no additional benefit in regards to neonatal sepsis (RR=2.16; 95% CI, 0.20–23.21) or neonatal death (RR=0.72; 95% CI, 0.12–4.16).1 There was a trend towards a decrease in the incidence of postpartum endometritis in women who received ampicillin, gentamicin, and clindamycin, but this did not reach statistical significance (RR=0.54; 95% CI, 0.19–1.49).4

 

 

 

Recommendations from others

A 2002 bulletin from American College of Obstetricians and Gynecologists (ACOG) and the American Academy of Pediatrics5 recommended the combination of ampicillin 2 gm IV every 4 to 6 hours or penicillin 5 million units IV every 4 to 6 hours, plus an aminoglycoside (such as gentamicin 1.5 mg/kg IV every 8 hours), since this regimen provides appropriate coverage for typical organisms associated with acute chorioamnionitis. At the time the bulletin was published, the use of single daily dosing of aminoglycoside did not have sufficient studies to back its use. In addition, ACOG recommends adding clindamycin, metronidazole, or an extended-spectrum third-generation cephalosporin to the treatment regimen if cesarean section is required, to provide coverage for anaerobic organisms. They recommend clindamycin 900 mg IV every 8 hours to replace amoxicillin in penicillin-allergic patients. The Nottingham Guideline Development Group recommends amoxicillin 2 gm IV initially then 1 gm every 8 hours, and in place of gentamicin, recommends metronidazole 500 mg IV, every 8 hours (or 1 gm PR twice a day).6 Both recommendations suggest clindamycin 900 mg IV every 8 hours to replace amoxicillin in penicillin-allergic patients. For patients with nonanaphylactic reactions to penicillin, they recommend cefotaxime 1 g IV every 8 hours.

Acknowledgments

The opinions and assertions contained herein are the private views of the author and not to be construed as official, or as reflecting the views of the US Air Force Medical Service or the US Air Force at large.

EVIDENCE-BASED ANSWER

A combination of beta-lactam and aminoglycoside antibiotics are the recommended empiric agents for the treatment of acute chorioamnionitis, given that no head-to-head trials exist (strength of recommendation [SOR]: C, based on expert opinion). Intrapartum antibiotic treatment is not superior to postpartum antibiotics for reducing neonatal sepsis and pneumonia (SOR: C, based on patient-oriented, underpowered randomized trials).

Clinical commentary

Carefully follow laboring patients with fever for other signs of chorioamnionitis
Jon O. Neher, MD
Valley Family Medicine, Renton, Wash

The data on the best antibiotic treatment of clinical chorioamnionitis remains as slim as ever, it appears. But since experts continue to recommend potentially toxic gentamicin as part of therapy, you should carefully monitor laboring patients at term who develop a fever for the development of other diagnostic signs of chorioamnionitis. While maternal and fetal tachycardia are frequently caused by conditions other than infection, their appearance in a febrile gravida should prompt full chorioamnionitis therapy (even in patients already on empiric antibiotics for group B streptococci). With epidural anesthesia, uterine tenderness is an unreliable sign of infection. Purulent amniotic fluid is a late sign and rarely contributes clinically.

 

Evidence summary

Acute chorioamnionitis (or intra-amniotic infection) poses a high risk of maternal and neonatal morbidity. Neonatal sepsis or pneumonia occurs in up to 24% of infants born to mothers with chorioamnionitis;1 1% to 2% of pregnancies complicated by chorioamnionitis end in neonatal death.1,2

Acute chorioamnionitis is defined as intrapartum maternal fever and maternal tachycardia, fetal tachycardia, uterine tenderness, or purulent amniotic fluid.1,3 Antibiotic treatment of acute chorioamnionitis is widely accepted, yet in vivo studies to determine the most effective empiric antibiotic regimens are lacking.

Intrapartum antibiotics probably reduce sepsis

Although few well-designed trials stand out, a Cochrane review4 summarizing 2 relevant studies is available. Gibbs et al3 performed an underpowered, randomized comparative trial of intrapartum vs postpartum treatment of chorioamnionitis, with both groups (45 patients total) receiving ampicillin 2 g IV every 6 hours plus gentamicin 1.5 mg/kg IV every 8 hours.3 Those women who underwent cesarean section also received clindamycin 900 mg IV every 8 hours starting at cord clamping. In this study, investigators reported neonatal sepsis was significantly reduced with intrapartum treatment (0 vs 21%; P=.03, number needed to treat=4.8), as were neonatal hospital stays (3.8 vs 5.7 days; P=.02), regardless of delivery method. The study had been planned for 92 patients; it was stopped early (n=48) after an interim analysis.

Because of the small sample size, other findings from the study must be viewed with caution. Intrapartum treatment with antibiotics was associated with a “significant” clinical reduction in neonatal sepsis (relative risk [RR]=0.08; 95% confidence interval [CI], 0.00–1.44) and pneumonia (RR=0.15; 95% CI, 0.01–2.92) compared with treatment given immediately postpartum; however, neither value was truly statistically significant according to the Cochrane review.4

The research suggests a potential benefit to adding clindamycin to ampicillin and gentamicin. In an effort to test this, 1 study randomized 133 women into 2 arms—treatment with ampicillin, gentamicin, and clindamycin compared with ampicillin and gentamicin alone—and found no additional benefit in regards to neonatal sepsis (RR=2.16; 95% CI, 0.20–23.21) or neonatal death (RR=0.72; 95% CI, 0.12–4.16).1 There was a trend towards a decrease in the incidence of postpartum endometritis in women who received ampicillin, gentamicin, and clindamycin, but this did not reach statistical significance (RR=0.54; 95% CI, 0.19–1.49).4

 

 

 

Recommendations from others

A 2002 bulletin from American College of Obstetricians and Gynecologists (ACOG) and the American Academy of Pediatrics5 recommended the combination of ampicillin 2 gm IV every 4 to 6 hours or penicillin 5 million units IV every 4 to 6 hours, plus an aminoglycoside (such as gentamicin 1.5 mg/kg IV every 8 hours), since this regimen provides appropriate coverage for typical organisms associated with acute chorioamnionitis. At the time the bulletin was published, the use of single daily dosing of aminoglycoside did not have sufficient studies to back its use. In addition, ACOG recommends adding clindamycin, metronidazole, or an extended-spectrum third-generation cephalosporin to the treatment regimen if cesarean section is required, to provide coverage for anaerobic organisms. They recommend clindamycin 900 mg IV every 8 hours to replace amoxicillin in penicillin-allergic patients. The Nottingham Guideline Development Group recommends amoxicillin 2 gm IV initially then 1 gm every 8 hours, and in place of gentamicin, recommends metronidazole 500 mg IV, every 8 hours (or 1 gm PR twice a day).6 Both recommendations suggest clindamycin 900 mg IV every 8 hours to replace amoxicillin in penicillin-allergic patients. For patients with nonanaphylactic reactions to penicillin, they recommend cefotaxime 1 g IV every 8 hours.

Acknowledgments

The opinions and assertions contained herein are the private views of the author and not to be construed as official, or as reflecting the views of the US Air Force Medical Service or the US Air Force at large.

References

1. Maberry MC, Gilstrap LC, 3rd. Intrapartum antibiotic therapy for suspected intraamniotic infection: impact on the fetus and neonate. Clin Obstet Gyn 1991;34:345-351.

2. Hauth JC, Gilstrap LC, Hankins GD, Conner KD. Term maternal and neonatal complications of acute chorioamnionitis. Obstet Gyn 1985;66:59-62.

3. Gibbs RS, Dinsmoor MJ, Newton ER, et al. A randomized trial of intrapartum versus immediate postpartum treatment of women with intra-amniotic infection. Obstet Gyn 1988;72:823-828.

4. Hopkins L, Smaill F. Antibiotic regimens for management of intraamniotic infection. Cochrane Database Syst Rev 2002;(3):CD003254.-

5. American College of Obstetricians and Gynecologists, American Academy of Pediatrics. Guidelines for Perinatal Care. 5th ed. Washington, DC: ACOG;2002:165-166.

6. Hayman R, Kean L. Guidelines for the Prevention of Neonatal Group B Streptococcal Infection. Nottingham: Nottingham City Hospital, National Health Service; 2002. Revised 2005. Available at: www.nuh.nhs.uk/nch/antibiotics. Accessed on March 30, 2007.

References

1. Maberry MC, Gilstrap LC, 3rd. Intrapartum antibiotic therapy for suspected intraamniotic infection: impact on the fetus and neonate. Clin Obstet Gyn 1991;34:345-351.

2. Hauth JC, Gilstrap LC, Hankins GD, Conner KD. Term maternal and neonatal complications of acute chorioamnionitis. Obstet Gyn 1985;66:59-62.

3. Gibbs RS, Dinsmoor MJ, Newton ER, et al. A randomized trial of intrapartum versus immediate postpartum treatment of women with intra-amniotic infection. Obstet Gyn 1988;72:823-828.

4. Hopkins L, Smaill F. Antibiotic regimens for management of intraamniotic infection. Cochrane Database Syst Rev 2002;(3):CD003254.-

5. American College of Obstetricians and Gynecologists, American Academy of Pediatrics. Guidelines for Perinatal Care. 5th ed. Washington, DC: ACOG;2002:165-166.

6. Hayman R, Kean L. Guidelines for the Prevention of Neonatal Group B Streptococcal Infection. Nottingham: Nottingham City Hospital, National Health Service; 2002. Revised 2005. Available at: www.nuh.nhs.uk/nch/antibiotics. Accessed on March 30, 2007.

Issue
The Journal of Family Practice - 56(5)
Issue
The Journal of Family Practice - 56(5)
Page Number
401-402
Page Number
401-402
Publications
Publications
Topics
Article Type
Display Headline
What treatment approach to intrapartum maternal fever has the best fetal outcomes?
Display Headline
What treatment approach to intrapartum maternal fever has the best fetal outcomes?
Legacy Keywords
fever; women’s health; pregnancy; birth; intrapartum; intrapartum fever; neonatal; obstetric
Legacy Keywords
fever; women’s health; pregnancy; birth; intrapartum; intrapartum fever; neonatal; obstetric
Sections
PURLs Copyright

Evidence-based answers from the Family Physicians Inquiries Network

Disallow All Ads
Alternative CME
Article PDF Media

What’s the best treatment for cradle cap?

Article Type
Changed
Mon, 01/14/2019 - 11:19
Display Headline
What’s the best treatment for cradle cap?
EVIDENCE-BASED ANSWER

Ketoconazole (Nizoral) shampoo appears to be a safe and efficacious treatment for infants with cradle cap (strength of recommendation [SOR]: C, consensus, usual practice, opinion, disease-oriented evidence, and case series). Limit topical corticosteroids to severe cases because of possible systemic absorption (SOR: C). Overnight application of emollients followed by gentle brushing and washing with baby shampoo helps to remove the scale associated with cradle cap (SOR: C).

Clinical commentary

If parents can’t leave it be, recommend mineral oil and a brush to loosen scale
Valerie J. King, MD, MPH
Oregon Health Sciences University, Portland

Cradle cap is distressing to parents. They want everyone else to see how gorgeous their new baby is, and cradle cap can make their beautiful little one look scruffy. My standard therapy has been to stress to the parents that it isn’t a problem for the baby.

If the parents still want to do something about it, I recommend mineral oil and a soft brush to loosen the scale. Although no evidence supports this, it seems safe and is somewhat effective.

This review makes me feel more comfortable with recommending ketoconazole shampoo when mineral oil proves insufficient. For resistant cases, a cute hat can work wonders.

 

Evidence summary

Cradle cap is a form of seborrheic dermatitis that manifests as greasy patches of scaling on the scalp of infants between the second week and sixth month of life.1,2 Untreated, it usually resolves at 8 months.1 It’s generally nonpruritic and doesn’t bother the infant, though it can be a stressor for parents.1

Researchers have noted a potential link with increased concentrations of the yeast Malassezia furfur (formerly Pityrosporum ovale), but a causative mechanism has not been identified.1,2 Overnight use of emollients such as mineral oil to soften scales followed by gentle brushing and washing with baby shampoo is an accepted treatment, although no trials could be found to show its efficacy for infants.1,3

Numerous treatments for seborrheic dermatitis with proven efficacy for adults have been adopted for use for infants. These include topical antifungals, anti-dandruff shampoos with zinc pyrithione or selenium sulfide, coal tar preparations, and episodic topical corticosteroids.1,4 Although each of these agents is used for infants with cradle cap, significantly sized randomized controlled trials in this age group are essentially absent.

Although limited evidence exists for seborrhea treatment in any age group, ketoconazole shampoo appears to be backed by the strongest evidence. For example, an uncontrolled multicenter trial with 575 adults found ketoconazole shampoo was superior to placebo for treatment of scalp seborrheic dermatitis with an 88% “excellent response” rate (P<.0001, no relative risk or confidence intervals given).4

 

 

 

Based on small studies, ketoconazole appears safe and effective for infants. A small (n=13) phase I safety trial of infants demonstrated that ketoconazole shampoo applied twice weekly for 1 month resulted in no detectable serum ketoconazole levels or elevation in liver function tests.5 In another small (n=19) uncontrolled study of once-daily ketoconazole 2% cream, 79% of infants affected with seborrheic dermatitis of the scalp and diaper area showed good response by day 10 (no statistical methods reported). Peak plasma ketoconazole levels in this study were only 1% to 2% of those documented after systemic administration.6

Studies conducted on topical steroids have also shown weak data. An unblinded uncontrolled comparative study of 2% ketoconazole cream and 1% hydrocortisone cream in the treatment of infantile seborrheic dermatitis revealed no statistical difference (31% vs 35%) in severity for 48 infants. All skin lesions in both treatment groups were cleared by the end of the second week of treatment.2

Multiple authors note safety concerns when considering treatment for mild and self-limited conditions such as cradle cap. Several studies have demonstrated systemic absorption and, in some cases, adrenocortical suppression when using mild topical steroids such as 1% hydrocortisone cream in pediatric populations.1,3,7

Recommendations from others

The guidance from PRODIGY (the UK’s National Health Service primary care database) recommends regular washing with baby shampoo followed by gentle brushing. Alternatively, softening the scale with mineral oil, followed by gentle brushing and shampooing is an alternative approach. Ketoconazole 2% shampoo or cream once a day has been shown to be effective; PRODIGY recommends avoiding topical corticosteroids.1

A review article recommends daily shampooing with an unmedicated shampoo. If this doesn’t work, the authors recommend trying a dandruff shampoo and softening the scales with mineral oil before washing.8 While the American Academy of Dermatology has no official guidelines on this subject, their patient-oriented pamphlet Dermatology Insights suggests that “cradle cap is treated with anti-dandruff or baby shampoo, with or without hydrocortisone lotion or cream, depending on the severity.”9

Acknowledgments

The opinions and assertions contained herein are the private views of the authors and not to be construed as official, or as reflecting the views of the US Air Force Medical Service or the US Air Force at large.

References

1. PRODIGY [database]. Seborrhoeic dermatitis. Knowledge Guidance structured review (2006). Sowerby Centre for Health Informatics at Newcastle Ltd (SCHIN). Available at: www.prodigy.nhs.uk/seborrhoeic_dermatitis. Accessed on February 6, 2007.

2. Wannanukul S, Chiabunkana J. Comparative study of 2% ketoconazole cream and 1% hydrocortisone cream in the treatment of infantile seborrheic dermatitis. J Med Assoc Thai 2004;87:S68-S71.

3. Janniger CK. Infantile seborrheic dermatitis: An approach to cradle cap. Cutis 1993;51:233-235.

4. Peter RU, Richarz-Barthauer U. Successful treatment and prophylaxis of scalp seborrhoeic dermatitis and dandruff with 2% ketoconazole shampoo: results of a multicentre, double-blind, placebo-controlled trial. Br J Dermatol 1995;132:441-445.

5. Brodell R, Patel S, Venglarick J, Moses D, Gemmel D. The safety of ketoconazole shampoo for infantile seborrheic dermatitis. Pediatr Dermatol 1998;15:406-407.

6. Taieb A, Legrain V, Palmier C, Lejean S, Six M, Maleville J. Topical ketoconazole for infantile seborrhoeic dermatitis. Dermatologica 1990;181:26-32.

7. Turpeinen M, Salo O, Leisti S. Effect of percutaneous absorption of hydrocortisone on adrenocortical responsiveness in infants with severe skin disease. Br J Dermatol 1986;115:475-484.

8. Seborrhea: What it is and how to treat it. Am Fam Physician 2000;61:2173-2174.

9. When to be concerned about childhood hair shedding. Dermatology Insights 2003;4(1):24.-Available at: www.aad.org/NR/rdonlyres/0AA67E605-01E104C7A-B493-9959923A8282/0/di_spring03.pdf#page=24. Accessed on February 6, 2007.

Article PDF
Author and Disclosure Information

Ryan C. Sheffield, MD
Paul Crawford, MD
Eglin Air Force Base Family Medicine Residency, Eglin Air Force Base, Fla

Sarah Towner Wright, MLS
University of North Carolina at Chapel Hill

Issue
The Journal of Family Practice - 56(03)
Publications
Topics
Page Number
232-233
Legacy Keywords
cradle cap; Ryan C. Sheffield;MD; Paul Crawford;MD; Sarah Towner Wright;MLS; Sheffield RC; Crawford P; Wright ST; Valerie J. King;MD;MPH; King VJ; ketoconazole; Nizoral;
Sections
Author and Disclosure Information

Ryan C. Sheffield, MD
Paul Crawford, MD
Eglin Air Force Base Family Medicine Residency, Eglin Air Force Base, Fla

Sarah Towner Wright, MLS
University of North Carolina at Chapel Hill

Author and Disclosure Information

Ryan C. Sheffield, MD
Paul Crawford, MD
Eglin Air Force Base Family Medicine Residency, Eglin Air Force Base, Fla

Sarah Towner Wright, MLS
University of North Carolina at Chapel Hill

Article PDF
Article PDF
EVIDENCE-BASED ANSWER

Ketoconazole (Nizoral) shampoo appears to be a safe and efficacious treatment for infants with cradle cap (strength of recommendation [SOR]: C, consensus, usual practice, opinion, disease-oriented evidence, and case series). Limit topical corticosteroids to severe cases because of possible systemic absorption (SOR: C). Overnight application of emollients followed by gentle brushing and washing with baby shampoo helps to remove the scale associated with cradle cap (SOR: C).

Clinical commentary

If parents can’t leave it be, recommend mineral oil and a brush to loosen scale
Valerie J. King, MD, MPH
Oregon Health Sciences University, Portland

Cradle cap is distressing to parents. They want everyone else to see how gorgeous their new baby is, and cradle cap can make their beautiful little one look scruffy. My standard therapy has been to stress to the parents that it isn’t a problem for the baby.

If the parents still want to do something about it, I recommend mineral oil and a soft brush to loosen the scale. Although no evidence supports this, it seems safe and is somewhat effective.

This review makes me feel more comfortable with recommending ketoconazole shampoo when mineral oil proves insufficient. For resistant cases, a cute hat can work wonders.

 

Evidence summary

Cradle cap is a form of seborrheic dermatitis that manifests as greasy patches of scaling on the scalp of infants between the second week and sixth month of life.1,2 Untreated, it usually resolves at 8 months.1 It’s generally nonpruritic and doesn’t bother the infant, though it can be a stressor for parents.1

Researchers have noted a potential link with increased concentrations of the yeast Malassezia furfur (formerly Pityrosporum ovale), but a causative mechanism has not been identified.1,2 Overnight use of emollients such as mineral oil to soften scales followed by gentle brushing and washing with baby shampoo is an accepted treatment, although no trials could be found to show its efficacy for infants.1,3

Numerous treatments for seborrheic dermatitis with proven efficacy for adults have been adopted for use for infants. These include topical antifungals, anti-dandruff shampoos with zinc pyrithione or selenium sulfide, coal tar preparations, and episodic topical corticosteroids.1,4 Although each of these agents is used for infants with cradle cap, significantly sized randomized controlled trials in this age group are essentially absent.

Although limited evidence exists for seborrhea treatment in any age group, ketoconazole shampoo appears to be backed by the strongest evidence. For example, an uncontrolled multicenter trial with 575 adults found ketoconazole shampoo was superior to placebo for treatment of scalp seborrheic dermatitis with an 88% “excellent response” rate (P<.0001, no relative risk or confidence intervals given).4

 

 

 

Based on small studies, ketoconazole appears safe and effective for infants. A small (n=13) phase I safety trial of infants demonstrated that ketoconazole shampoo applied twice weekly for 1 month resulted in no detectable serum ketoconazole levels or elevation in liver function tests.5 In another small (n=19) uncontrolled study of once-daily ketoconazole 2% cream, 79% of infants affected with seborrheic dermatitis of the scalp and diaper area showed good response by day 10 (no statistical methods reported). Peak plasma ketoconazole levels in this study were only 1% to 2% of those documented after systemic administration.6

Studies conducted on topical steroids have also shown weak data. An unblinded uncontrolled comparative study of 2% ketoconazole cream and 1% hydrocortisone cream in the treatment of infantile seborrheic dermatitis revealed no statistical difference (31% vs 35%) in severity for 48 infants. All skin lesions in both treatment groups were cleared by the end of the second week of treatment.2

Multiple authors note safety concerns when considering treatment for mild and self-limited conditions such as cradle cap. Several studies have demonstrated systemic absorption and, in some cases, adrenocortical suppression when using mild topical steroids such as 1% hydrocortisone cream in pediatric populations.1,3,7

Recommendations from others

The guidance from PRODIGY (the UK’s National Health Service primary care database) recommends regular washing with baby shampoo followed by gentle brushing. Alternatively, softening the scale with mineral oil, followed by gentle brushing and shampooing is an alternative approach. Ketoconazole 2% shampoo or cream once a day has been shown to be effective; PRODIGY recommends avoiding topical corticosteroids.1

A review article recommends daily shampooing with an unmedicated shampoo. If this doesn’t work, the authors recommend trying a dandruff shampoo and softening the scales with mineral oil before washing.8 While the American Academy of Dermatology has no official guidelines on this subject, their patient-oriented pamphlet Dermatology Insights suggests that “cradle cap is treated with anti-dandruff or baby shampoo, with or without hydrocortisone lotion or cream, depending on the severity.”9

Acknowledgments

The opinions and assertions contained herein are the private views of the authors and not to be construed as official, or as reflecting the views of the US Air Force Medical Service or the US Air Force at large.

EVIDENCE-BASED ANSWER

Ketoconazole (Nizoral) shampoo appears to be a safe and efficacious treatment for infants with cradle cap (strength of recommendation [SOR]: C, consensus, usual practice, opinion, disease-oriented evidence, and case series). Limit topical corticosteroids to severe cases because of possible systemic absorption (SOR: C). Overnight application of emollients followed by gentle brushing and washing with baby shampoo helps to remove the scale associated with cradle cap (SOR: C).

Clinical commentary

If parents can’t leave it be, recommend mineral oil and a brush to loosen scale
Valerie J. King, MD, MPH
Oregon Health Sciences University, Portland

Cradle cap is distressing to parents. They want everyone else to see how gorgeous their new baby is, and cradle cap can make their beautiful little one look scruffy. My standard therapy has been to stress to the parents that it isn’t a problem for the baby.

If the parents still want to do something about it, I recommend mineral oil and a soft brush to loosen the scale. Although no evidence supports this, it seems safe and is somewhat effective.

This review makes me feel more comfortable with recommending ketoconazole shampoo when mineral oil proves insufficient. For resistant cases, a cute hat can work wonders.

 

Evidence summary

Cradle cap is a form of seborrheic dermatitis that manifests as greasy patches of scaling on the scalp of infants between the second week and sixth month of life.1,2 Untreated, it usually resolves at 8 months.1 It’s generally nonpruritic and doesn’t bother the infant, though it can be a stressor for parents.1

Researchers have noted a potential link with increased concentrations of the yeast Malassezia furfur (formerly Pityrosporum ovale), but a causative mechanism has not been identified.1,2 Overnight use of emollients such as mineral oil to soften scales followed by gentle brushing and washing with baby shampoo is an accepted treatment, although no trials could be found to show its efficacy for infants.1,3

Numerous treatments for seborrheic dermatitis with proven efficacy for adults have been adopted for use for infants. These include topical antifungals, anti-dandruff shampoos with zinc pyrithione or selenium sulfide, coal tar preparations, and episodic topical corticosteroids.1,4 Although each of these agents is used for infants with cradle cap, significantly sized randomized controlled trials in this age group are essentially absent.

Although limited evidence exists for seborrhea treatment in any age group, ketoconazole shampoo appears to be backed by the strongest evidence. For example, an uncontrolled multicenter trial with 575 adults found ketoconazole shampoo was superior to placebo for treatment of scalp seborrheic dermatitis with an 88% “excellent response” rate (P<.0001, no relative risk or confidence intervals given).4

 

 

 

Based on small studies, ketoconazole appears safe and effective for infants. A small (n=13) phase I safety trial of infants demonstrated that ketoconazole shampoo applied twice weekly for 1 month resulted in no detectable serum ketoconazole levels or elevation in liver function tests.5 In another small (n=19) uncontrolled study of once-daily ketoconazole 2% cream, 79% of infants affected with seborrheic dermatitis of the scalp and diaper area showed good response by day 10 (no statistical methods reported). Peak plasma ketoconazole levels in this study were only 1% to 2% of those documented after systemic administration.6

Studies conducted on topical steroids have also shown weak data. An unblinded uncontrolled comparative study of 2% ketoconazole cream and 1% hydrocortisone cream in the treatment of infantile seborrheic dermatitis revealed no statistical difference (31% vs 35%) in severity for 48 infants. All skin lesions in both treatment groups were cleared by the end of the second week of treatment.2

Multiple authors note safety concerns when considering treatment for mild and self-limited conditions such as cradle cap. Several studies have demonstrated systemic absorption and, in some cases, adrenocortical suppression when using mild topical steroids such as 1% hydrocortisone cream in pediatric populations.1,3,7

Recommendations from others

The guidance from PRODIGY (the UK’s National Health Service primary care database) recommends regular washing with baby shampoo followed by gentle brushing. Alternatively, softening the scale with mineral oil, followed by gentle brushing and shampooing is an alternative approach. Ketoconazole 2% shampoo or cream once a day has been shown to be effective; PRODIGY recommends avoiding topical corticosteroids.1

A review article recommends daily shampooing with an unmedicated shampoo. If this doesn’t work, the authors recommend trying a dandruff shampoo and softening the scales with mineral oil before washing.8 While the American Academy of Dermatology has no official guidelines on this subject, their patient-oriented pamphlet Dermatology Insights suggests that “cradle cap is treated with anti-dandruff or baby shampoo, with or without hydrocortisone lotion or cream, depending on the severity.”9

Acknowledgments

The opinions and assertions contained herein are the private views of the authors and not to be construed as official, or as reflecting the views of the US Air Force Medical Service or the US Air Force at large.

References

1. PRODIGY [database]. Seborrhoeic dermatitis. Knowledge Guidance structured review (2006). Sowerby Centre for Health Informatics at Newcastle Ltd (SCHIN). Available at: www.prodigy.nhs.uk/seborrhoeic_dermatitis. Accessed on February 6, 2007.

2. Wannanukul S, Chiabunkana J. Comparative study of 2% ketoconazole cream and 1% hydrocortisone cream in the treatment of infantile seborrheic dermatitis. J Med Assoc Thai 2004;87:S68-S71.

3. Janniger CK. Infantile seborrheic dermatitis: An approach to cradle cap. Cutis 1993;51:233-235.

4. Peter RU, Richarz-Barthauer U. Successful treatment and prophylaxis of scalp seborrhoeic dermatitis and dandruff with 2% ketoconazole shampoo: results of a multicentre, double-blind, placebo-controlled trial. Br J Dermatol 1995;132:441-445.

5. Brodell R, Patel S, Venglarick J, Moses D, Gemmel D. The safety of ketoconazole shampoo for infantile seborrheic dermatitis. Pediatr Dermatol 1998;15:406-407.

6. Taieb A, Legrain V, Palmier C, Lejean S, Six M, Maleville J. Topical ketoconazole for infantile seborrhoeic dermatitis. Dermatologica 1990;181:26-32.

7. Turpeinen M, Salo O, Leisti S. Effect of percutaneous absorption of hydrocortisone on adrenocortical responsiveness in infants with severe skin disease. Br J Dermatol 1986;115:475-484.

8. Seborrhea: What it is and how to treat it. Am Fam Physician 2000;61:2173-2174.

9. When to be concerned about childhood hair shedding. Dermatology Insights 2003;4(1):24.-Available at: www.aad.org/NR/rdonlyres/0AA67E605-01E104C7A-B493-9959923A8282/0/di_spring03.pdf#page=24. Accessed on February 6, 2007.

References

1. PRODIGY [database]. Seborrhoeic dermatitis. Knowledge Guidance structured review (2006). Sowerby Centre for Health Informatics at Newcastle Ltd (SCHIN). Available at: www.prodigy.nhs.uk/seborrhoeic_dermatitis. Accessed on February 6, 2007.

2. Wannanukul S, Chiabunkana J. Comparative study of 2% ketoconazole cream and 1% hydrocortisone cream in the treatment of infantile seborrheic dermatitis. J Med Assoc Thai 2004;87:S68-S71.

3. Janniger CK. Infantile seborrheic dermatitis: An approach to cradle cap. Cutis 1993;51:233-235.

4. Peter RU, Richarz-Barthauer U. Successful treatment and prophylaxis of scalp seborrhoeic dermatitis and dandruff with 2% ketoconazole shampoo: results of a multicentre, double-blind, placebo-controlled trial. Br J Dermatol 1995;132:441-445.

5. Brodell R, Patel S, Venglarick J, Moses D, Gemmel D. The safety of ketoconazole shampoo for infantile seborrheic dermatitis. Pediatr Dermatol 1998;15:406-407.

6. Taieb A, Legrain V, Palmier C, Lejean S, Six M, Maleville J. Topical ketoconazole for infantile seborrhoeic dermatitis. Dermatologica 1990;181:26-32.

7. Turpeinen M, Salo O, Leisti S. Effect of percutaneous absorption of hydrocortisone on adrenocortical responsiveness in infants with severe skin disease. Br J Dermatol 1986;115:475-484.

8. Seborrhea: What it is and how to treat it. Am Fam Physician 2000;61:2173-2174.

9. When to be concerned about childhood hair shedding. Dermatology Insights 2003;4(1):24.-Available at: www.aad.org/NR/rdonlyres/0AA67E605-01E104C7A-B493-9959923A8282/0/di_spring03.pdf#page=24. Accessed on February 6, 2007.

Issue
The Journal of Family Practice - 56(03)
Issue
The Journal of Family Practice - 56(03)
Page Number
232-233
Page Number
232-233
Publications
Publications
Topics
Article Type
Display Headline
What’s the best treatment for cradle cap?
Display Headline
What’s the best treatment for cradle cap?
Legacy Keywords
cradle cap; Ryan C. Sheffield;MD; Paul Crawford;MD; Sarah Towner Wright;MLS; Sheffield RC; Crawford P; Wright ST; Valerie J. King;MD;MPH; King VJ; ketoconazole; Nizoral;
Legacy Keywords
cradle cap; Ryan C. Sheffield;MD; Paul Crawford;MD; Sarah Towner Wright;MLS; Sheffield RC; Crawford P; Wright ST; Valerie J. King;MD;MPH; King VJ; ketoconazole; Nizoral;
Sections
PURLs Copyright

Evidence-based answers from the Family Physicians Inquiries Network

Disallow All Ads
Article PDF Media

What is the dietary treatment for low HDL cholesterol?

Article Type
Changed
Mon, 01/14/2019 - 11:18
Display Headline
What is the dietary treatment for low HDL cholesterol?
EVIDENCE-BASED ANSWER

Low-carbohydrate diets raise high-density lipoprotein (HDL) cholesterol levels by approximately 10%; soy protein with isoflavones raises HDL by 3% (strength of recommendation [SOR]: C, based on meta-analysis of physiologic parameters). The Dietary Approaches to Stop Hypertension (DASH) diet and multivitamin supplementation raise HDL 21% to 33% (SOR: C, based on single randomized trial each measuring physiologic parameters). No other dietary interventions studied raise HDL (SOR: C, based on meta-analysis of physiologic parameters).

CLINICAL COMMENTARY

Michael K. Park, MD
University of Colorado Health Sciences Center, Rose Family Medicine Residency, Denver

Even modest increases in HDL can be clinically important; exercise, weight loss, and tobacco cessation can help When it comes to HDL, most of our patients are not as fortunate as natives of Limone sul Garda, Italy (famously low but efficient HDL) or Honshu, Japan (high HDL). Medications based on these protective genetic anomalies are being developed. Also, the flushing resulting from niacin may soon be more effectively mitigated than with aspirin. Until these new therapies are available, urge multifaceted lifestyle modification—if only for its more robust cardiovascular benefits.

A low HDL can elicit a clinical fatalism from even the best of us. But each increase in baseline HDL of 1 mg/dL is associated with a 5% decrease in the risk of death from coronary disease,1 so even modest increases in HDL can be clinically important. In addition to the dietary measures described above, evidence exists that exercise, alcohol in moderation, weight loss, and tobacco cessation also increase HDL. Unfortunately, the magnitude of even these small improvements appear to be directly proportional to baseline HDL levels.2

So … when are those new medications coming?

 

Evidence summary

Low HDL is recognized as a risk factor for atherosclerosis. Clinicians find raising HDL a challenge, and patients often inquire about dietary advice that may help raise HDL.

No quality evidence exists that specifically looks at the effect of a dietary intervention on HDL or whether it affects survival. However, several dietary intervention studies in specific populations include HDL as a secondary endpoint in the study. This leaves clinicians to act on physiologic data that may or may not increase the overall health and survival of patients. Dietary interventions that raised HDL include low-carbohydrate diets, the DASH diet, supplementation with soy protein including isoflavones, and multivitamin supplementation.

TABLE
Summary of studies evaluating the effect of various diets on HDL cholesterol

STUDYINTERVENTIONMETHODSHDL EFFECT
Nordmann, et al 20063; Wood et al 20064Low-carbohydrate dietsSystematic review with meta-analysis of 5 RCTs of low-carbohydrate vs low-fat diets. 10% increase in HDL. Absolute increase 4.6 mg/dL (95% CI, 1.5–8.1). Subsequent uncontrolled prospective trial consistent with systematic review (12% increase in HDL)++
Zhan and Ho 20057Soy protein with isoflavonesSystematic review with meta-analysis of 23 RCTs evaluating effect of various amounts of soy protein with isoflavones on lipid profile. 3% increase in HDL. Absolute difference 1.5 mg/dL (95% CI, 0.0–2.8)+
Morcos 19996MultivitaminRCT of 46 subjects in placebo controlled crossover study. 31% increase in HDL++++
Azadbacht et al 20055DASH diet in metabolic syndromeRCT of 116 patients randomized to control diet, weight control or DASH diet. Absolute increase of 7 mg/dL for men (21%). Absolute increase of 10 mg/dL for women (33%). No confidence intervals given+++
Kelly et al 20048Low glycemic dietSystematic review of 15 RCTs with low glycemic diets for patients with coronary heart disease. Heterogeneity prevented meta-analysis. No effect on HDL.0
Brunner et al 20059Dietary adviceSystematic review of 23 RCTs comparing dietary advice and no advice to reduce cardiovascular risk. 956 participants. No effect on HDL.0
Chen et al 200610Soluble fiberRCT of 110 adults randomized to 8 g of soluble fiber vs control. No effect on HDL.0
Lewis et al 200411Omega-3 fatty acids in hyper-triglyceridemiaSystematic review of 10 RCTs comparing omega-3 fatty acids. Considered poor to moderate quality. Variable results from RCTs???
Farmer et al 200112Omega-3 fatty acids in type 2 diabetesSystematic review with meta-analysis of 18 trials with 823 patients. No effect on HDL0
Hooper et al 200413Omega-3 fatty acids for prevention of CVDSystematic review with meta-analysis of 48 trials of 36,913 participants taking omega-3 fatty acids for prevention of cardiovascular disease. No effect on HDL0
Tapsell et al 2004;14 Spiller et al 199815Walnuts and almondsOne RCT and one prospective cohort trial of nuts added to the diet. No significant effect0

Several overall diet interventions appear to raise HDL, but whether this affects cardiovascular events or mortality is unknown. A systematic review with meta-analysis of 5 randomized controlled trials (RCTs) of low-carbohydrate versus low-fat diets showed a 10% increase in HDL attributed to the low-carbohydrate diet, which translated to an absolute increase of 4.6 mg/dL (95% confidence interval [CI], 1.5–8.1).1 A subsequent uncontrolled prospective trial was consistent with consistent with this systematic review and showed a 12% increase in HDL.2 The DASH diet was studied as an intervention in a RCT of 116 patients with the metabolic syndrome. Men responded with an in crease of 21% and women with an increase of 33%.3

 

 

 

Supplementation with several food additives and nutritional supplements has been tested. A systematic review with meta-analysis of 23 RCTs evaluating effect of various amounts of soy protein with isoflavones on lipid profile found a 3% increase in HDL with an absolute difference 1.5 mg/dL (95% CI, 0.0–2.8).4 Supplementation with standard multivitamins in a single small, crossover RCT showed a 31% increase in HDL.5

Many other strategies, supplements, and plans have been tested in different populations. Other than the above interventions, no other interventions raise HDL when subjected to meta-analysis or quality randomized trials (TABLE).

Recommendations from others

No specific guidelines on dietary therapy of HDL exist; however, the American Heart Association (AHA) published diet and lifestyle recommendations in 2006.14 These guidelines recommend a diet low in fat, saturated fat, trans fat, and cholesterol in addition to minimizing sodium, added sugars, and alcohol. The AHA also recommends for consumption of oily fish and the DASH diet.

Acknowledgments

The opinions and assertions contained herein are the private views of the author and not to be construed as official, or as reflecting the views of the US Air Force Medical Service or the US Air Force at large.

References

1. Gordon DJ, Knoke J, Probstfield JL, Superko R, Tyroler HA. High-density lipoprotein cholesterol and coronary heart disease in hypercholesterolemic men: the Lipid Research Clinics Coronary Primary Prevention Trial. Circulation 1986;74:1217-1225.

2. Williams PT. The relationships of vigorous exercise, alcohol, and adiposity to low and high high-density lipoproteincholesterol levels. Metabolism 2004;53:700-709.

3. Nordmann AJ, Nordmann A, Briel M, et al. Effects of low-carbohydrate vs low-fat diets on weight loss and cardiovascular risk factors: a meta-analysis of randomized controlled trials. Arch Intern Med 2006;166:285-293.

4. Wood RJ, Volek JS, Y Liu, NS Schacter, JH Contois, ML Fernandez. Carbohydrate restriction alters lipoprotein metabolism by modifying VLDL, LDL, and HDL subfraction distribution and size in overweight men. J Nutr 2006;136:384-389.

5. Azadbakht L, Mirmiran P, Esmaillzadeh A, Azizi T, Azizi F. Beneficial effects of a Dietary Approaches to Stop Hypertension eating plan on features of the metabolic syndrome. Diabetes Care 2005;28:2823-2831.

6. Zhan S, Ho SC. Meta-analysis of the effects of soy protein containing isoflavones on the lipid profile. Am J Clin Nutr 2005;81:397-408.

7. Morcos N. Increase in serum high-density lipoprotein following multivitamin and multimineral supplementation in adults with cardiovascular risk factors. Med Sci Res 1999;27:121-125.

8. Kelly S, Frost G, Whittaker V, Summerbell C. Low glycaemic index diets for coronary heart disease. Cochrane Database Syst Rev 2004;(4):CD004467.-

9. Brunner EJ, Thorogood M, Rees K, Hewitt G. Dietary advice for reducing cardiovascular risk. Cochrane Database Syst Rev 2005;(4):CD002128.-

10. Chen J, He J, Wildman RP, Reynolds K, Streiffer RH, Whelton PK. A randomized controlled trial of dietary fiber intake on serum lipids. Eur J Clin Nutr 2006;60:62-68.

11. Lewis A, Lookinland S, Beckstrand RL, Tiedeman ME. Treatment of hypertriglyceridemia with omega-3 fatty acids: A systematic review. J Am Acad Nurse Pract 2004;16:384-395.

12. Farmer A, Montori V, Dinneen S, Clar C. Fish oil in people with type 2 diabetes mellitus. Cochrane Database Syst Rev 2001;(3):CD003205.-

13. Hooper L, Thompson RL, Harrison RA, et al. Omega 3 fatty acids for prevention and treatment of cardiovascular disease. Cochrane Database Syst Rev 2004;(4):CD003177.-

14. Tapsell LC, Gillen LJ, Patch CS, et al. Including walnuts in a low-fat/modified-fat diet improves HDL cholesterol-to-total cholesterol ratios in patients with type 2 diabetes. Diabetes Care 2004;27:2777-2783.

15. Spiller GA, Jenkins DA, Bosello O, Gates JE, Cragen LN, Bruce B. Nuts and plasma lipids: an almond-based diet lowers LDL-C while preserving HDL-C. J Am Coll Nutr 1998;17:285-290.

16. Lichtenstein AH, Appel LJ, Brands M, et al. Diet and lifestyle recommendations revision 2006: A scientific statement from the American Heart Association Nutrition Committee. Circulation 2006;114:82-96.

Article PDF
Author and Disclosure Information

Paul Crawford, MD
Eglin Air Force Base Family Medicine Residency, Eglin AFB, FL

Shelley L. Paden, MLS, AHIP
Academic Information Technology and Libraries, University of Cincinnati

Issue
The Journal of Family Practice - 55(12)
Publications
Topics
Page Number
1076-1078
Sections
Author and Disclosure Information

Paul Crawford, MD
Eglin Air Force Base Family Medicine Residency, Eglin AFB, FL

Shelley L. Paden, MLS, AHIP
Academic Information Technology and Libraries, University of Cincinnati

Author and Disclosure Information

Paul Crawford, MD
Eglin Air Force Base Family Medicine Residency, Eglin AFB, FL

Shelley L. Paden, MLS, AHIP
Academic Information Technology and Libraries, University of Cincinnati

Article PDF
Article PDF
EVIDENCE-BASED ANSWER

Low-carbohydrate diets raise high-density lipoprotein (HDL) cholesterol levels by approximately 10%; soy protein with isoflavones raises HDL by 3% (strength of recommendation [SOR]: C, based on meta-analysis of physiologic parameters). The Dietary Approaches to Stop Hypertension (DASH) diet and multivitamin supplementation raise HDL 21% to 33% (SOR: C, based on single randomized trial each measuring physiologic parameters). No other dietary interventions studied raise HDL (SOR: C, based on meta-analysis of physiologic parameters).

CLINICAL COMMENTARY

Michael K. Park, MD
University of Colorado Health Sciences Center, Rose Family Medicine Residency, Denver

Even modest increases in HDL can be clinically important; exercise, weight loss, and tobacco cessation can help When it comes to HDL, most of our patients are not as fortunate as natives of Limone sul Garda, Italy (famously low but efficient HDL) or Honshu, Japan (high HDL). Medications based on these protective genetic anomalies are being developed. Also, the flushing resulting from niacin may soon be more effectively mitigated than with aspirin. Until these new therapies are available, urge multifaceted lifestyle modification—if only for its more robust cardiovascular benefits.

A low HDL can elicit a clinical fatalism from even the best of us. But each increase in baseline HDL of 1 mg/dL is associated with a 5% decrease in the risk of death from coronary disease,1 so even modest increases in HDL can be clinically important. In addition to the dietary measures described above, evidence exists that exercise, alcohol in moderation, weight loss, and tobacco cessation also increase HDL. Unfortunately, the magnitude of even these small improvements appear to be directly proportional to baseline HDL levels.2

So … when are those new medications coming?

 

Evidence summary

Low HDL is recognized as a risk factor for atherosclerosis. Clinicians find raising HDL a challenge, and patients often inquire about dietary advice that may help raise HDL.

No quality evidence exists that specifically looks at the effect of a dietary intervention on HDL or whether it affects survival. However, several dietary intervention studies in specific populations include HDL as a secondary endpoint in the study. This leaves clinicians to act on physiologic data that may or may not increase the overall health and survival of patients. Dietary interventions that raised HDL include low-carbohydrate diets, the DASH diet, supplementation with soy protein including isoflavones, and multivitamin supplementation.

TABLE
Summary of studies evaluating the effect of various diets on HDL cholesterol

STUDYINTERVENTIONMETHODSHDL EFFECT
Nordmann, et al 20063; Wood et al 20064Low-carbohydrate dietsSystematic review with meta-analysis of 5 RCTs of low-carbohydrate vs low-fat diets. 10% increase in HDL. Absolute increase 4.6 mg/dL (95% CI, 1.5–8.1). Subsequent uncontrolled prospective trial consistent with systematic review (12% increase in HDL)++
Zhan and Ho 20057Soy protein with isoflavonesSystematic review with meta-analysis of 23 RCTs evaluating effect of various amounts of soy protein with isoflavones on lipid profile. 3% increase in HDL. Absolute difference 1.5 mg/dL (95% CI, 0.0–2.8)+
Morcos 19996MultivitaminRCT of 46 subjects in placebo controlled crossover study. 31% increase in HDL++++
Azadbacht et al 20055DASH diet in metabolic syndromeRCT of 116 patients randomized to control diet, weight control or DASH diet. Absolute increase of 7 mg/dL for men (21%). Absolute increase of 10 mg/dL for women (33%). No confidence intervals given+++
Kelly et al 20048Low glycemic dietSystematic review of 15 RCTs with low glycemic diets for patients with coronary heart disease. Heterogeneity prevented meta-analysis. No effect on HDL.0
Brunner et al 20059Dietary adviceSystematic review of 23 RCTs comparing dietary advice and no advice to reduce cardiovascular risk. 956 participants. No effect on HDL.0
Chen et al 200610Soluble fiberRCT of 110 adults randomized to 8 g of soluble fiber vs control. No effect on HDL.0
Lewis et al 200411Omega-3 fatty acids in hyper-triglyceridemiaSystematic review of 10 RCTs comparing omega-3 fatty acids. Considered poor to moderate quality. Variable results from RCTs???
Farmer et al 200112Omega-3 fatty acids in type 2 diabetesSystematic review with meta-analysis of 18 trials with 823 patients. No effect on HDL0
Hooper et al 200413Omega-3 fatty acids for prevention of CVDSystematic review with meta-analysis of 48 trials of 36,913 participants taking omega-3 fatty acids for prevention of cardiovascular disease. No effect on HDL0
Tapsell et al 2004;14 Spiller et al 199815Walnuts and almondsOne RCT and one prospective cohort trial of nuts added to the diet. No significant effect0

Several overall diet interventions appear to raise HDL, but whether this affects cardiovascular events or mortality is unknown. A systematic review with meta-analysis of 5 randomized controlled trials (RCTs) of low-carbohydrate versus low-fat diets showed a 10% increase in HDL attributed to the low-carbohydrate diet, which translated to an absolute increase of 4.6 mg/dL (95% confidence interval [CI], 1.5–8.1).1 A subsequent uncontrolled prospective trial was consistent with consistent with this systematic review and showed a 12% increase in HDL.2 The DASH diet was studied as an intervention in a RCT of 116 patients with the metabolic syndrome. Men responded with an in crease of 21% and women with an increase of 33%.3

 

 

 

Supplementation with several food additives and nutritional supplements has been tested. A systematic review with meta-analysis of 23 RCTs evaluating effect of various amounts of soy protein with isoflavones on lipid profile found a 3% increase in HDL with an absolute difference 1.5 mg/dL (95% CI, 0.0–2.8).4 Supplementation with standard multivitamins in a single small, crossover RCT showed a 31% increase in HDL.5

Many other strategies, supplements, and plans have been tested in different populations. Other than the above interventions, no other interventions raise HDL when subjected to meta-analysis or quality randomized trials (TABLE).

Recommendations from others

No specific guidelines on dietary therapy of HDL exist; however, the American Heart Association (AHA) published diet and lifestyle recommendations in 2006.14 These guidelines recommend a diet low in fat, saturated fat, trans fat, and cholesterol in addition to minimizing sodium, added sugars, and alcohol. The AHA also recommends for consumption of oily fish and the DASH diet.

Acknowledgments

The opinions and assertions contained herein are the private views of the author and not to be construed as official, or as reflecting the views of the US Air Force Medical Service or the US Air Force at large.

EVIDENCE-BASED ANSWER

Low-carbohydrate diets raise high-density lipoprotein (HDL) cholesterol levels by approximately 10%; soy protein with isoflavones raises HDL by 3% (strength of recommendation [SOR]: C, based on meta-analysis of physiologic parameters). The Dietary Approaches to Stop Hypertension (DASH) diet and multivitamin supplementation raise HDL 21% to 33% (SOR: C, based on single randomized trial each measuring physiologic parameters). No other dietary interventions studied raise HDL (SOR: C, based on meta-analysis of physiologic parameters).

CLINICAL COMMENTARY

Michael K. Park, MD
University of Colorado Health Sciences Center, Rose Family Medicine Residency, Denver

Even modest increases in HDL can be clinically important; exercise, weight loss, and tobacco cessation can help When it comes to HDL, most of our patients are not as fortunate as natives of Limone sul Garda, Italy (famously low but efficient HDL) or Honshu, Japan (high HDL). Medications based on these protective genetic anomalies are being developed. Also, the flushing resulting from niacin may soon be more effectively mitigated than with aspirin. Until these new therapies are available, urge multifaceted lifestyle modification—if only for its more robust cardiovascular benefits.

A low HDL can elicit a clinical fatalism from even the best of us. But each increase in baseline HDL of 1 mg/dL is associated with a 5% decrease in the risk of death from coronary disease,1 so even modest increases in HDL can be clinically important. In addition to the dietary measures described above, evidence exists that exercise, alcohol in moderation, weight loss, and tobacco cessation also increase HDL. Unfortunately, the magnitude of even these small improvements appear to be directly proportional to baseline HDL levels.2

So … when are those new medications coming?

 

Evidence summary

Low HDL is recognized as a risk factor for atherosclerosis. Clinicians find raising HDL a challenge, and patients often inquire about dietary advice that may help raise HDL.

No quality evidence exists that specifically looks at the effect of a dietary intervention on HDL or whether it affects survival. However, several dietary intervention studies in specific populations include HDL as a secondary endpoint in the study. This leaves clinicians to act on physiologic data that may or may not increase the overall health and survival of patients. Dietary interventions that raised HDL include low-carbohydrate diets, the DASH diet, supplementation with soy protein including isoflavones, and multivitamin supplementation.

TABLE
Summary of studies evaluating the effect of various diets on HDL cholesterol

STUDYINTERVENTIONMETHODSHDL EFFECT
Nordmann, et al 20063; Wood et al 20064Low-carbohydrate dietsSystematic review with meta-analysis of 5 RCTs of low-carbohydrate vs low-fat diets. 10% increase in HDL. Absolute increase 4.6 mg/dL (95% CI, 1.5–8.1). Subsequent uncontrolled prospective trial consistent with systematic review (12% increase in HDL)++
Zhan and Ho 20057Soy protein with isoflavonesSystematic review with meta-analysis of 23 RCTs evaluating effect of various amounts of soy protein with isoflavones on lipid profile. 3% increase in HDL. Absolute difference 1.5 mg/dL (95% CI, 0.0–2.8)+
Morcos 19996MultivitaminRCT of 46 subjects in placebo controlled crossover study. 31% increase in HDL++++
Azadbacht et al 20055DASH diet in metabolic syndromeRCT of 116 patients randomized to control diet, weight control or DASH diet. Absolute increase of 7 mg/dL for men (21%). Absolute increase of 10 mg/dL for women (33%). No confidence intervals given+++
Kelly et al 20048Low glycemic dietSystematic review of 15 RCTs with low glycemic diets for patients with coronary heart disease. Heterogeneity prevented meta-analysis. No effect on HDL.0
Brunner et al 20059Dietary adviceSystematic review of 23 RCTs comparing dietary advice and no advice to reduce cardiovascular risk. 956 participants. No effect on HDL.0
Chen et al 200610Soluble fiberRCT of 110 adults randomized to 8 g of soluble fiber vs control. No effect on HDL.0
Lewis et al 200411Omega-3 fatty acids in hyper-triglyceridemiaSystematic review of 10 RCTs comparing omega-3 fatty acids. Considered poor to moderate quality. Variable results from RCTs???
Farmer et al 200112Omega-3 fatty acids in type 2 diabetesSystematic review with meta-analysis of 18 trials with 823 patients. No effect on HDL0
Hooper et al 200413Omega-3 fatty acids for prevention of CVDSystematic review with meta-analysis of 48 trials of 36,913 participants taking omega-3 fatty acids for prevention of cardiovascular disease. No effect on HDL0
Tapsell et al 2004;14 Spiller et al 199815Walnuts and almondsOne RCT and one prospective cohort trial of nuts added to the diet. No significant effect0

Several overall diet interventions appear to raise HDL, but whether this affects cardiovascular events or mortality is unknown. A systematic review with meta-analysis of 5 randomized controlled trials (RCTs) of low-carbohydrate versus low-fat diets showed a 10% increase in HDL attributed to the low-carbohydrate diet, which translated to an absolute increase of 4.6 mg/dL (95% confidence interval [CI], 1.5–8.1).1 A subsequent uncontrolled prospective trial was consistent with consistent with this systematic review and showed a 12% increase in HDL.2 The DASH diet was studied as an intervention in a RCT of 116 patients with the metabolic syndrome. Men responded with an in crease of 21% and women with an increase of 33%.3

 

 

 

Supplementation with several food additives and nutritional supplements has been tested. A systematic review with meta-analysis of 23 RCTs evaluating effect of various amounts of soy protein with isoflavones on lipid profile found a 3% increase in HDL with an absolute difference 1.5 mg/dL (95% CI, 0.0–2.8).4 Supplementation with standard multivitamins in a single small, crossover RCT showed a 31% increase in HDL.5

Many other strategies, supplements, and plans have been tested in different populations. Other than the above interventions, no other interventions raise HDL when subjected to meta-analysis or quality randomized trials (TABLE).

Recommendations from others

No specific guidelines on dietary therapy of HDL exist; however, the American Heart Association (AHA) published diet and lifestyle recommendations in 2006.14 These guidelines recommend a diet low in fat, saturated fat, trans fat, and cholesterol in addition to minimizing sodium, added sugars, and alcohol. The AHA also recommends for consumption of oily fish and the DASH diet.

Acknowledgments

The opinions and assertions contained herein are the private views of the author and not to be construed as official, or as reflecting the views of the US Air Force Medical Service or the US Air Force at large.

References

1. Gordon DJ, Knoke J, Probstfield JL, Superko R, Tyroler HA. High-density lipoprotein cholesterol and coronary heart disease in hypercholesterolemic men: the Lipid Research Clinics Coronary Primary Prevention Trial. Circulation 1986;74:1217-1225.

2. Williams PT. The relationships of vigorous exercise, alcohol, and adiposity to low and high high-density lipoproteincholesterol levels. Metabolism 2004;53:700-709.

3. Nordmann AJ, Nordmann A, Briel M, et al. Effects of low-carbohydrate vs low-fat diets on weight loss and cardiovascular risk factors: a meta-analysis of randomized controlled trials. Arch Intern Med 2006;166:285-293.

4. Wood RJ, Volek JS, Y Liu, NS Schacter, JH Contois, ML Fernandez. Carbohydrate restriction alters lipoprotein metabolism by modifying VLDL, LDL, and HDL subfraction distribution and size in overweight men. J Nutr 2006;136:384-389.

5. Azadbakht L, Mirmiran P, Esmaillzadeh A, Azizi T, Azizi F. Beneficial effects of a Dietary Approaches to Stop Hypertension eating plan on features of the metabolic syndrome. Diabetes Care 2005;28:2823-2831.

6. Zhan S, Ho SC. Meta-analysis of the effects of soy protein containing isoflavones on the lipid profile. Am J Clin Nutr 2005;81:397-408.

7. Morcos N. Increase in serum high-density lipoprotein following multivitamin and multimineral supplementation in adults with cardiovascular risk factors. Med Sci Res 1999;27:121-125.

8. Kelly S, Frost G, Whittaker V, Summerbell C. Low glycaemic index diets for coronary heart disease. Cochrane Database Syst Rev 2004;(4):CD004467.-

9. Brunner EJ, Thorogood M, Rees K, Hewitt G. Dietary advice for reducing cardiovascular risk. Cochrane Database Syst Rev 2005;(4):CD002128.-

10. Chen J, He J, Wildman RP, Reynolds K, Streiffer RH, Whelton PK. A randomized controlled trial of dietary fiber intake on serum lipids. Eur J Clin Nutr 2006;60:62-68.

11. Lewis A, Lookinland S, Beckstrand RL, Tiedeman ME. Treatment of hypertriglyceridemia with omega-3 fatty acids: A systematic review. J Am Acad Nurse Pract 2004;16:384-395.

12. Farmer A, Montori V, Dinneen S, Clar C. Fish oil in people with type 2 diabetes mellitus. Cochrane Database Syst Rev 2001;(3):CD003205.-

13. Hooper L, Thompson RL, Harrison RA, et al. Omega 3 fatty acids for prevention and treatment of cardiovascular disease. Cochrane Database Syst Rev 2004;(4):CD003177.-

14. Tapsell LC, Gillen LJ, Patch CS, et al. Including walnuts in a low-fat/modified-fat diet improves HDL cholesterol-to-total cholesterol ratios in patients with type 2 diabetes. Diabetes Care 2004;27:2777-2783.

15. Spiller GA, Jenkins DA, Bosello O, Gates JE, Cragen LN, Bruce B. Nuts and plasma lipids: an almond-based diet lowers LDL-C while preserving HDL-C. J Am Coll Nutr 1998;17:285-290.

16. Lichtenstein AH, Appel LJ, Brands M, et al. Diet and lifestyle recommendations revision 2006: A scientific statement from the American Heart Association Nutrition Committee. Circulation 2006;114:82-96.

References

1. Gordon DJ, Knoke J, Probstfield JL, Superko R, Tyroler HA. High-density lipoprotein cholesterol and coronary heart disease in hypercholesterolemic men: the Lipid Research Clinics Coronary Primary Prevention Trial. Circulation 1986;74:1217-1225.

2. Williams PT. The relationships of vigorous exercise, alcohol, and adiposity to low and high high-density lipoproteincholesterol levels. Metabolism 2004;53:700-709.

3. Nordmann AJ, Nordmann A, Briel M, et al. Effects of low-carbohydrate vs low-fat diets on weight loss and cardiovascular risk factors: a meta-analysis of randomized controlled trials. Arch Intern Med 2006;166:285-293.

4. Wood RJ, Volek JS, Y Liu, NS Schacter, JH Contois, ML Fernandez. Carbohydrate restriction alters lipoprotein metabolism by modifying VLDL, LDL, and HDL subfraction distribution and size in overweight men. J Nutr 2006;136:384-389.

5. Azadbakht L, Mirmiran P, Esmaillzadeh A, Azizi T, Azizi F. Beneficial effects of a Dietary Approaches to Stop Hypertension eating plan on features of the metabolic syndrome. Diabetes Care 2005;28:2823-2831.

6. Zhan S, Ho SC. Meta-analysis of the effects of soy protein containing isoflavones on the lipid profile. Am J Clin Nutr 2005;81:397-408.

7. Morcos N. Increase in serum high-density lipoprotein following multivitamin and multimineral supplementation in adults with cardiovascular risk factors. Med Sci Res 1999;27:121-125.

8. Kelly S, Frost G, Whittaker V, Summerbell C. Low glycaemic index diets for coronary heart disease. Cochrane Database Syst Rev 2004;(4):CD004467.-

9. Brunner EJ, Thorogood M, Rees K, Hewitt G. Dietary advice for reducing cardiovascular risk. Cochrane Database Syst Rev 2005;(4):CD002128.-

10. Chen J, He J, Wildman RP, Reynolds K, Streiffer RH, Whelton PK. A randomized controlled trial of dietary fiber intake on serum lipids. Eur J Clin Nutr 2006;60:62-68.

11. Lewis A, Lookinland S, Beckstrand RL, Tiedeman ME. Treatment of hypertriglyceridemia with omega-3 fatty acids: A systematic review. J Am Acad Nurse Pract 2004;16:384-395.

12. Farmer A, Montori V, Dinneen S, Clar C. Fish oil in people with type 2 diabetes mellitus. Cochrane Database Syst Rev 2001;(3):CD003205.-

13. Hooper L, Thompson RL, Harrison RA, et al. Omega 3 fatty acids for prevention and treatment of cardiovascular disease. Cochrane Database Syst Rev 2004;(4):CD003177.-

14. Tapsell LC, Gillen LJ, Patch CS, et al. Including walnuts in a low-fat/modified-fat diet improves HDL cholesterol-to-total cholesterol ratios in patients with type 2 diabetes. Diabetes Care 2004;27:2777-2783.

15. Spiller GA, Jenkins DA, Bosello O, Gates JE, Cragen LN, Bruce B. Nuts and plasma lipids: an almond-based diet lowers LDL-C while preserving HDL-C. J Am Coll Nutr 1998;17:285-290.

16. Lichtenstein AH, Appel LJ, Brands M, et al. Diet and lifestyle recommendations revision 2006: A scientific statement from the American Heart Association Nutrition Committee. Circulation 2006;114:82-96.

Issue
The Journal of Family Practice - 55(12)
Issue
The Journal of Family Practice - 55(12)
Page Number
1076-1078
Page Number
1076-1078
Publications
Publications
Topics
Article Type
Display Headline
What is the dietary treatment for low HDL cholesterol?
Display Headline
What is the dietary treatment for low HDL cholesterol?
Sections
PURLs Copyright

Evidence-based answers from the Family Physicians Inquiries Network

Disallow All Ads
Alternative CME
Article PDF Media

Does stopping a statin increase the short-term risk of a cardiovascular event?

Article Type
Changed
Mon, 01/14/2019 - 11:16
Display Headline
Does stopping a statin increase the short-term risk of a cardiovascular event?
EVIDENCE-BASED ANSWER

When hydroxymethyl glutaryl coenzyme A (HMG CoA)inhibitors (statins) are stopped by asymptomatic patients, there appears to be no increased risk of cardiovascular events (strength of recommendation [SOR]: B,). However, for patients who have recently experienced a cardiovascular event, discontinuation of statins increases the risk of further events and death (SOR: B,).

CLINICAL COMMENTARY

Rely on low-tech skills, like shared decision-making, to improve adherence
Vincent Lo, MD
San Joaquin Family Medicine Residency, San Joaquin General Hospital, French Camp, Calif

One might hope that all patients taking statins would have excellent compliance, given these drugs’ well-established benefit. Unfortunately, long-term adherence remains suboptimal, and patients are going to stop their statins.1 A Canadian study2 found that patients aged >65 years, with and without recent acute coronary syndrome (ACS), had low rates of adherence to statins 2 years after initiation of therapy (40.1% for ACS, 36.1% for chronic coronary artery disease, and 25.4% for primary prevention). A recent Cochrane review3 found small improved adherence despite attempted intervention (range improvement: −3% to 25%). They concluded no intervention aimed at improving adherence to lipid-lowing drugs can be recommended over another, given the limited effects.

Clinicians are left to rely on low-tech skills, such as focusing on the patient’s perspective and shared decision-making, to improve their patients’ adherence. This focus is especially important for those who had a recent cardiovascular event. Nevertheless, it is reassuring that this review did not find significantly increased harm after abrupt stopping of statins among stable patients without recent ACS.

 

Evidence summary

The benefits of statin therapy appear to extend beyond the realm of their cholesterol-lowering properties.4 These benefits, such as reduction in post—myocardial infarction (MI) deaths and reinfarctions, are seen quickly after initiation of therapy. Other drugs, such as aspirin and beta-blockers, have also been shown to improve early outcomes when started after cardiovascular events, although waiting until the patient is hemodynamically stable to initiate beta-blockade reduces the risk of cardiogenic shock.5 If standard agents are either not started or withdrawn after a cardiovascular event, patients are at increased risk of harm.6,7

Physiological research. Studies of patients with stroke and those with only risk factors for cardiovascular disease show that platelet activity is increased when statins are discontinued.8,9 Additionally, tissue plasminogen activator levels are decreased after discontinuation of statins, resulting in a relatively hypercoagulable state.10 Animal studies of stroke in mice showed mice whose statin was abruptly stopped had more damage from stroke than those whose statin was continued.11

High-risk cardiovascular patients. Preliminary human data suggested that stopping statins increased risk of recurrent events for patients who recently had a primary cardiovascular event. One retrospective case-control study evaluated 4870 patients who had statin therapy withdrawn on admission to the hospital for non-ST segment elevation MI (NSTEMI). Patients who had their statins withheld had increased rates of heart failure, arrhythmia, shock, and death (hazard ratio=2.32; 95% confidence interval [CI], 2.02–2.67).12 A post-hoc analysis of data from the PRISM trial found that among the 86 patients who were admitted for chest pain and had their statin withdrawn, a higher rate of death and nonfatal MI was observed, compared with the 379 patients whose statins were continued (hazard ratio=2.93; 95% CI, 1.64–6.27). This effect was seen in the first week and was independent of cholesterol levels and measures of severity of illness.13

Low-risk cardiovascular patients. A post-hoc analysis of the washout period of a prospective study of 9473 asymptomatic outpatients who were previously taking statins showed that for these lower-risk patients, similar rates of cardiovascular events could be expected during withdrawal (any statin) or initiation of atorvastatin therapy. The monthly event rate during the discontinuation phase was 0.20% and during initiation was 0.26% (P=NS).14

Recommendations from others

Currently, no expert panels or specialty bodies make recommendations regarding how or when to discontinue statins. The Institute for Clinical Systems Improvement recommends that all patients with chronic stable coronary artery disease should be considered for statin use regardless of their lipid levels; however, no mention of discontinuing statins is made.15

References

1. Benner JS, Glynn RJ, Neumann PJ, Mogun H, Weinsten MC, Avorn J. Long-term persistence in use of statin therapy in elderly patients. JAMA 2002;288:455-461.

2. Jackevicius CA, Mamdani M, Tu JV. Adherence with statin therapy in elderly patients with and without acute coronary syndromes. JAMA 2002;288:462-467.

3. Schedlbauer A, Schroeder K, Peters TJ, Fahey T. Interventions to improve adherence to lipid lowering medication. Cochrane Database Syst Rev 2004;(4)CD004371.

4. Thompson PL, Meredeth I, Amerena J, et al. Effect of pravastatin compared with placebo initiated within 24 hours of onset of acute myocardial infarction or unstable angina: the Pravastatin in Acute Coronary Treatment (PACT) trial. Am Heart J 2004;148:e2.

5. Chen ZM, Pan HC, Chen YP, et al. Early intravenous then oral metoprolol in 45,852 patients with acute myocardial infarction: randomised placebo-controlled trial. Lancet 2005;366:1622-1632.

6. Olsson G, Oden A, Johansson L, Sjogren A, Rehnqvist N. Prognosis after withdrawal of chronic postinfarction metoprolol treatment: a 2-7 year follow-up. Eur Heart J 1988;9:365-372.

7. ISIS-2 (Second International Study of Infarct Survival) Collaborative Group. Randomised trial of intravenous streptokinase, oral aspirin, both, or neither among 17,187 cases of suspected acute myocardial infarction: ISIS-2. Lancet 1988; 2(8607):349–360.

8. Puccetti L, Pasqui AL, Pastorelli M, et al. Platelet hyperactivity after statin treatment discontinuation. Thromb Haemost 2003;90:476-482.

9. Cha JK, Jeong MH, Kim JW. Statin reduces the platelet P-selectin expression in atherosclerotic ischemic stroke. J Thrombosis Thrombolysis 2004;18:39-42.

10. Lai WT, Lee KT, Chu CS, et al. Influence of withdrawal of statin treatment on proinflammatory response and fibrinolytic activity in humans: an effect independent on cholesterol elevation. Int J Cardiol 2005;98:459-464.

11. Gertz K, Laufs U, Lindauer U, et al. Withdrawal of statin treatment abrogates stroke protection in mice. Stroke 2003;34:551-557.

12. Spencer FA, Fonarow GC, Frederick PD, et al. Early withdrawal of statin therapy in patients with non-ST-segment elevation myocardial infarction: national registry of myocardial infarction. Arch Intern Med 2004;164:2162-2168.

13. Heeschen C, Hamm CW, Laufs U, et al. Withdrawal of statins increases event rates in patients with acute coronary syndromes. Circulation 2002;105:1446-1452.

14. McGowan MP. and the Treating to New Target (TNT) Study Group. There is no evidence for an increase in acute coronary syndromes after short-term abrupt discontinuation of statins in stable cardiac patients. Circulation 2004;110:2333-2335.

15. Stable coronary artery disease. Institute for Clinical Systems Improvement. Bloomington, Minn: Institute for Clinical Systems Improvement (ICSI); July 1994. Revised April 2005. Available at: www.icsi.org/knowledge/detail.asp?catID=29&itemID=192. Accessed on May 17, 2006.

Article PDF
Author and Disclosure Information

Paul Crawford, MD
Kristin Hitchcock, MSI
Eglin Air Force Base Family Medicine Residency, Eglin AFB, Fla
Department of Family Medicine, The University of Chicago

Issue
The Journal of Family Practice - 55(6)
Publications
Topics
Page Number
533-534
Sections
Author and Disclosure Information

Paul Crawford, MD
Kristin Hitchcock, MSI
Eglin Air Force Base Family Medicine Residency, Eglin AFB, Fla
Department of Family Medicine, The University of Chicago

Author and Disclosure Information

Paul Crawford, MD
Kristin Hitchcock, MSI
Eglin Air Force Base Family Medicine Residency, Eglin AFB, Fla
Department of Family Medicine, The University of Chicago

Article PDF
Article PDF
EVIDENCE-BASED ANSWER

When hydroxymethyl glutaryl coenzyme A (HMG CoA)inhibitors (statins) are stopped by asymptomatic patients, there appears to be no increased risk of cardiovascular events (strength of recommendation [SOR]: B,). However, for patients who have recently experienced a cardiovascular event, discontinuation of statins increases the risk of further events and death (SOR: B,).

CLINICAL COMMENTARY

Rely on low-tech skills, like shared decision-making, to improve adherence
Vincent Lo, MD
San Joaquin Family Medicine Residency, San Joaquin General Hospital, French Camp, Calif

One might hope that all patients taking statins would have excellent compliance, given these drugs’ well-established benefit. Unfortunately, long-term adherence remains suboptimal, and patients are going to stop their statins.1 A Canadian study2 found that patients aged >65 years, with and without recent acute coronary syndrome (ACS), had low rates of adherence to statins 2 years after initiation of therapy (40.1% for ACS, 36.1% for chronic coronary artery disease, and 25.4% for primary prevention). A recent Cochrane review3 found small improved adherence despite attempted intervention (range improvement: −3% to 25%). They concluded no intervention aimed at improving adherence to lipid-lowing drugs can be recommended over another, given the limited effects.

Clinicians are left to rely on low-tech skills, such as focusing on the patient’s perspective and shared decision-making, to improve their patients’ adherence. This focus is especially important for those who had a recent cardiovascular event. Nevertheless, it is reassuring that this review did not find significantly increased harm after abrupt stopping of statins among stable patients without recent ACS.

 

Evidence summary

The benefits of statin therapy appear to extend beyond the realm of their cholesterol-lowering properties.4 These benefits, such as reduction in post—myocardial infarction (MI) deaths and reinfarctions, are seen quickly after initiation of therapy. Other drugs, such as aspirin and beta-blockers, have also been shown to improve early outcomes when started after cardiovascular events, although waiting until the patient is hemodynamically stable to initiate beta-blockade reduces the risk of cardiogenic shock.5 If standard agents are either not started or withdrawn after a cardiovascular event, patients are at increased risk of harm.6,7

Physiological research. Studies of patients with stroke and those with only risk factors for cardiovascular disease show that platelet activity is increased when statins are discontinued.8,9 Additionally, tissue plasminogen activator levels are decreased after discontinuation of statins, resulting in a relatively hypercoagulable state.10 Animal studies of stroke in mice showed mice whose statin was abruptly stopped had more damage from stroke than those whose statin was continued.11

High-risk cardiovascular patients. Preliminary human data suggested that stopping statins increased risk of recurrent events for patients who recently had a primary cardiovascular event. One retrospective case-control study evaluated 4870 patients who had statin therapy withdrawn on admission to the hospital for non-ST segment elevation MI (NSTEMI). Patients who had their statins withheld had increased rates of heart failure, arrhythmia, shock, and death (hazard ratio=2.32; 95% confidence interval [CI], 2.02–2.67).12 A post-hoc analysis of data from the PRISM trial found that among the 86 patients who were admitted for chest pain and had their statin withdrawn, a higher rate of death and nonfatal MI was observed, compared with the 379 patients whose statins were continued (hazard ratio=2.93; 95% CI, 1.64–6.27). This effect was seen in the first week and was independent of cholesterol levels and measures of severity of illness.13

Low-risk cardiovascular patients. A post-hoc analysis of the washout period of a prospective study of 9473 asymptomatic outpatients who were previously taking statins showed that for these lower-risk patients, similar rates of cardiovascular events could be expected during withdrawal (any statin) or initiation of atorvastatin therapy. The monthly event rate during the discontinuation phase was 0.20% and during initiation was 0.26% (P=NS).14

Recommendations from others

Currently, no expert panels or specialty bodies make recommendations regarding how or when to discontinue statins. The Institute for Clinical Systems Improvement recommends that all patients with chronic stable coronary artery disease should be considered for statin use regardless of their lipid levels; however, no mention of discontinuing statins is made.15

EVIDENCE-BASED ANSWER

When hydroxymethyl glutaryl coenzyme A (HMG CoA)inhibitors (statins) are stopped by asymptomatic patients, there appears to be no increased risk of cardiovascular events (strength of recommendation [SOR]: B,). However, for patients who have recently experienced a cardiovascular event, discontinuation of statins increases the risk of further events and death (SOR: B,).

CLINICAL COMMENTARY

Rely on low-tech skills, like shared decision-making, to improve adherence
Vincent Lo, MD
San Joaquin Family Medicine Residency, San Joaquin General Hospital, French Camp, Calif

One might hope that all patients taking statins would have excellent compliance, given these drugs’ well-established benefit. Unfortunately, long-term adherence remains suboptimal, and patients are going to stop their statins.1 A Canadian study2 found that patients aged >65 years, with and without recent acute coronary syndrome (ACS), had low rates of adherence to statins 2 years after initiation of therapy (40.1% for ACS, 36.1% for chronic coronary artery disease, and 25.4% for primary prevention). A recent Cochrane review3 found small improved adherence despite attempted intervention (range improvement: −3% to 25%). They concluded no intervention aimed at improving adherence to lipid-lowing drugs can be recommended over another, given the limited effects.

Clinicians are left to rely on low-tech skills, such as focusing on the patient’s perspective and shared decision-making, to improve their patients’ adherence. This focus is especially important for those who had a recent cardiovascular event. Nevertheless, it is reassuring that this review did not find significantly increased harm after abrupt stopping of statins among stable patients without recent ACS.

 

Evidence summary

The benefits of statin therapy appear to extend beyond the realm of their cholesterol-lowering properties.4 These benefits, such as reduction in post—myocardial infarction (MI) deaths and reinfarctions, are seen quickly after initiation of therapy. Other drugs, such as aspirin and beta-blockers, have also been shown to improve early outcomes when started after cardiovascular events, although waiting until the patient is hemodynamically stable to initiate beta-blockade reduces the risk of cardiogenic shock.5 If standard agents are either not started or withdrawn after a cardiovascular event, patients are at increased risk of harm.6,7

Physiological research. Studies of patients with stroke and those with only risk factors for cardiovascular disease show that platelet activity is increased when statins are discontinued.8,9 Additionally, tissue plasminogen activator levels are decreased after discontinuation of statins, resulting in a relatively hypercoagulable state.10 Animal studies of stroke in mice showed mice whose statin was abruptly stopped had more damage from stroke than those whose statin was continued.11

High-risk cardiovascular patients. Preliminary human data suggested that stopping statins increased risk of recurrent events for patients who recently had a primary cardiovascular event. One retrospective case-control study evaluated 4870 patients who had statin therapy withdrawn on admission to the hospital for non-ST segment elevation MI (NSTEMI). Patients who had their statins withheld had increased rates of heart failure, arrhythmia, shock, and death (hazard ratio=2.32; 95% confidence interval [CI], 2.02–2.67).12 A post-hoc analysis of data from the PRISM trial found that among the 86 patients who were admitted for chest pain and had their statin withdrawn, a higher rate of death and nonfatal MI was observed, compared with the 379 patients whose statins were continued (hazard ratio=2.93; 95% CI, 1.64–6.27). This effect was seen in the first week and was independent of cholesterol levels and measures of severity of illness.13

Low-risk cardiovascular patients. A post-hoc analysis of the washout period of a prospective study of 9473 asymptomatic outpatients who were previously taking statins showed that for these lower-risk patients, similar rates of cardiovascular events could be expected during withdrawal (any statin) or initiation of atorvastatin therapy. The monthly event rate during the discontinuation phase was 0.20% and during initiation was 0.26% (P=NS).14

Recommendations from others

Currently, no expert panels or specialty bodies make recommendations regarding how or when to discontinue statins. The Institute for Clinical Systems Improvement recommends that all patients with chronic stable coronary artery disease should be considered for statin use regardless of their lipid levels; however, no mention of discontinuing statins is made.15

References

1. Benner JS, Glynn RJ, Neumann PJ, Mogun H, Weinsten MC, Avorn J. Long-term persistence in use of statin therapy in elderly patients. JAMA 2002;288:455-461.

2. Jackevicius CA, Mamdani M, Tu JV. Adherence with statin therapy in elderly patients with and without acute coronary syndromes. JAMA 2002;288:462-467.

3. Schedlbauer A, Schroeder K, Peters TJ, Fahey T. Interventions to improve adherence to lipid lowering medication. Cochrane Database Syst Rev 2004;(4)CD004371.

4. Thompson PL, Meredeth I, Amerena J, et al. Effect of pravastatin compared with placebo initiated within 24 hours of onset of acute myocardial infarction or unstable angina: the Pravastatin in Acute Coronary Treatment (PACT) trial. Am Heart J 2004;148:e2.

5. Chen ZM, Pan HC, Chen YP, et al. Early intravenous then oral metoprolol in 45,852 patients with acute myocardial infarction: randomised placebo-controlled trial. Lancet 2005;366:1622-1632.

6. Olsson G, Oden A, Johansson L, Sjogren A, Rehnqvist N. Prognosis after withdrawal of chronic postinfarction metoprolol treatment: a 2-7 year follow-up. Eur Heart J 1988;9:365-372.

7. ISIS-2 (Second International Study of Infarct Survival) Collaborative Group. Randomised trial of intravenous streptokinase, oral aspirin, both, or neither among 17,187 cases of suspected acute myocardial infarction: ISIS-2. Lancet 1988; 2(8607):349–360.

8. Puccetti L, Pasqui AL, Pastorelli M, et al. Platelet hyperactivity after statin treatment discontinuation. Thromb Haemost 2003;90:476-482.

9. Cha JK, Jeong MH, Kim JW. Statin reduces the platelet P-selectin expression in atherosclerotic ischemic stroke. J Thrombosis Thrombolysis 2004;18:39-42.

10. Lai WT, Lee KT, Chu CS, et al. Influence of withdrawal of statin treatment on proinflammatory response and fibrinolytic activity in humans: an effect independent on cholesterol elevation. Int J Cardiol 2005;98:459-464.

11. Gertz K, Laufs U, Lindauer U, et al. Withdrawal of statin treatment abrogates stroke protection in mice. Stroke 2003;34:551-557.

12. Spencer FA, Fonarow GC, Frederick PD, et al. Early withdrawal of statin therapy in patients with non-ST-segment elevation myocardial infarction: national registry of myocardial infarction. Arch Intern Med 2004;164:2162-2168.

13. Heeschen C, Hamm CW, Laufs U, et al. Withdrawal of statins increases event rates in patients with acute coronary syndromes. Circulation 2002;105:1446-1452.

14. McGowan MP. and the Treating to New Target (TNT) Study Group. There is no evidence for an increase in acute coronary syndromes after short-term abrupt discontinuation of statins in stable cardiac patients. Circulation 2004;110:2333-2335.

15. Stable coronary artery disease. Institute for Clinical Systems Improvement. Bloomington, Minn: Institute for Clinical Systems Improvement (ICSI); July 1994. Revised April 2005. Available at: www.icsi.org/knowledge/detail.asp?catID=29&itemID=192. Accessed on May 17, 2006.

References

1. Benner JS, Glynn RJ, Neumann PJ, Mogun H, Weinsten MC, Avorn J. Long-term persistence in use of statin therapy in elderly patients. JAMA 2002;288:455-461.

2. Jackevicius CA, Mamdani M, Tu JV. Adherence with statin therapy in elderly patients with and without acute coronary syndromes. JAMA 2002;288:462-467.

3. Schedlbauer A, Schroeder K, Peters TJ, Fahey T. Interventions to improve adherence to lipid lowering medication. Cochrane Database Syst Rev 2004;(4)CD004371.

4. Thompson PL, Meredeth I, Amerena J, et al. Effect of pravastatin compared with placebo initiated within 24 hours of onset of acute myocardial infarction or unstable angina: the Pravastatin in Acute Coronary Treatment (PACT) trial. Am Heart J 2004;148:e2.

5. Chen ZM, Pan HC, Chen YP, et al. Early intravenous then oral metoprolol in 45,852 patients with acute myocardial infarction: randomised placebo-controlled trial. Lancet 2005;366:1622-1632.

6. Olsson G, Oden A, Johansson L, Sjogren A, Rehnqvist N. Prognosis after withdrawal of chronic postinfarction metoprolol treatment: a 2-7 year follow-up. Eur Heart J 1988;9:365-372.

7. ISIS-2 (Second International Study of Infarct Survival) Collaborative Group. Randomised trial of intravenous streptokinase, oral aspirin, both, or neither among 17,187 cases of suspected acute myocardial infarction: ISIS-2. Lancet 1988; 2(8607):349–360.

8. Puccetti L, Pasqui AL, Pastorelli M, et al. Platelet hyperactivity after statin treatment discontinuation. Thromb Haemost 2003;90:476-482.

9. Cha JK, Jeong MH, Kim JW. Statin reduces the platelet P-selectin expression in atherosclerotic ischemic stroke. J Thrombosis Thrombolysis 2004;18:39-42.

10. Lai WT, Lee KT, Chu CS, et al. Influence of withdrawal of statin treatment on proinflammatory response and fibrinolytic activity in humans: an effect independent on cholesterol elevation. Int J Cardiol 2005;98:459-464.

11. Gertz K, Laufs U, Lindauer U, et al. Withdrawal of statin treatment abrogates stroke protection in mice. Stroke 2003;34:551-557.

12. Spencer FA, Fonarow GC, Frederick PD, et al. Early withdrawal of statin therapy in patients with non-ST-segment elevation myocardial infarction: national registry of myocardial infarction. Arch Intern Med 2004;164:2162-2168.

13. Heeschen C, Hamm CW, Laufs U, et al. Withdrawal of statins increases event rates in patients with acute coronary syndromes. Circulation 2002;105:1446-1452.

14. McGowan MP. and the Treating to New Target (TNT) Study Group. There is no evidence for an increase in acute coronary syndromes after short-term abrupt discontinuation of statins in stable cardiac patients. Circulation 2004;110:2333-2335.

15. Stable coronary artery disease. Institute for Clinical Systems Improvement. Bloomington, Minn: Institute for Clinical Systems Improvement (ICSI); July 1994. Revised April 2005. Available at: www.icsi.org/knowledge/detail.asp?catID=29&itemID=192. Accessed on May 17, 2006.

Issue
The Journal of Family Practice - 55(6)
Issue
The Journal of Family Practice - 55(6)
Page Number
533-534
Page Number
533-534
Publications
Publications
Topics
Article Type
Display Headline
Does stopping a statin increase the short-term risk of a cardiovascular event?
Display Headline
Does stopping a statin increase the short-term risk of a cardiovascular event?
Sections
PURLs Copyright

Evidence-based answers from the Family Physicians Inquiries Network

Disallow All Ads
Alternative CME
Article PDF Media

How safe is vaginal birth after cesarean section for the mother and fetus?

Article Type
Changed
Mon, 01/14/2019 - 11:16
Display Headline
How safe is vaginal birth after cesarean section for the mother and fetus?
EVIDENCE-BASED ANSWER

Compared with planned repeat low-transverse cesarean section, vaginal birth after cesarean section (VBAC) is not associated with increased risk of maternal or neonatal mortality (strength of recommendation [SOR]: B). Morbidity is slightly increased, as evidenced by higher uterine rupture rates and some neonatal outcome measures (SOR: B).

CLINICAL COMMENTARY

Risks of C-section and labor must be considered when counseling regarding route of delivery
Lynda DeArmond, MD
Waco Family Practice Residency Program, Waco, Texas

Another question to pose is: how safe is repeat cesarean section for the mother and fetus? How much do morbidity and mortality increase with each new intra-abdominal procedure? Each time the belly is opened there is new scar, with increased likelihood of adhesions and potential for future bowel obstruction. Consider these risks when counseling regarding route of delivery. Risk of uterine rupture appears to be higher in trials of labor (and confers a statistically significant but small increase in morbidity but not mortality). However, the uterine scar can silently fail without labor—as is sometimes discovered at a scheduled repeat section, usually without untoward effects on mother or fetus.

Remember that you are sending a young woman home with a new baby to care for (along with other children) and a major abdominal procedure (through an old scar) to recover from, which one could certainly define as morbidity. Cesarean section is an important tool, but we must be careful to practice best possible care and consider all patient factors and preferences. And data are still lacking to support the notion that VBAC is unsafe.

 

Evidence summary

Contrary to the goals of Healthy People 2010, the rate of cesarean sections is increasing.1 The repeat cesarean rate for low-risk women of all ages and racial groups is now 88.7%, the highest rate since the Centers for Disease Control and Prevention (CDC) began tracking the statistic in 1989. Is VBAC safe, or is a trial of labor no longer supported by the data?

The most recent Cochrane Review found that both VBAC and repeat lowtransverse cesarean section have benefits and risks associated with them; however, after reviewing the limited data, they concluded that no trial exists to adequately help women and their caregivers make an informed decision between the two.2 A strong theme in the Cochrane Review, echoed in most reviews, was the absence of high-quality prospective randomized data.

In an attempt to quantify the risks of VBAC, a systematic review determined that attempted VBAC, compared with repeat low-transverse cesarean section, increased the risk of uterine rupture by 2.7 per 1000 cases (95% confidence interval [CI], 0.73–4.73).3 This additional risk rate is often quoted in VBAC reviews and was cited in the Agency for Healthcare Research and Quality evidence report; it is based on 1 prospective, nonrandomized cohort trial and 1 retrospective cohort study.4,5

No randomized controlled trials exist for determining maternal safety of VBAC, although another recent systematic review found 2 nonrandomized prospective trials of sufficient quality to analyze. The authors concluded there were “no statistically significant differences between planned elective repeat cesarean section and planned VBAC.”6 Upon closer review in PubMed, one of the cited studies did not study 312 patients for VBAC outcomes as alleged; rather, it investigated patient attitudes towards VBAC.7

Since publication of that review, a large, multicenter, prospective, nonrandomized trial involving 33,699 patients found no significant difference between VBAC and planned cesarean for hysterectomy (0.2% vs 0.3%; odds ratio [OR]=0.77; 95% CI, 0.51–1.17), maternal death (0.02% vs 0.04%; OR=0.38; 95% CI, 0.1–1.46), and neonatal death (0.08% vs 0.05%; OR=1.82; 95% CI, 0.73–4.57).8 Significant associations were found for uterine rupture rates in spontaneous labor (24/6685 [0.4%] vs no cases; number needed to harm [NNH]=279) and neonatal hypoxic-ischemic encephalopathy (0.46 cases per 1000 vs no cases; NNH=2174).8

A retrospective Canadian cohort trial of 308,755 women also demonstrated an association of VBAC with uterine rupture(0.65% of trial-of-labor cases; OR=2.38; 95% CI, 2.12–2.67), and a trend towards higher maternal mortality in the cesarean group (1.6 per 100,000 for VBAC vs 5.6 per 100,000 for planned cesarean; OR=0.32; 95% CI, 0.07–1.47).9

The effect of VBAC on neonatal morbidity and mortality is unclear. In contrast to the negative larger trial,8 a smaller retrospective cohort of 24,529 births found a higher association of perinatal death for trial of labor (adjusted OR=11.7; 95% CI, 1.4–101.6).10 The perinatal death rate was similar to rates in nulliparous women. Regarding morbidity, one retrospective cohort trial showed VBAC was associated with an increase in neonatal sepsis (1% vs 0%; CI not given) compared with planned cesarean, but VBAC resulted in less transient tachypnea (5% vs 7%) and hyper-bilirubinemia (2% vs 6%).11

 

 

 

Recommendations from others

Both the American College of Obstetricians and Gynecologists and the Society of Obstetricians and Gynecologists of Canada state that women with 1 previous low-transverse cesarean section should be offered a trial of labor after appropriate counseling of the risks and benefits.12,13 Furthermore, induction with oxytocin is allowed, but the use of prostaglandins is not recommended. Based on expert opinion, both organizations encourage VBAC only in institutions staffed with surgeons and anesthesiologists immediately available to provide emergent cesarean.

Acknowledgments

The opinions and assertions contained herein are the private views of the author and are not to be construed as official, or as reflecting the views of the US Air Force medical department or the US Air Force at large.

References

1. Menacker F. Trends in cesarean rates for first births and repeat cesarean rates for low-risk women: United States, 1990-2003. Natl Vital Stat Rep 2005;54:1-8.

2. Dodd J, Crowther C, Huertas E, Guise J, Horey D. Planned elective repeat caesarean section versus planned vaginal birth for women with a previous caesarean birth. Cochrane Database Syst Rev 2005;(4):CD004224.

3. Guise J, McDonagh M, Osterweil P, Nygren P, Chan B, Helfand M. Systematic review of the incidence and consequences of uterine rupture in women with previous caesarean section. BMJ 2004;329:19-25.

4. Vaginal Birth After Cesarean (VBAC). Summary. Evidence Report/Technology Assessment, No. 71. AHRQ publication 03-E017. Rockville, Md: Agency for Healthcare Research and Quality; 2003. Available at: www.ahrq.gov/clinic/epcsums/vbacsum.htm. Accessed on January 9, 2006.

5. McMahon MJ, Luther ER, Bowes WA, Olshan AF. Comparison of a trial of labor with an elective second cesarean section. N Engl J Med 1996;335:689-695.

6. Dodd J, Crowther C. Vaginal birth after Caesarean versus elective repeat Caesarean for women with a single prior Caesarean birth: a systematic review of the literature. Aust N Z J Obstet Gynaecol 2004;44:387-391.

7. Abitbol MM, Castillo I, Taylor UB, Rochelson BL, Shmoys S, Monheit AG. Vaginal birth after cesarean section: the patient’s point of view. Am Fam Physician 1993;47:129-134.

8. Landon M, Hauth J, Leveno K, et al. for the National Institute of Child Health and Human Development Maternal-Fetal Medicine Units Network. Maternal and perinatal outcomes associated with a trial of labor after prior cesarean delivery. N Engl J Med 2004;351:2581-2589.

9. Wen S, Rusen I, Walker M, et al. Comparison of maternal mortality and morbidity between trial of labor and elective cesarean section among women with previous cesarean delivery. Am J Obstet Gynecol 2004;191:1263-1269.

10. Smith G, Pell J, Cameron A, Dobbie R. Risk of perinatal death associated with labor after previous cesarean delivery in uncomplicated term pregnancies. JAMA 2002;287:2684-2690.

11. Hook B, Kiwi R, Amini S, Fanaroff A, Hack M. Neonatal morbidity after elective repeat cesarean section and trial of labor. Pediatrics 1997;100:348-353.

12. American College of Obstetricians and Gynecologists Committee on Practice Bulletins—Obstetrics. ACOG Practice Bulletin. Vaginal Birth After Cesarean Delivery. Obstet Gynecol 2004;104:203-212.

13. Martel M, MacKinnon C, and the Clinical Practice Obstetrics Committee, Society of Obstetricians and Gynaecologists of Canada. Guidelines for vaginal birth after previous Caesarean birth. J Obstet Gynaecol Can 2005;27:164-188.

Article PDF
Author and Disclosure Information

Paul Crawford, MD
Eglin Air Force Base Family Medicine Residency, Eglin AFB, Florida

Leonora Kaufmann, MLIS
Carolinas Health Care System, Charlotte, NC

Issue
The Journal of Family Practice - 55(2)
Publications
Topics
Page Number
149-151
Sections
Author and Disclosure Information

Paul Crawford, MD
Eglin Air Force Base Family Medicine Residency, Eglin AFB, Florida

Leonora Kaufmann, MLIS
Carolinas Health Care System, Charlotte, NC

Author and Disclosure Information

Paul Crawford, MD
Eglin Air Force Base Family Medicine Residency, Eglin AFB, Florida

Leonora Kaufmann, MLIS
Carolinas Health Care System, Charlotte, NC

Article PDF
Article PDF
EVIDENCE-BASED ANSWER

Compared with planned repeat low-transverse cesarean section, vaginal birth after cesarean section (VBAC) is not associated with increased risk of maternal or neonatal mortality (strength of recommendation [SOR]: B). Morbidity is slightly increased, as evidenced by higher uterine rupture rates and some neonatal outcome measures (SOR: B).

CLINICAL COMMENTARY

Risks of C-section and labor must be considered when counseling regarding route of delivery
Lynda DeArmond, MD
Waco Family Practice Residency Program, Waco, Texas

Another question to pose is: how safe is repeat cesarean section for the mother and fetus? How much do morbidity and mortality increase with each new intra-abdominal procedure? Each time the belly is opened there is new scar, with increased likelihood of adhesions and potential for future bowel obstruction. Consider these risks when counseling regarding route of delivery. Risk of uterine rupture appears to be higher in trials of labor (and confers a statistically significant but small increase in morbidity but not mortality). However, the uterine scar can silently fail without labor—as is sometimes discovered at a scheduled repeat section, usually without untoward effects on mother or fetus.

Remember that you are sending a young woman home with a new baby to care for (along with other children) and a major abdominal procedure (through an old scar) to recover from, which one could certainly define as morbidity. Cesarean section is an important tool, but we must be careful to practice best possible care and consider all patient factors and preferences. And data are still lacking to support the notion that VBAC is unsafe.

 

Evidence summary

Contrary to the goals of Healthy People 2010, the rate of cesarean sections is increasing.1 The repeat cesarean rate for low-risk women of all ages and racial groups is now 88.7%, the highest rate since the Centers for Disease Control and Prevention (CDC) began tracking the statistic in 1989. Is VBAC safe, or is a trial of labor no longer supported by the data?

The most recent Cochrane Review found that both VBAC and repeat lowtransverse cesarean section have benefits and risks associated with them; however, after reviewing the limited data, they concluded that no trial exists to adequately help women and their caregivers make an informed decision between the two.2 A strong theme in the Cochrane Review, echoed in most reviews, was the absence of high-quality prospective randomized data.

In an attempt to quantify the risks of VBAC, a systematic review determined that attempted VBAC, compared with repeat low-transverse cesarean section, increased the risk of uterine rupture by 2.7 per 1000 cases (95% confidence interval [CI], 0.73–4.73).3 This additional risk rate is often quoted in VBAC reviews and was cited in the Agency for Healthcare Research and Quality evidence report; it is based on 1 prospective, nonrandomized cohort trial and 1 retrospective cohort study.4,5

No randomized controlled trials exist for determining maternal safety of VBAC, although another recent systematic review found 2 nonrandomized prospective trials of sufficient quality to analyze. The authors concluded there were “no statistically significant differences between planned elective repeat cesarean section and planned VBAC.”6 Upon closer review in PubMed, one of the cited studies did not study 312 patients for VBAC outcomes as alleged; rather, it investigated patient attitudes towards VBAC.7

Since publication of that review, a large, multicenter, prospective, nonrandomized trial involving 33,699 patients found no significant difference between VBAC and planned cesarean for hysterectomy (0.2% vs 0.3%; odds ratio [OR]=0.77; 95% CI, 0.51–1.17), maternal death (0.02% vs 0.04%; OR=0.38; 95% CI, 0.1–1.46), and neonatal death (0.08% vs 0.05%; OR=1.82; 95% CI, 0.73–4.57).8 Significant associations were found for uterine rupture rates in spontaneous labor (24/6685 [0.4%] vs no cases; number needed to harm [NNH]=279) and neonatal hypoxic-ischemic encephalopathy (0.46 cases per 1000 vs no cases; NNH=2174).8

A retrospective Canadian cohort trial of 308,755 women also demonstrated an association of VBAC with uterine rupture(0.65% of trial-of-labor cases; OR=2.38; 95% CI, 2.12–2.67), and a trend towards higher maternal mortality in the cesarean group (1.6 per 100,000 for VBAC vs 5.6 per 100,000 for planned cesarean; OR=0.32; 95% CI, 0.07–1.47).9

The effect of VBAC on neonatal morbidity and mortality is unclear. In contrast to the negative larger trial,8 a smaller retrospective cohort of 24,529 births found a higher association of perinatal death for trial of labor (adjusted OR=11.7; 95% CI, 1.4–101.6).10 The perinatal death rate was similar to rates in nulliparous women. Regarding morbidity, one retrospective cohort trial showed VBAC was associated with an increase in neonatal sepsis (1% vs 0%; CI not given) compared with planned cesarean, but VBAC resulted in less transient tachypnea (5% vs 7%) and hyper-bilirubinemia (2% vs 6%).11

 

 

 

Recommendations from others

Both the American College of Obstetricians and Gynecologists and the Society of Obstetricians and Gynecologists of Canada state that women with 1 previous low-transverse cesarean section should be offered a trial of labor after appropriate counseling of the risks and benefits.12,13 Furthermore, induction with oxytocin is allowed, but the use of prostaglandins is not recommended. Based on expert opinion, both organizations encourage VBAC only in institutions staffed with surgeons and anesthesiologists immediately available to provide emergent cesarean.

Acknowledgments

The opinions and assertions contained herein are the private views of the author and are not to be construed as official, or as reflecting the views of the US Air Force medical department or the US Air Force at large.

EVIDENCE-BASED ANSWER

Compared with planned repeat low-transverse cesarean section, vaginal birth after cesarean section (VBAC) is not associated with increased risk of maternal or neonatal mortality (strength of recommendation [SOR]: B). Morbidity is slightly increased, as evidenced by higher uterine rupture rates and some neonatal outcome measures (SOR: B).

CLINICAL COMMENTARY

Risks of C-section and labor must be considered when counseling regarding route of delivery
Lynda DeArmond, MD
Waco Family Practice Residency Program, Waco, Texas

Another question to pose is: how safe is repeat cesarean section for the mother and fetus? How much do morbidity and mortality increase with each new intra-abdominal procedure? Each time the belly is opened there is new scar, with increased likelihood of adhesions and potential for future bowel obstruction. Consider these risks when counseling regarding route of delivery. Risk of uterine rupture appears to be higher in trials of labor (and confers a statistically significant but small increase in morbidity but not mortality). However, the uterine scar can silently fail without labor—as is sometimes discovered at a scheduled repeat section, usually without untoward effects on mother or fetus.

Remember that you are sending a young woman home with a new baby to care for (along with other children) and a major abdominal procedure (through an old scar) to recover from, which one could certainly define as morbidity. Cesarean section is an important tool, but we must be careful to practice best possible care and consider all patient factors and preferences. And data are still lacking to support the notion that VBAC is unsafe.

 

Evidence summary

Contrary to the goals of Healthy People 2010, the rate of cesarean sections is increasing.1 The repeat cesarean rate for low-risk women of all ages and racial groups is now 88.7%, the highest rate since the Centers for Disease Control and Prevention (CDC) began tracking the statistic in 1989. Is VBAC safe, or is a trial of labor no longer supported by the data?

The most recent Cochrane Review found that both VBAC and repeat lowtransverse cesarean section have benefits and risks associated with them; however, after reviewing the limited data, they concluded that no trial exists to adequately help women and their caregivers make an informed decision between the two.2 A strong theme in the Cochrane Review, echoed in most reviews, was the absence of high-quality prospective randomized data.

In an attempt to quantify the risks of VBAC, a systematic review determined that attempted VBAC, compared with repeat low-transverse cesarean section, increased the risk of uterine rupture by 2.7 per 1000 cases (95% confidence interval [CI], 0.73–4.73).3 This additional risk rate is often quoted in VBAC reviews and was cited in the Agency for Healthcare Research and Quality evidence report; it is based on 1 prospective, nonrandomized cohort trial and 1 retrospective cohort study.4,5

No randomized controlled trials exist for determining maternal safety of VBAC, although another recent systematic review found 2 nonrandomized prospective trials of sufficient quality to analyze. The authors concluded there were “no statistically significant differences between planned elective repeat cesarean section and planned VBAC.”6 Upon closer review in PubMed, one of the cited studies did not study 312 patients for VBAC outcomes as alleged; rather, it investigated patient attitudes towards VBAC.7

Since publication of that review, a large, multicenter, prospective, nonrandomized trial involving 33,699 patients found no significant difference between VBAC and planned cesarean for hysterectomy (0.2% vs 0.3%; odds ratio [OR]=0.77; 95% CI, 0.51–1.17), maternal death (0.02% vs 0.04%; OR=0.38; 95% CI, 0.1–1.46), and neonatal death (0.08% vs 0.05%; OR=1.82; 95% CI, 0.73–4.57).8 Significant associations were found for uterine rupture rates in spontaneous labor (24/6685 [0.4%] vs no cases; number needed to harm [NNH]=279) and neonatal hypoxic-ischemic encephalopathy (0.46 cases per 1000 vs no cases; NNH=2174).8

A retrospective Canadian cohort trial of 308,755 women also demonstrated an association of VBAC with uterine rupture(0.65% of trial-of-labor cases; OR=2.38; 95% CI, 2.12–2.67), and a trend towards higher maternal mortality in the cesarean group (1.6 per 100,000 for VBAC vs 5.6 per 100,000 for planned cesarean; OR=0.32; 95% CI, 0.07–1.47).9

The effect of VBAC on neonatal morbidity and mortality is unclear. In contrast to the negative larger trial,8 a smaller retrospective cohort of 24,529 births found a higher association of perinatal death for trial of labor (adjusted OR=11.7; 95% CI, 1.4–101.6).10 The perinatal death rate was similar to rates in nulliparous women. Regarding morbidity, one retrospective cohort trial showed VBAC was associated with an increase in neonatal sepsis (1% vs 0%; CI not given) compared with planned cesarean, but VBAC resulted in less transient tachypnea (5% vs 7%) and hyper-bilirubinemia (2% vs 6%).11

 

 

 

Recommendations from others

Both the American College of Obstetricians and Gynecologists and the Society of Obstetricians and Gynecologists of Canada state that women with 1 previous low-transverse cesarean section should be offered a trial of labor after appropriate counseling of the risks and benefits.12,13 Furthermore, induction with oxytocin is allowed, but the use of prostaglandins is not recommended. Based on expert opinion, both organizations encourage VBAC only in institutions staffed with surgeons and anesthesiologists immediately available to provide emergent cesarean.

Acknowledgments

The opinions and assertions contained herein are the private views of the author and are not to be construed as official, or as reflecting the views of the US Air Force medical department or the US Air Force at large.

References

1. Menacker F. Trends in cesarean rates for first births and repeat cesarean rates for low-risk women: United States, 1990-2003. Natl Vital Stat Rep 2005;54:1-8.

2. Dodd J, Crowther C, Huertas E, Guise J, Horey D. Planned elective repeat caesarean section versus planned vaginal birth for women with a previous caesarean birth. Cochrane Database Syst Rev 2005;(4):CD004224.

3. Guise J, McDonagh M, Osterweil P, Nygren P, Chan B, Helfand M. Systematic review of the incidence and consequences of uterine rupture in women with previous caesarean section. BMJ 2004;329:19-25.

4. Vaginal Birth After Cesarean (VBAC). Summary. Evidence Report/Technology Assessment, No. 71. AHRQ publication 03-E017. Rockville, Md: Agency for Healthcare Research and Quality; 2003. Available at: www.ahrq.gov/clinic/epcsums/vbacsum.htm. Accessed on January 9, 2006.

5. McMahon MJ, Luther ER, Bowes WA, Olshan AF. Comparison of a trial of labor with an elective second cesarean section. N Engl J Med 1996;335:689-695.

6. Dodd J, Crowther C. Vaginal birth after Caesarean versus elective repeat Caesarean for women with a single prior Caesarean birth: a systematic review of the literature. Aust N Z J Obstet Gynaecol 2004;44:387-391.

7. Abitbol MM, Castillo I, Taylor UB, Rochelson BL, Shmoys S, Monheit AG. Vaginal birth after cesarean section: the patient’s point of view. Am Fam Physician 1993;47:129-134.

8. Landon M, Hauth J, Leveno K, et al. for the National Institute of Child Health and Human Development Maternal-Fetal Medicine Units Network. Maternal and perinatal outcomes associated with a trial of labor after prior cesarean delivery. N Engl J Med 2004;351:2581-2589.

9. Wen S, Rusen I, Walker M, et al. Comparison of maternal mortality and morbidity between trial of labor and elective cesarean section among women with previous cesarean delivery. Am J Obstet Gynecol 2004;191:1263-1269.

10. Smith G, Pell J, Cameron A, Dobbie R. Risk of perinatal death associated with labor after previous cesarean delivery in uncomplicated term pregnancies. JAMA 2002;287:2684-2690.

11. Hook B, Kiwi R, Amini S, Fanaroff A, Hack M. Neonatal morbidity after elective repeat cesarean section and trial of labor. Pediatrics 1997;100:348-353.

12. American College of Obstetricians and Gynecologists Committee on Practice Bulletins—Obstetrics. ACOG Practice Bulletin. Vaginal Birth After Cesarean Delivery. Obstet Gynecol 2004;104:203-212.

13. Martel M, MacKinnon C, and the Clinical Practice Obstetrics Committee, Society of Obstetricians and Gynaecologists of Canada. Guidelines for vaginal birth after previous Caesarean birth. J Obstet Gynaecol Can 2005;27:164-188.

References

1. Menacker F. Trends in cesarean rates for first births and repeat cesarean rates for low-risk women: United States, 1990-2003. Natl Vital Stat Rep 2005;54:1-8.

2. Dodd J, Crowther C, Huertas E, Guise J, Horey D. Planned elective repeat caesarean section versus planned vaginal birth for women with a previous caesarean birth. Cochrane Database Syst Rev 2005;(4):CD004224.

3. Guise J, McDonagh M, Osterweil P, Nygren P, Chan B, Helfand M. Systematic review of the incidence and consequences of uterine rupture in women with previous caesarean section. BMJ 2004;329:19-25.

4. Vaginal Birth After Cesarean (VBAC). Summary. Evidence Report/Technology Assessment, No. 71. AHRQ publication 03-E017. Rockville, Md: Agency for Healthcare Research and Quality; 2003. Available at: www.ahrq.gov/clinic/epcsums/vbacsum.htm. Accessed on January 9, 2006.

5. McMahon MJ, Luther ER, Bowes WA, Olshan AF. Comparison of a trial of labor with an elective second cesarean section. N Engl J Med 1996;335:689-695.

6. Dodd J, Crowther C. Vaginal birth after Caesarean versus elective repeat Caesarean for women with a single prior Caesarean birth: a systematic review of the literature. Aust N Z J Obstet Gynaecol 2004;44:387-391.

7. Abitbol MM, Castillo I, Taylor UB, Rochelson BL, Shmoys S, Monheit AG. Vaginal birth after cesarean section: the patient’s point of view. Am Fam Physician 1993;47:129-134.

8. Landon M, Hauth J, Leveno K, et al. for the National Institute of Child Health and Human Development Maternal-Fetal Medicine Units Network. Maternal and perinatal outcomes associated with a trial of labor after prior cesarean delivery. N Engl J Med 2004;351:2581-2589.

9. Wen S, Rusen I, Walker M, et al. Comparison of maternal mortality and morbidity between trial of labor and elective cesarean section among women with previous cesarean delivery. Am J Obstet Gynecol 2004;191:1263-1269.

10. Smith G, Pell J, Cameron A, Dobbie R. Risk of perinatal death associated with labor after previous cesarean delivery in uncomplicated term pregnancies. JAMA 2002;287:2684-2690.

11. Hook B, Kiwi R, Amini S, Fanaroff A, Hack M. Neonatal morbidity after elective repeat cesarean section and trial of labor. Pediatrics 1997;100:348-353.

12. American College of Obstetricians and Gynecologists Committee on Practice Bulletins—Obstetrics. ACOG Practice Bulletin. Vaginal Birth After Cesarean Delivery. Obstet Gynecol 2004;104:203-212.

13. Martel M, MacKinnon C, and the Clinical Practice Obstetrics Committee, Society of Obstetricians and Gynaecologists of Canada. Guidelines for vaginal birth after previous Caesarean birth. J Obstet Gynaecol Can 2005;27:164-188.

Issue
The Journal of Family Practice - 55(2)
Issue
The Journal of Family Practice - 55(2)
Page Number
149-151
Page Number
149-151
Publications
Publications
Topics
Article Type
Display Headline
How safe is vaginal birth after cesarean section for the mother and fetus?
Display Headline
How safe is vaginal birth after cesarean section for the mother and fetus?
Sections
PURLs Copyright

Evidence-based answers from the Family Physicians Inquiries Network

Disallow All Ads
Alternative CME
Article PDF Media

What dietary modifications are indicated for migraines?

Article Type
Changed
Mon, 01/14/2019 - 11:15
Display Headline
What dietary modifications are indicated for migraines?
EVIDENCE-BASED ANSWER

Migraine frequency, duration, and severity are not increased by dietary choices (strength of recommendation [SOR]: A, individual randomized trial [RCT]); they can be decreased by a low-fat diet (SOR: B). Regular supplementation with high-dose riboflavin or magnesium reduces frequency and intensity of migraines (SOR: B, single RCT).

CLINICAL COMMENTARY

Have patients keep a migraine diary; experiment with dietary/activity modifications
Jennifer Hoock, MD
University of Washington, Seattle

Interestingly, this review of the literature seems to both disavow commonly accepted beliefs about migraine triggers and suggest new dietary interventions. In my experience, foods like chocolate, cheese, and citrus are rarely reported by patients as migraine triggers. Alcohol is reported as a trigger, though the possibility that it is a cofactor with stress and fatigue seems plausible. Certainly patients perceive that various foods and activities trigger their migraines. It is possible that no universal food triggers exist, but that persons have individual triggers. In the end, I think the practical approach remains to have patients keep a diary of the events surrounding their migraines, identify patterns and experiment with dietary and activity modifications. An empiric trial of magnesium or riboflavin certainly seems worth consideration, as does a recommendation for a healthy low-fat diet that incorporates omega-3 fatty acids/olive oil. Migraine treatment remains a process of educated trial and error to find the optimal combination of lifestyle modifications and medications.

 

Evidence summary

Contrary to what many physicians learned from their mentors—and to what many patients believe—no food or food additive has been proven to cause migraine headaches; and in fact, good evidence disproves this notion. The primary foods once thought to trigger migraines were cheese, alcohol, chocolate and citrus fruit.1 Conversely, it appears that regular supplementation with some nutrients reduces the frequency and intensity of migraines (TABLE 1).

Vasoactive amines. Vasoactive amines (ie, tyramine and phenylethylamine) are present in aged cheese and red wine. One randomized trial of 80 patients with frequent migraines showed that tyramine and placebo induced migraine at the same rate.2 A systematic review on the relation of vasoactive amines to migraine found no evidence that any biogenic amines in red wine, cheese, or chocolate cause migraine.3 Furthermore, an uncontrolled prospective trial failed to show that amount or type of alcohol correlates with migraines, but it did find a correlation between stressful events and migraines. These stressful events also correlated with a higher alcohol intake.4 One final small randomized controlled trial enrolling children found no difference in migraine frequency between high fiber/high vasoactive amine and a high fiber/low vasoactive amine diet.5 In contrast to these RCTs, one series of lower-evidence-level patient surveys from a tertiary clinic reported that approximately 12% to 28% of patients perceived migraines were triggered by various foods (ie, cheese, wine, beer, chocolate).6

Chocolate. The role of chocolate in instigating headache was investigated in a 63-subject double-blind RCT comparing chocolate with carob. Chocolate was not more likely to provoke headache than carob in any of the headache diagnostic groups (P=.83). These results were independent of subjects’ beliefs regarding the role of chocolate in the instigation of headache (P=.39). Unfortunately, this trial included multiple headache types, with only 50% being migraine.7

Omega-3 fatty acids. A small double-blind crossover study of 27 adolescents over 5 months showed no difference between fish oil supplementation and “placebo supplementation” with olive oil. The dose of fish oil used is approximately equivalent to 1.5 g of the recently approved Omacor fish oil capsules. Interestingly, the subjects reported dramatic decreases in headache frequency (15 per month down to 2 episodes per month) and decreases in headache severity (reduction from 5 to 3 on a 7-point Likert scale) with both compounds.8 The possibility of olive oil being an active comparator muddles interpretation of the results.

Riboflavin. A good-quality RCT compared riboflavin 400 mg/d with placebo for prophylaxis of migraines.9 Using intention-to-treat analysis, riboflavin was superior to placebo in reducing attack frequency (P=.005) and headache days (P=.012). The proportion of patients who improved by at least 50% was 15% for placebo and 59% for riboflavin (P=.002). The number needed to treat (NNT) was 2.3. Adverse events were very rare—1 case of diarrhea was reported causing withdrawal (number needed to harm [NNH]=33.3). The effect of riboflavin on migraine began at 1 month but was maximal at 3 months, when this study ended. The most pronounced effect was shorter migraine attacks followed by fewer migraine attacks. An additional large case series found that high-dose riboflavin reduced headache days by 50% (P<.05) and use of abortive medicines by 36% (P<.05).10

 

 

 

Magnesium. A good-quality RCT of 81 adults given 600 mg of magnesium or placebo showed that by weeks 9 to 12 frequency of attacks was reduced by 41.6% in the magnesium group and by 15.8% in the placebo group compared with the baseline (P<.05; NNT=3.9). However, diarrhea was reported among 18.6% of magnesium recipients (NNH=5.2).11 Additionally, a very small randomized trial of 20 women found that magnesium 360 mg/d in the luteal phase reduced the number of days with menstrual-related migraines (P<.03) when compared with placebo. However, the absolute magnitude of the difference was not reported, so it is unclear if this study is clinically useful or can be extended to all patients with migraines.12

Low-fat diet. A prospective cohort trial of 54 patients evaluated the effect of lowering total fat intake per day. The dietary intervention successfully lowered fat intake from 65.9 to 27.8 g/d and was associated with statistically significant decreases in headache frequency (median decrease from 6 to 1 per week), intensity (median decrease 2.9 to 0.5 on a scale from 0 to 5), duration, and medication intake (P<.0001 for all measures, confidence interval not available).13

Caffeine. One case series found that adolescents and children ingesting over 1400 mg/wk of caffeine from cola drinks experienced resolution of headaches with gradual reduction in cola intake,14 but no prospective trials to confirm this observation have been completed. It is important to note that reduction in migraines may have been due to reduction in other ingredients, not just caffeine.

Riboflavin/magnesium/feverfew. One double-blind RCT compared a compound with daily doses of riboflavin 400 mg, magnesium 300 mg, and feverfew 100 mg with the proposed placebo of riboflavin 25 mg. There was no difference in response between the compound and the riboflavin placebo; however, response to riboflavin 25 mg was higher than expected for a placebo (44%). Further study with a placebo free of active ingredients is required to determine the ultimate effectiveness of this compound.15

TABLE
Effects of dietary compounds on migraine headaches

DIETARY COMPOUND OR DIET TYPEEFFECT ON MIGRAINESADVERSE EFFECTS
Vasoactive amines00
Chocolate00
Omega-3 fatty acids00
Low-fat diet (<20 g/day)+0
Riboflavin 400 mg/day++++
Magnesium++++
Caffeine??
Riboflavin/magnesium/feverfew??
Key: 0=no effect, +=slight effect, ++=moderate effect, +++=large effect, ++++=very large effect.

Recommendations from others

The American Academy of Neurology makes no mention of dietary therapy in its most recent guideline on migraine, but it does identify both riboflavin (fair evidence) and magnesium (weak evidence) as safe options for preventing migraine.16 The National Headache Foundation makes no statement regarding dietary therapy for migraines, but it does identify riboflavin and magnesium as possible preventive therapies.17

Acknowledgments

The opinions and assertions contained herein are the private views of the author and not to be construed as official, or as reflecting the views of the US Air Force Medical Service or the US Air Force at large.

References

1. Peatfield RC, Glover V, Littlewood JT, Sandler M, Clifford Rose F. The prevalence of diet-induced migraine. Cephalalgia 1984;4:179-183.

2. Ziegler DK, Stewart R. Failure of tyramine to induce migraine. Neurology 1977;27:725-726.

3. Jansen SC, van Dusseldorp M, Bottema KC, Dubois AE. Intolerance to dietary biogenic amines: a review. Ann Allergy Asthma Immunol 2003;91:233-240.

4. Nicolodi M, Sicuteri F. Wine and migraine: compatibility or incompatibility? Drugs Exp Clin Res 1999;25:147-153.

5. Salfield SA, Wardley BL, Houlsby WT, et al. Controlled study of exclusion of dietary vasoactive amines in migraine. Arch Dis Child 1987;62:458-460.

6. Peatfield R. Relationships between food, wine, and beer-precipitated migrainous headaches. Headache 1995;35:355-357.

7. Marcus DA, Scharff L, Turk D, Gourley LM. A double-blind provocative study of chocolate as a trigger of headache. Cephalalgia 1997;17:855-862.

8. Harel Z, Gascon G, Riggs S, Vaz R, Brown W, Exil G. Supplementation with omega-3 polyunsaturated fatty acids in the management of recurrent migraines in adolescents. J Adoles Health 2002;31:154-161.

9. Schoenen J, Jacquoy J, Lenaerts M. Effectiveness of high-dose riboflavin in migraine prophylaxis. A randomized controlled trial. Neurology 1998;50:466-470.

10. Boehnke C, Reuter U, Flach U, Schuh-Hofer S, Einhaupl KM, Arnold G. High-dose riboflavin treatment is efficacious in migraine prophylaxis: an open study in a tertiary care centre. Eur J Neurol 2004;11:475-477.

11. Peikert A, Wilimzig C, Köhne-Volland R. Prophylaxis of migraine with oral magnesium: results from a prospective, multi-center, placebo-controlled and double-blind randomized study. Cephalalgia 1996;16:257-263.

12. Facchinetti F, Sances G, Borella P, Genazzani AR, Nappi G. Magnesium prophylaxis of menstrual migraine: effects on intracellular magnesium. Headache 1991;31:298-301.

13. Bic Z, Blix GG, Hopp HP, Leslie FM, Schell MJ. The influence of a low-fat diet on incidence and severity of migraine headaches. J Womens Health Gend Based Med 1999;8:623-630.

14. Hering-Hanit R, Gadoth N. Caffeine-induced headache in children and adolescents. Cephalalgia 2003;23:332-335.

15. Maizels M, Blumenfeld A, Burchette R. A combination of riboflavin, magnesium, and feverfew for migraine prophylaxis: a randomized trial. Headache 2004;44:885-890.

16. Silberstein SD. Practice parameter: evidence-based guidelines for migraine headache (an evidence-based review): report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology 2000;55:754.Available at: www.guideline.gov/summary/summary.aspx?doc_id=2820. Accessed on December 7, 2005.

17. Mauskop A, Graff Radford S. Special treatment situations: alternative headache treatments. In: Standards of Care for Headache Diagnosis and Treatment. Chicago, Ill: National Headache Foundation; 2004:115. Available at: www.guideline.gov/summary/summary.aspx?doc_id=6588. Accessed on December 8, 2005.

Article PDF
Author and Disclosure Information

Paul Crawford, MD
Eglin Air Force Base Family Medicine Residency, Eglin Air Force Base, Fla

Michael Simmons, MLIS
Sparrow Health System, Lansing, Mich

Issue
The Journal of Family Practice - 55(1)
Publications
Topics
Page Number
62-66
Sections
Author and Disclosure Information

Paul Crawford, MD
Eglin Air Force Base Family Medicine Residency, Eglin Air Force Base, Fla

Michael Simmons, MLIS
Sparrow Health System, Lansing, Mich

Author and Disclosure Information

Paul Crawford, MD
Eglin Air Force Base Family Medicine Residency, Eglin Air Force Base, Fla

Michael Simmons, MLIS
Sparrow Health System, Lansing, Mich

Article PDF
Article PDF
EVIDENCE-BASED ANSWER

Migraine frequency, duration, and severity are not increased by dietary choices (strength of recommendation [SOR]: A, individual randomized trial [RCT]); they can be decreased by a low-fat diet (SOR: B). Regular supplementation with high-dose riboflavin or magnesium reduces frequency and intensity of migraines (SOR: B, single RCT).

CLINICAL COMMENTARY

Have patients keep a migraine diary; experiment with dietary/activity modifications
Jennifer Hoock, MD
University of Washington, Seattle

Interestingly, this review of the literature seems to both disavow commonly accepted beliefs about migraine triggers and suggest new dietary interventions. In my experience, foods like chocolate, cheese, and citrus are rarely reported by patients as migraine triggers. Alcohol is reported as a trigger, though the possibility that it is a cofactor with stress and fatigue seems plausible. Certainly patients perceive that various foods and activities trigger their migraines. It is possible that no universal food triggers exist, but that persons have individual triggers. In the end, I think the practical approach remains to have patients keep a diary of the events surrounding their migraines, identify patterns and experiment with dietary and activity modifications. An empiric trial of magnesium or riboflavin certainly seems worth consideration, as does a recommendation for a healthy low-fat diet that incorporates omega-3 fatty acids/olive oil. Migraine treatment remains a process of educated trial and error to find the optimal combination of lifestyle modifications and medications.

 

Evidence summary

Contrary to what many physicians learned from their mentors—and to what many patients believe—no food or food additive has been proven to cause migraine headaches; and in fact, good evidence disproves this notion. The primary foods once thought to trigger migraines were cheese, alcohol, chocolate and citrus fruit.1 Conversely, it appears that regular supplementation with some nutrients reduces the frequency and intensity of migraines (TABLE 1).

Vasoactive amines. Vasoactive amines (ie, tyramine and phenylethylamine) are present in aged cheese and red wine. One randomized trial of 80 patients with frequent migraines showed that tyramine and placebo induced migraine at the same rate.2 A systematic review on the relation of vasoactive amines to migraine found no evidence that any biogenic amines in red wine, cheese, or chocolate cause migraine.3 Furthermore, an uncontrolled prospective trial failed to show that amount or type of alcohol correlates with migraines, but it did find a correlation between stressful events and migraines. These stressful events also correlated with a higher alcohol intake.4 One final small randomized controlled trial enrolling children found no difference in migraine frequency between high fiber/high vasoactive amine and a high fiber/low vasoactive amine diet.5 In contrast to these RCTs, one series of lower-evidence-level patient surveys from a tertiary clinic reported that approximately 12% to 28% of patients perceived migraines were triggered by various foods (ie, cheese, wine, beer, chocolate).6

Chocolate. The role of chocolate in instigating headache was investigated in a 63-subject double-blind RCT comparing chocolate with carob. Chocolate was not more likely to provoke headache than carob in any of the headache diagnostic groups (P=.83). These results were independent of subjects’ beliefs regarding the role of chocolate in the instigation of headache (P=.39). Unfortunately, this trial included multiple headache types, with only 50% being migraine.7

Omega-3 fatty acids. A small double-blind crossover study of 27 adolescents over 5 months showed no difference between fish oil supplementation and “placebo supplementation” with olive oil. The dose of fish oil used is approximately equivalent to 1.5 g of the recently approved Omacor fish oil capsules. Interestingly, the subjects reported dramatic decreases in headache frequency (15 per month down to 2 episodes per month) and decreases in headache severity (reduction from 5 to 3 on a 7-point Likert scale) with both compounds.8 The possibility of olive oil being an active comparator muddles interpretation of the results.

Riboflavin. A good-quality RCT compared riboflavin 400 mg/d with placebo for prophylaxis of migraines.9 Using intention-to-treat analysis, riboflavin was superior to placebo in reducing attack frequency (P=.005) and headache days (P=.012). The proportion of patients who improved by at least 50% was 15% for placebo and 59% for riboflavin (P=.002). The number needed to treat (NNT) was 2.3. Adverse events were very rare—1 case of diarrhea was reported causing withdrawal (number needed to harm [NNH]=33.3). The effect of riboflavin on migraine began at 1 month but was maximal at 3 months, when this study ended. The most pronounced effect was shorter migraine attacks followed by fewer migraine attacks. An additional large case series found that high-dose riboflavin reduced headache days by 50% (P<.05) and use of abortive medicines by 36% (P<.05).10

 

 

 

Magnesium. A good-quality RCT of 81 adults given 600 mg of magnesium or placebo showed that by weeks 9 to 12 frequency of attacks was reduced by 41.6% in the magnesium group and by 15.8% in the placebo group compared with the baseline (P<.05; NNT=3.9). However, diarrhea was reported among 18.6% of magnesium recipients (NNH=5.2).11 Additionally, a very small randomized trial of 20 women found that magnesium 360 mg/d in the luteal phase reduced the number of days with menstrual-related migraines (P<.03) when compared with placebo. However, the absolute magnitude of the difference was not reported, so it is unclear if this study is clinically useful or can be extended to all patients with migraines.12

Low-fat diet. A prospective cohort trial of 54 patients evaluated the effect of lowering total fat intake per day. The dietary intervention successfully lowered fat intake from 65.9 to 27.8 g/d and was associated with statistically significant decreases in headache frequency (median decrease from 6 to 1 per week), intensity (median decrease 2.9 to 0.5 on a scale from 0 to 5), duration, and medication intake (P<.0001 for all measures, confidence interval not available).13

Caffeine. One case series found that adolescents and children ingesting over 1400 mg/wk of caffeine from cola drinks experienced resolution of headaches with gradual reduction in cola intake,14 but no prospective trials to confirm this observation have been completed. It is important to note that reduction in migraines may have been due to reduction in other ingredients, not just caffeine.

Riboflavin/magnesium/feverfew. One double-blind RCT compared a compound with daily doses of riboflavin 400 mg, magnesium 300 mg, and feverfew 100 mg with the proposed placebo of riboflavin 25 mg. There was no difference in response between the compound and the riboflavin placebo; however, response to riboflavin 25 mg was higher than expected for a placebo (44%). Further study with a placebo free of active ingredients is required to determine the ultimate effectiveness of this compound.15

TABLE
Effects of dietary compounds on migraine headaches

DIETARY COMPOUND OR DIET TYPEEFFECT ON MIGRAINESADVERSE EFFECTS
Vasoactive amines00
Chocolate00
Omega-3 fatty acids00
Low-fat diet (<20 g/day)+0
Riboflavin 400 mg/day++++
Magnesium++++
Caffeine??
Riboflavin/magnesium/feverfew??
Key: 0=no effect, +=slight effect, ++=moderate effect, +++=large effect, ++++=very large effect.

Recommendations from others

The American Academy of Neurology makes no mention of dietary therapy in its most recent guideline on migraine, but it does identify both riboflavin (fair evidence) and magnesium (weak evidence) as safe options for preventing migraine.16 The National Headache Foundation makes no statement regarding dietary therapy for migraines, but it does identify riboflavin and magnesium as possible preventive therapies.17

Acknowledgments

The opinions and assertions contained herein are the private views of the author and not to be construed as official, or as reflecting the views of the US Air Force Medical Service or the US Air Force at large.

EVIDENCE-BASED ANSWER

Migraine frequency, duration, and severity are not increased by dietary choices (strength of recommendation [SOR]: A, individual randomized trial [RCT]); they can be decreased by a low-fat diet (SOR: B). Regular supplementation with high-dose riboflavin or magnesium reduces frequency and intensity of migraines (SOR: B, single RCT).

CLINICAL COMMENTARY

Have patients keep a migraine diary; experiment with dietary/activity modifications
Jennifer Hoock, MD
University of Washington, Seattle

Interestingly, this review of the literature seems to both disavow commonly accepted beliefs about migraine triggers and suggest new dietary interventions. In my experience, foods like chocolate, cheese, and citrus are rarely reported by patients as migraine triggers. Alcohol is reported as a trigger, though the possibility that it is a cofactor with stress and fatigue seems plausible. Certainly patients perceive that various foods and activities trigger their migraines. It is possible that no universal food triggers exist, but that persons have individual triggers. In the end, I think the practical approach remains to have patients keep a diary of the events surrounding their migraines, identify patterns and experiment with dietary and activity modifications. An empiric trial of magnesium or riboflavin certainly seems worth consideration, as does a recommendation for a healthy low-fat diet that incorporates omega-3 fatty acids/olive oil. Migraine treatment remains a process of educated trial and error to find the optimal combination of lifestyle modifications and medications.

 

Evidence summary

Contrary to what many physicians learned from their mentors—and to what many patients believe—no food or food additive has been proven to cause migraine headaches; and in fact, good evidence disproves this notion. The primary foods once thought to trigger migraines were cheese, alcohol, chocolate and citrus fruit.1 Conversely, it appears that regular supplementation with some nutrients reduces the frequency and intensity of migraines (TABLE 1).

Vasoactive amines. Vasoactive amines (ie, tyramine and phenylethylamine) are present in aged cheese and red wine. One randomized trial of 80 patients with frequent migraines showed that tyramine and placebo induced migraine at the same rate.2 A systematic review on the relation of vasoactive amines to migraine found no evidence that any biogenic amines in red wine, cheese, or chocolate cause migraine.3 Furthermore, an uncontrolled prospective trial failed to show that amount or type of alcohol correlates with migraines, but it did find a correlation between stressful events and migraines. These stressful events also correlated with a higher alcohol intake.4 One final small randomized controlled trial enrolling children found no difference in migraine frequency between high fiber/high vasoactive amine and a high fiber/low vasoactive amine diet.5 In contrast to these RCTs, one series of lower-evidence-level patient surveys from a tertiary clinic reported that approximately 12% to 28% of patients perceived migraines were triggered by various foods (ie, cheese, wine, beer, chocolate).6

Chocolate. The role of chocolate in instigating headache was investigated in a 63-subject double-blind RCT comparing chocolate with carob. Chocolate was not more likely to provoke headache than carob in any of the headache diagnostic groups (P=.83). These results were independent of subjects’ beliefs regarding the role of chocolate in the instigation of headache (P=.39). Unfortunately, this trial included multiple headache types, with only 50% being migraine.7

Omega-3 fatty acids. A small double-blind crossover study of 27 adolescents over 5 months showed no difference between fish oil supplementation and “placebo supplementation” with olive oil. The dose of fish oil used is approximately equivalent to 1.5 g of the recently approved Omacor fish oil capsules. Interestingly, the subjects reported dramatic decreases in headache frequency (15 per month down to 2 episodes per month) and decreases in headache severity (reduction from 5 to 3 on a 7-point Likert scale) with both compounds.8 The possibility of olive oil being an active comparator muddles interpretation of the results.

Riboflavin. A good-quality RCT compared riboflavin 400 mg/d with placebo for prophylaxis of migraines.9 Using intention-to-treat analysis, riboflavin was superior to placebo in reducing attack frequency (P=.005) and headache days (P=.012). The proportion of patients who improved by at least 50% was 15% for placebo and 59% for riboflavin (P=.002). The number needed to treat (NNT) was 2.3. Adverse events were very rare—1 case of diarrhea was reported causing withdrawal (number needed to harm [NNH]=33.3). The effect of riboflavin on migraine began at 1 month but was maximal at 3 months, when this study ended. The most pronounced effect was shorter migraine attacks followed by fewer migraine attacks. An additional large case series found that high-dose riboflavin reduced headache days by 50% (P<.05) and use of abortive medicines by 36% (P<.05).10

 

 

 

Magnesium. A good-quality RCT of 81 adults given 600 mg of magnesium or placebo showed that by weeks 9 to 12 frequency of attacks was reduced by 41.6% in the magnesium group and by 15.8% in the placebo group compared with the baseline (P<.05; NNT=3.9). However, diarrhea was reported among 18.6% of magnesium recipients (NNH=5.2).11 Additionally, a very small randomized trial of 20 women found that magnesium 360 mg/d in the luteal phase reduced the number of days with menstrual-related migraines (P<.03) when compared with placebo. However, the absolute magnitude of the difference was not reported, so it is unclear if this study is clinically useful or can be extended to all patients with migraines.12

Low-fat diet. A prospective cohort trial of 54 patients evaluated the effect of lowering total fat intake per day. The dietary intervention successfully lowered fat intake from 65.9 to 27.8 g/d and was associated with statistically significant decreases in headache frequency (median decrease from 6 to 1 per week), intensity (median decrease 2.9 to 0.5 on a scale from 0 to 5), duration, and medication intake (P<.0001 for all measures, confidence interval not available).13

Caffeine. One case series found that adolescents and children ingesting over 1400 mg/wk of caffeine from cola drinks experienced resolution of headaches with gradual reduction in cola intake,14 but no prospective trials to confirm this observation have been completed. It is important to note that reduction in migraines may have been due to reduction in other ingredients, not just caffeine.

Riboflavin/magnesium/feverfew. One double-blind RCT compared a compound with daily doses of riboflavin 400 mg, magnesium 300 mg, and feverfew 100 mg with the proposed placebo of riboflavin 25 mg. There was no difference in response between the compound and the riboflavin placebo; however, response to riboflavin 25 mg was higher than expected for a placebo (44%). Further study with a placebo free of active ingredients is required to determine the ultimate effectiveness of this compound.15

TABLE
Effects of dietary compounds on migraine headaches

DIETARY COMPOUND OR DIET TYPEEFFECT ON MIGRAINESADVERSE EFFECTS
Vasoactive amines00
Chocolate00
Omega-3 fatty acids00
Low-fat diet (<20 g/day)+0
Riboflavin 400 mg/day++++
Magnesium++++
Caffeine??
Riboflavin/magnesium/feverfew??
Key: 0=no effect, +=slight effect, ++=moderate effect, +++=large effect, ++++=very large effect.

Recommendations from others

The American Academy of Neurology makes no mention of dietary therapy in its most recent guideline on migraine, but it does identify both riboflavin (fair evidence) and magnesium (weak evidence) as safe options for preventing migraine.16 The National Headache Foundation makes no statement regarding dietary therapy for migraines, but it does identify riboflavin and magnesium as possible preventive therapies.17

Acknowledgments

The opinions and assertions contained herein are the private views of the author and not to be construed as official, or as reflecting the views of the US Air Force Medical Service or the US Air Force at large.

References

1. Peatfield RC, Glover V, Littlewood JT, Sandler M, Clifford Rose F. The prevalence of diet-induced migraine. Cephalalgia 1984;4:179-183.

2. Ziegler DK, Stewart R. Failure of tyramine to induce migraine. Neurology 1977;27:725-726.

3. Jansen SC, van Dusseldorp M, Bottema KC, Dubois AE. Intolerance to dietary biogenic amines: a review. Ann Allergy Asthma Immunol 2003;91:233-240.

4. Nicolodi M, Sicuteri F. Wine and migraine: compatibility or incompatibility? Drugs Exp Clin Res 1999;25:147-153.

5. Salfield SA, Wardley BL, Houlsby WT, et al. Controlled study of exclusion of dietary vasoactive amines in migraine. Arch Dis Child 1987;62:458-460.

6. Peatfield R. Relationships between food, wine, and beer-precipitated migrainous headaches. Headache 1995;35:355-357.

7. Marcus DA, Scharff L, Turk D, Gourley LM. A double-blind provocative study of chocolate as a trigger of headache. Cephalalgia 1997;17:855-862.

8. Harel Z, Gascon G, Riggs S, Vaz R, Brown W, Exil G. Supplementation with omega-3 polyunsaturated fatty acids in the management of recurrent migraines in adolescents. J Adoles Health 2002;31:154-161.

9. Schoenen J, Jacquoy J, Lenaerts M. Effectiveness of high-dose riboflavin in migraine prophylaxis. A randomized controlled trial. Neurology 1998;50:466-470.

10. Boehnke C, Reuter U, Flach U, Schuh-Hofer S, Einhaupl KM, Arnold G. High-dose riboflavin treatment is efficacious in migraine prophylaxis: an open study in a tertiary care centre. Eur J Neurol 2004;11:475-477.

11. Peikert A, Wilimzig C, Köhne-Volland R. Prophylaxis of migraine with oral magnesium: results from a prospective, multi-center, placebo-controlled and double-blind randomized study. Cephalalgia 1996;16:257-263.

12. Facchinetti F, Sances G, Borella P, Genazzani AR, Nappi G. Magnesium prophylaxis of menstrual migraine: effects on intracellular magnesium. Headache 1991;31:298-301.

13. Bic Z, Blix GG, Hopp HP, Leslie FM, Schell MJ. The influence of a low-fat diet on incidence and severity of migraine headaches. J Womens Health Gend Based Med 1999;8:623-630.

14. Hering-Hanit R, Gadoth N. Caffeine-induced headache in children and adolescents. Cephalalgia 2003;23:332-335.

15. Maizels M, Blumenfeld A, Burchette R. A combination of riboflavin, magnesium, and feverfew for migraine prophylaxis: a randomized trial. Headache 2004;44:885-890.

16. Silberstein SD. Practice parameter: evidence-based guidelines for migraine headache (an evidence-based review): report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology 2000;55:754.Available at: www.guideline.gov/summary/summary.aspx?doc_id=2820. Accessed on December 7, 2005.

17. Mauskop A, Graff Radford S. Special treatment situations: alternative headache treatments. In: Standards of Care for Headache Diagnosis and Treatment. Chicago, Ill: National Headache Foundation; 2004:115. Available at: www.guideline.gov/summary/summary.aspx?doc_id=6588. Accessed on December 8, 2005.

References

1. Peatfield RC, Glover V, Littlewood JT, Sandler M, Clifford Rose F. The prevalence of diet-induced migraine. Cephalalgia 1984;4:179-183.

2. Ziegler DK, Stewart R. Failure of tyramine to induce migraine. Neurology 1977;27:725-726.

3. Jansen SC, van Dusseldorp M, Bottema KC, Dubois AE. Intolerance to dietary biogenic amines: a review. Ann Allergy Asthma Immunol 2003;91:233-240.

4. Nicolodi M, Sicuteri F. Wine and migraine: compatibility or incompatibility? Drugs Exp Clin Res 1999;25:147-153.

5. Salfield SA, Wardley BL, Houlsby WT, et al. Controlled study of exclusion of dietary vasoactive amines in migraine. Arch Dis Child 1987;62:458-460.

6. Peatfield R. Relationships between food, wine, and beer-precipitated migrainous headaches. Headache 1995;35:355-357.

7. Marcus DA, Scharff L, Turk D, Gourley LM. A double-blind provocative study of chocolate as a trigger of headache. Cephalalgia 1997;17:855-862.

8. Harel Z, Gascon G, Riggs S, Vaz R, Brown W, Exil G. Supplementation with omega-3 polyunsaturated fatty acids in the management of recurrent migraines in adolescents. J Adoles Health 2002;31:154-161.

9. Schoenen J, Jacquoy J, Lenaerts M. Effectiveness of high-dose riboflavin in migraine prophylaxis. A randomized controlled trial. Neurology 1998;50:466-470.

10. Boehnke C, Reuter U, Flach U, Schuh-Hofer S, Einhaupl KM, Arnold G. High-dose riboflavin treatment is efficacious in migraine prophylaxis: an open study in a tertiary care centre. Eur J Neurol 2004;11:475-477.

11. Peikert A, Wilimzig C, Köhne-Volland R. Prophylaxis of migraine with oral magnesium: results from a prospective, multi-center, placebo-controlled and double-blind randomized study. Cephalalgia 1996;16:257-263.

12. Facchinetti F, Sances G, Borella P, Genazzani AR, Nappi G. Magnesium prophylaxis of menstrual migraine: effects on intracellular magnesium. Headache 1991;31:298-301.

13. Bic Z, Blix GG, Hopp HP, Leslie FM, Schell MJ. The influence of a low-fat diet on incidence and severity of migraine headaches. J Womens Health Gend Based Med 1999;8:623-630.

14. Hering-Hanit R, Gadoth N. Caffeine-induced headache in children and adolescents. Cephalalgia 2003;23:332-335.

15. Maizels M, Blumenfeld A, Burchette R. A combination of riboflavin, magnesium, and feverfew for migraine prophylaxis: a randomized trial. Headache 2004;44:885-890.

16. Silberstein SD. Practice parameter: evidence-based guidelines for migraine headache (an evidence-based review): report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology 2000;55:754.Available at: www.guideline.gov/summary/summary.aspx?doc_id=2820. Accessed on December 7, 2005.

17. Mauskop A, Graff Radford S. Special treatment situations: alternative headache treatments. In: Standards of Care for Headache Diagnosis and Treatment. Chicago, Ill: National Headache Foundation; 2004:115. Available at: www.guideline.gov/summary/summary.aspx?doc_id=6588. Accessed on December 8, 2005.

Issue
The Journal of Family Practice - 55(1)
Issue
The Journal of Family Practice - 55(1)
Page Number
62-66
Page Number
62-66
Publications
Publications
Topics
Article Type
Display Headline
What dietary modifications are indicated for migraines?
Display Headline
What dietary modifications are indicated for migraines?
Sections
PURLs Copyright

Evidence-based answers from the Family Physicians Inquiries Network

Disallow All Ads
Alternative CME
Article PDF Media