Appendices for U.S. Medical Eligibility Criteria for Contraceptive Use, 2024

View associated article

 

Appendix A: Summary of Changes from U.S. Medical Eligibility Criteria for Contraceptive Use, 2016

The classification additions, deletions, and modifications from the 2016 U.S. Medical Eligibility Criteria for Contraceptive Use (U.S. MEC) are summarized in this appendix (Box A1) (Tables A1, A2, and A3). For conditions for which classifications changed for one or more contraceptive methods or for which the condition description underwent a substantive modification, the changes or modifications are noted (Tables A1, A2, and A3). Conditions that do not appear in this table remain unchanged from the 2016 U.S. MEC.

Return to your place in the textBOX A1. Categories for classifying intrauterine devices and hormonal contraceptives

U.S. MEC 1 = A condition for which there is no restriction for the use of the contraceptive method

U.S. MEC 2 = A condition for which the advantages of using the method generally outweigh the theoretical or proven risks

U.S. MEC 3 = A condition for which the theoretical or proven risks usually outweigh the advantages of using the method

U.S. MEC 4 = A condition that represents an unacceptable health risk if the contraceptive method is used

Abbreviation: U.S. MEC = U.S. Medical Eligibility Criteria for Contraceptive Use.



TABLE A1. Summary of changes in classifications for hormonal contraceptive methods and intrauterine devices from U.S. Medical Eligibility Criteria for Contraceptive Use, 2016
Condition Cu-IUD LNG-IUD Implant DMPA POP CHC Clarification
Breastfeeding
a. <21 days postpartum 2 2 2 4 Breastfeeding provides important health benefits for breastfeeding parent and infant. The U.S. Dietary Guidelines for Americans and American Academy of Pediatrics recommend that infants be exclusively breastfed for about the first 6 months with continued breastfeeding while introducing appropriate complementary foods for 1 year or longer (1) or up to age 2 years or longer (2).
b. 21 to <30 days postpartum
  i. With other risk factors for VTE (e.g., age ≥35 years, previous VTE, thrombophilia, immobility, transfusion at delivery, peripartum cardiomyopathy, BMI ≥30 kg/m2, postpartum hemorrhage, postcesarean delivery, preeclampsia, or smoking) 2 2 2 3 CHC: For persons with other risk factors for VTE, these risk factors might increase the classification to a category 4.
Breastfeeding: Breastfeeding provides important health benefits for breastfeeding parent and infant. The U.S. Dietary Guidelines for Americans and American Academy of Pediatrics recommend that infants be exclusively breastfed for about the first 6 months with continued breastfeeding while introducing appropriate complementary foods for 1 year or longer (1) or up to age 2 years or longer (2).
  ii. Without other risk factors for VTE 2 2 2 3 Breastfeeding provides important health benefits for breastfeeding parent and infant. The U.S. Dietary Guidelines for Americans and American Academy of Pediatrics recommend that infants be exclusively breastfed for about the first 6 months with continued breastfeeding while introducing appropriate complementary foods for 1 year or longer (1) or up to age 2 years or longer (2).
c. 30–42 days postpartum
  i. With other risk factors for VTE (e.g., age ≥35 years, previous VTE, thrombophilia, immobility, transfusion at delivery, peripartum cardiomyopathy, BMI ≥30 kg/m2, postpartum hemorrhage, postcesarean delivery, preeclampsia, or smoking) 1 2* 1 3 CHC: For persons with other risk factors for VTE, these risk factors might increase the classification to a category 4.
Breastfeeding: Breastfeeding provides important health benefits for breastfeeding parent and infant. The U.S. Dietary Guidelines for Americans and American Academy of Pediatrics recommend that infants be exclusively breastfed for about the first 6 months with continued breastfeeding while introducing appropriate complementary foods for 1 year or longer (1) or up to age 2 years or longer (2).
  ii. Without other risk factors for VTE 1 1 1 2 Breastfeeding provides important health benefits for breastfeeding parent and infant. The U.S. Dietary Guidelines for Americans and American Academy of Pediatrics recommend that infants be exclusively breastfed for about the first 6 months with continued breastfeeding while introducing appropriate complementary foods for 1 year or longer (1) or up to age 2 years or longer (2).
d. >42 days postpartum 1 1 1 2 Breastfeeding provides important health benefits for breastfeeding parent and infant. The U.S. Dietary Guidelines for Americans and American Academy of Pediatrics recommend that infants be exclusively breastfed for about the first 6 months with continued breastfeeding while introducing appropriate complementary foods for 1 year or longer (1) or up to age 2 years or longer (2).
Postpartum (nonbreastfeeding)
a. <21 days postpartum 1 2* 1 4
b. 21–42 days postpartum
  i. With other risk factors for VTE (e.g., age ≥35 years, previous VTE, thrombophilia, immobility, transfusion at delivery, peripartum cardiomyopathy, BMI ≥30 kg/m2, postpartum hemorrhage, postcesarean delivery, preeclampsia, or smoking) 1 2* 1 3 CHC: For persons with other risk factors for VTE, these risk factors might increase the classification to a category 4.
  ii. Without other risk factors for VTE 1 1 1 2
c. >42 days postpartum 1 1 1 1
Postpartum (including cesarean
delivery, breastfeeding, or
nonbreastfeeding)
a. <10 minutes after delivery of the placenta 2* 2* IUD: Postpartum placement of IUDs is safe and does not appear to increase health risks associated with IUD use such as infection. Higher rates of expulsion during the postpartum period should be considered as they relate to effectiveness, along with patient access to interval placement (i.e., not related to pregnancy) when expulsion rates are lower.
Breastfeeding: Breastfeeding provides important health benefits for breastfeeding parent and infant. The U.S. Dietary Guidelines for Americans and American Academy of Pediatrics recommend that infants be exclusively breastfed for about the first 6 months with continued breastfeeding while introducing appropriate complementary foods for 1 year or longer (1) or up to age 2 years or longer (2).
b. 10 minutes after delivery of the placenta to <4 weeks 2 2 IUD: Postpartum placement of IUDs is safe and does not appear to increase health risks associated with IUD use such as infection. Higher rates of expulsion during the postpartum period should be considered as they relate to effectiveness, along with patient access to interval placement (i.e., not related to pregnancy) when expulsion rates are lower.
Breastfeeding: Breastfeeding provides important health benefits for breastfeeding parent and infant. The U.S. Dietary Guidelines for Americans and American Academy of Pediatrics recommend that infants be exclusively breastfed for about the first 6 months with continued breastfeeding while introducing appropriate complementary foods for 1 year or longer (1) or up to age 2 years or longer (2).
c. ≥4 weeks 1 1 IUD: Postpartum placement of IUDs is safe and does not appear to increase health risks associated with IUD use such as infection. Higher rates of expulsion during the postpartum period should be considered as they relate to effectiveness, along with patient access to interval placement (i.e., not related to pregnancy) when expulsion rates are lower.
Breastfeeding: Breastfeeding provides important health benefits for breastfeeding parent and infant. The U.S. Dietary Guidelines for Americans and American Academy of Pediatrics recommend that infants be exclusively breastfed for about the first 6 months with continued breastfeeding while introducing appropriate complementary foods for 1 year or longer (1) or up to age 2 years or longer (2).
d. Postpartum sepsis 4 4
Postabortion (spontaneous
or induced)
a. First trimester abortion
  i. Procedural (surgical)* 1 1 1 1 1 1 IUD: IUDs may be placed immediately after abortion completion.
POC: POCs may be started immediately after abortion completion or at time of medication abortion initiation.
DMPA: After a first trimester medication abortion that did not include mifepristone, there is no restriction for the use of DMPA (category 1). After a first trimester medication abortion that included mifepristone, there is no restriction for use of DMPA after abortion completion (category 1) and benefits generally outweigh risks with DMPA use immediately at time of medication abortion initiation (category 2). Concurrent administration of DMPA with mifepristone might slightly decrease medication abortion effectiveness and increase risk for ongoing pregnancy. Risk for ongoing pregnancy with concurrent administration of DMPA with mifepristone should be considered along with personal preference and access to follow-up abortion and contraceptive care.*
CHC: CHCs may be started immediately after abortion completion or at time of medication abortion initiation.
  ii. Medication* 1 1 1 1/2* 1 1
  iii. Spontaneous abortion with no intervention* 1 1 1 1 1 1
b. Second trimester
abortion
  i. Procedural (surgical)* 2 2 1 1 1 1 IUD: IUDs may be placed immediately after abortion completion.
POC: POCs may be started immediately after abortion completion or at time of medication abortion initiation.
CHC: CHCs may be started immediately after abortion completion or at time of medication abortion initiation.
  ii. Medication* 2 2 1 1 1 1
  iii. Spontaneous abortion with no intervention* 2 2 1 1 1 1
c. Immediate postseptic abortion 4 4 1 1 1 1 POC: POCs may be started immediately after abortion completion or at time of medication abortion initiation.
CHC: CHCs may be started immediately after abortion completion or at time of medication abortion initiation.
Obesity
a. BMI ≥30 kg/m2 1 1 1 1 1 2 CHC: Risk for thrombosis increases with multiple risk factors, such as obesity, older age (e.g., ≥40 years), diabetes, smoking, family history of thrombosis, and dyslipidemia. When a person has multiple risk factors, any of which alone would increase risk for thrombosis, use of CHCs might increase thrombosis risk to an unacceptable level. However, a simple addition of categories for multiple risk factors is not intended; for example, a combination of two category 2 risk factors might not necessarily warrant a higher category.*
b. Menarche to <18 years and BMI ≥30 kg/m2 1 1 1 2 1 2 CHC: Risk for thrombosis increases with multiple risk factors, such as obesity, older age (e.g., ≥40 years), diabetes, smoking, family history of thrombosis, and dyslipidemia. When a person has multiple risk factors, any of which alone would increase risk for thrombosis, use of CHCs might increase thrombosis risk to an unacceptable level. However, a simple addition of categories for multiple risk factors is not intended; for example, a combination of two category 2 risk factors might not necessarily warrant a higher category.*
Surgery
a. Minor surgery without immobilization 1 1 1 1 1 1
b. Major surgery
  i. Without prolonged immobilization 1 1 1 1 1 2
  ii. With prolonged immobilization 1 1* 1* 2 1* 4
Deep venous thrombosis/
Pulmonary embolism

This condition is associated
with increased risk for adverse
health events as a result of
pregnancy (Box 3).
a. Current or history of DVT/PE, receiving anticoagulant therapy (therapeutic dose) (e.g., acute DVT/PE or long-term therapeutic dose)* 2 2 2 2 2 3* Cu-IUD: Persons using anticoagulant therapy are at risk for gynecologic complications of therapy, such as heavy or prolonged bleeding. Cu-IUDs might worsen bleeding.*
LNG-IUD: Persons using anticoagulant therapy are at risk for gynecologic complications of therapy, such as heavy or prolonged bleeding. LNG-IUDs can be of benefit in preventing or treating this complication. When a contraceptive method is used as a therapy, rather than solely to prevent pregnancy, the risk/benefit ratio might differ and should be considered on a case-by-case basis.*
POC: Persons using anticoagulant therapy are at risk for gynecologic complications of therapy, such as heavy or prolonged bleeding and hemorrhagic ovarian cysts. POCs can be of benefit in preventing or treating these complications; benefits might vary by POC dose and formulation. When a contraceptive method is used as a therapy, rather than solely to prevent pregnancy, the risk/benefit ratio might differ and should be considered on a case-by-case basis.*
CHC: Persons using anticoagulant therapy are at risk for gynecologic complications of therapy, such as heavy or prolonged bleeding and hemorrhagic ovarian cysts. CHCs can be of benefit in preventing or treating these complications. When a contraceptive method is used as a therapy, rather than solely to prevent pregnancy, the risk/benefit ratio might differ and should be considered on a case-by-case basis.*
CHC: When a patient discontinues therapeutic dose of anticoagulant therapy, careful consideration should be given to transitioning from CHCs to a progestin-only or nonhormonal method, if acceptable to the patient.*
b. History of DVT/PE,
receiving anticoagulant
therapy (prophylactic dose)*
Cu-IUD: Persons using anticoagulant therapy are at risk for gynecologic complications of therapy, such as heavy or prolonged bleeding. Cu-IUDs might worsen bleeding.*
LNG-IUD: Persons using anticoagulant therapy are at risk for gynecologic complications of therapy, such as heavy or prolonged bleeding. LNG-IUDs can be of benefit in preventing or treating this complication. When a contraceptive method is used as a therapy, rather than solely to prevent pregnancy, the risk/benefit ratio might differ and should be considered on a case-by-case basis.*
POC: Persons using anticoagulant therapy are at risk for gynecologic complications of therapy, such as heavy or prolonged bleeding and hemorrhagic ovarian cysts. POCs can be of benefit in preventing or treating these complications; benefits might vary by POC dose and formulation. When a contraceptive method is used as a therapy, rather than solely to prevent pregnancy, the risk/benefit ratio might differ and should be considered on a case-by-case basis.*
CHC: Persons using anticoagulant therapy are at risk for gynecologic complications of therapy, such as heavy or prolonged bleeding and hemorrhagic ovarian cysts. CHCs can be of benefit in preventing or treating these complications. When a contraceptive method is used as a therapy, rather than solely to prevent pregnancy, the risk/benefit ratio might differ and should be considered on a case-by-case basis.*
  i. Higher risk for recurrent DVT/PE (one or more risk factors)* 2 2 2 3* 2 4
• Thrombophilia (e.g., factor V Leiden mutation; prothrombin gene mutation; protein S, protein C, and antithrombin deficiencies; or antiphospholipid syndrome)*
• Active cancer (metastatic, receiving therapy, or within 6 months after clinical remission), excluding nonmelanoma skin cancer*
• History of recurrent DVT/PE*
  ii. Lower risk for recurrent DVT/PE (no risk factors)* 2 2 2 2 2 3
c. History of DVT/PE, not
receiving anticoagulant therapy*
  i. Higher risk for recurrent DVT/PE (one or more risk factors)* 1 2 2 3* 2 4
• History of estrogen-associated DVT/PE
• Pregnancy-associated DVT/PE*
• Idiopathic DVT/PE*
• Thrombophilia (e.g., factor V Leiden mutation; prothrombin gene mutation; protein S, protein C, or antithrombin deficiencies; or antiphospholipid syndrome)*
• Active cancer (metastatic, receiving therapy, or within 6 months after clinical remission), excluding nonmelanoma skin
cancer*
• History of recurrent DVT/PE*
  ii. Lower risk for recurrent DVT/PE (no risk factors)* 1 2 2 2 2 3
d. Family history (first-degree relatives) 1 1 1 1 1 2
Thrombophilia (e.g., factor V Leiden mutation; prothrombin gene mutation; protein S, protein C, and antithrombin deficiencies; or antiphospholipid syndrome)
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 2 2 3* 2 4 Routine screening in the general population before contraceptive initiation is not recommended.
If a person has current or history of DVT/PE, see recommendations for DVT/PE.*
Classification of antiphospholipid syndrome includes presence of a clinical feature (e.g., thrombosis or obstetric morbidity) and persistently abnormal antiphospholipid antibody test on two or more occasions at least 12 weeks apart (3).*
Superficial venous disorders
a. Varicose veins 1 1 1 1 1 1
b. Superficial venous thrombosis (acute or history) 1 1 1 2* 1 3 CHC: Superficial venous thrombosis might be associated with an increased risk for VTE. If a person has risk factors for concurrent DVT (e.g., thrombophilia or cancer) or has current or history of DVT, see recommendations for DVT/PE. Superficial venous thrombosis associated with a peripheral intravenous catheter is less likely to be associated with additional thrombosis and use of CHCs may be considered.
Valvular heart disease
Complicated valvular heart
disease is a condition associated
with increased risk for adverse
health events as a result of
pregnancy (Box 3).
a. Uncomplicated 1 1 1 1 1 2
b. Complicated (pulmonary hypertension, risk for atrial fibrillation, or history of subacute bacterial endocarditis) 1 1 1 2* 1 4
Peripartum cardiomyopathy
This condition is associated with
increased risk for adverse health
events as a result of pregnancy
(Box 3).
a. Normal or mildly impaired
cardiac function (New York
Heart Association Functional
Class I or II: no limitation of
activities or slight, mild limitation
of activity) (4)
  i. <6 months 2 2 1 2* 1 4
  ii. ≥6 months 2 2 1 2* 1 3
b. Moderately or severely impaired cardiac function (New York Heart Association Functional Class III or IV: marked limitation of activity or should be at complete rest) (4) 2 2 2 3* 2 4
Chronic kidney disease*
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
Initiation Continuation Initiation Continuation
a. Current nephrotic syndrome* 1* 1* 2* 2* 2* 3* 2*
DRSP POP with known hyperkalemia: 4*
4* DRSP POP: Persons with known hyperkalemia should not use DRSP POPs because of the risk for worsening hyperkalemia (category 4). For persons with CKD without known hyperkalemia (category 2), consider checking serum potassium level during first cycle of DRSP POPs.*
b. Hemodialysis* 1* 1* 2* 2* 2* 3* 2*
DRSP POP with known hyperkalemia: 4*
4* DRSP POP: Persons with known hyperkalemia should not use DRSP POPs because of the risk for worsening hyperkalemia (category 4). For persons with CKD without known hyperkalemia (category 2), consider checking serum potassium level during first cycle of DRSP POPs.*
c. Peritoneal dialysis* 2* 1* 2* 2* 2* 3* 2*
DRSP POP with known hyperkalemia: 4*
4* DRSP POP: Persons with known hyperkalemia should not use DRSP POPs because of the risk for worsening hyperkalemia (category 4). For persons with CKD without known hyperkalemia (category 2), consider checking serum potassium level during first cycle of DRSP POPs.*
Systemic lupus erythematosus
This condition is associated with
increased risk for adverse health
events as a result of pregnancy
(Box 3).
Initiation Continuation
a. Positive (or unknown) antiphospholipid antibodies 1 1 2* 2* 3 3 2* 4 Persons with SLE are at increased risk for ischemic heart disease, stroke, and VTE. Categories assigned to such conditions in U.S. MEC should be the same for persons with SLE who have these conditions. For all subconditions of SLE, classifications are based on the assumption that no other risk factors for cardiovascular disease are present; these classifications must be modified in the presence of such risk factors.
b. Severe thrombocytopenia 3 2 2 2 3 2 2 2 Persons with SLE are at increased risk for ischemic heart disease, stroke, and VTE. Categories assigned to such conditions in U.S. MEC should be the same for persons with SLE who have these conditions. For all subconditions of SLE, classifications are based on the assumption that no other risk factors for cardiovascular disease are present; these classifications must be modified in the presence of such risk factors.
Severe thrombocytopenia increases the risk for bleeding. The category should be assessed according to the severity of thrombocytopenia and its clinical manifestations. In persons with very severe thrombocytopenia who are at risk for spontaneous bleeding, consultation with a specialist and certain pretreatments might be warranted.
c. Immunosuppressive therapy 2 1 2 2 2 2 2 2 Persons with SLE are at increased risk for ischemic heart disease, stroke, and VTE. Categories assigned to such conditions in U.S. MEC should be the same for persons with SLE who have these conditions. For all subconditions of SLE, classifications are based on the assumption that no other risk factors for cardiovascular disease are present; these classifications must be modified in the presence of such risk factors.
d. None of the above 1 1 2 2 2 2 2 2 Persons with SLE are at increased risk for ischemic heart disease, stroke, and VTE. Categories assigned to such conditions in U.S. MEC should be the same for persons with SLE who have these conditions. For all subconditions of SLE, classifications are based on the assumption that no other risk factors for cardiovascular disease are present; these classifications must be modified in the presence of such risk factors.
High risk for HIV infection Initiation Continuation Initiation Continuation 1 1 1 1 IUD: Many persons at high risk for HIV infection are also at risk for other STIs (see recommendations for Sexually transmitted infections in U.S. MEC and recommendations on STI screening before IUD placement in U.S. SPR [https://www.cdc.gov/contraception/hcp/usspr]) (5).*
1* 1* 1* 1*
Cirrhosis
Decompensated cirrhosis is
associated with increased risk
for adverse health events as a
result of pregnancy (Box 3).
a. Compensated (normal liver function) 1 1 1 1 1 1
b. Decompensated (impaired liver function) 1 2* 2* 3 2* 4
Liver tumors
Hepatocelluar adenoma
and malignant liver tumors are
associated with increased risk for
adverse health events as a result
of pregnancy (Box 3).
a. Benign
  i. Focal nodular hyperplasia 1 2 2 2 2 2
  ii. Hepatoceullular adenoma 1 2* 2* 3 2* 4
b. Malignant (hepatocellular carcinoma) 1 3 3 3 3 4
Sickle cell disease
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
2 1 1 2/3* 1 4* DMPA: The category should be assessed according to the severity of the condition and risk for thrombosis.*
Solid organ transplantation
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
Initiation Continuation Initiation Continuation
a. No graft failure 1* 1* 1* 1* 2 2/3* 2 2 DMPA: DMPA use among persons receiving long-term immunosuppressive therapy with a history of, or risk factors for, nontraumatic fractures is classified as category 3. Otherwise, DMPA use for persons with solid organ transplantation is classified as category 2.*
CHC: Persons with transplant due to Budd-Chiari syndrome should not use CHCs because of the increased risk for thrombosis.*
b. Graft failure 2* 1* 2* 1* 2 2/3* 2 4 DMPA: DMPA use among persons receiving long-term immunosuppressive therapy with a history of, or risk factors for, nontraumatic fractures is classified as category 3. Otherwise, DMPA use for persons with solid organ transplantation is classified as category 2.*
Antiretrovirals used for
prevention (PrEP) or treatment
of HIV infection*,†
See the following guidelines for the most up-to-date recommendations on drug-drug interactions between hormonal contraception and antiretrovirals: 1) Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions to Reduce Perinatal HIV Transmission in the United States (https://clinicalinfo.hiv.gov/en/guidelines/perinatal/prepregnancy-counseling-childbearing-age-overview?view=full#table-3) (6) and 2) Guidelines for the Use of Antiretroviral Agents in Adults and Adolescents With HIV (https://clinicalinfo.hiv.gov/en/guidelines/hiv-clinical-guidelines-adult-and-adolescent-arv/drug-interactions-overview?view=full) (7).

Abbreviations: ARV = antiretroviral; BMI = body mass index; CHC = combined hormonal contraceptive; CKD = chronic kidney disease; Cu-IUD = copper intrauterine device; DMPA = depot medroxyprogesterone acetate; DRSP = drospirenone; DVT = deep venous thrombosis; IUD = intrauterine device; LNG-IUD = levonorgestrel intrauterine device; PE = pulmonary embolism; POC = progestin-only contraceptive; POP = progestin-only pill; PrEP = pre-exposure prophylaxis; SLE = systemic lupus erythematous; STI = sexually transmitted infection; U.S. MEC = U.S. Medical Eligibility Criteria for Contraceptive Use; U.S. SPR = U.S. Selected Practice Recommendations for Contraceptive Use; VTE = venous thromboembolism.
* Indicates a condition for which the classification changed for one or more contraceptive methods or for which the condition description underwent a substantive modification.
U.S. MEC recommendations for concurrent use of hormonal contraceptives or IUDs and ARVs for treatment of HIV infection also apply to use of ARVs for PrEP.

TABLE A2. Summary of changes for barrier methods from U.S. Medical Eligibility Criteria for Contraceptive Use, 2016
Condition Condom Spermicide/Vaginal pH modulator*,† Diaphragm/Cap (with spermicide) Clarification
Chronic kidney disease*
This condition is associated with increased risk
for adverse health events as a result of
pregnancy (Box 3).
a. Current nephrotic syndrome* 1* 1* 1*
b. Hemodialysis* 1* 1* 1*
c. Peritoneal dialysis* 1* 1* 1*
Cervical cancer (awaiting treatment) 1 Vaginal pH modulator: 1*
Spermicide: 2
1 The cap should not be used. Diaphragm use has no restrictions.
High risk for HIV infection 1 Vaginal pH modulator: 1*
Spermicide: 4
4
HIV infection
For persons with HIV infection who are not clinically well or not receiving ARV therapy, this condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 Vaginal pH modulator: 1*
Spermicide: 3
3
Antiretrovirals used for prevention (PrEP) or treatment of HIV infection* 1 1/3/4* 3/4 No drug interaction between ARV therapy and barrier method use is known. HIV infection is classified as category 1 for vaginal pH modulator and category 3 for spermicide and diaphragm or cap (see recommendations for HIV infection). High risk for HIV infection is classified as category 1 for vaginal pH modulator and category 4 for spermicide and diaphragm or cap (see recommendations for High risk for HIV infection).*

Abbreviations: ARV = antiretroviral; PrEP = pre-exposure prophylaxis.
* Indicates a condition for which the classification changed for one or more contraceptive methods or for which the condition description underwent a substantive modification.
The contraceptive method “Spermicide” has been changed to “Spermicide/Vaginal pH modulator.” Recommendations for “Spermicide/Vaginal pH modulator” are the same as those previously for “Spermicide,” with exceptions noted.
§ U.S. Medical Eligibility Criteria for Contraceptive Use recommendations for concurrent use of barrier methods and ARVs for treatment of HIV infection also apply to use of ARVs for PrEP.

TABLE A3. Summary of changes for emergency contraception from U.S. Medical Eligibility Criteria for Contraceptive Use, 2016
Condition Category Clarification
Cu-IUD UPA LNG COC
Solid organ transplantation
This condition is associated with
increased risk for adverse health
events as a result of pregnancy
(Box 3).
a. No graft failure 1* 1 1 1
b. Graft failure 2* 1 1 1

Abbreviations: COC = combined oral contraceptive; Cu-IUD = copper intrauterine device; LNG = levonorgestrel; UPA = ulipristal acetate.
* Indicates a condition for which the classification changed for one or more contraceptive methods or for which the condition description underwent a substantive modification.

References

  1. US Department of Agriculture; US Department of Health and Human Services. Dietary guidelines for Americans, 2020–2025. 9th ed. Washington, DC: US Department of Agriculture and US Department of Health and Human Services; 2020. https://www.dietaryguidelines.gov/sites/default/files/2021-03/Dietary_Guidelines_for_Americans-2020-2025.pdf
  2. Meek JY, Noble L; Section on Breastfeeding. Policy statement: breastfeeding and the use of human milk. 2022;150:e2022057988. https://doi.org/10.1542/peds.2022-057988 PMID:35921640
  3. Barbhaiya M, Zuily S, Naden R, et al.; ACR/EULAR APS Classification Criteria Collaborators. The 2023 ACR/EULAR Antiphospholipid Syndrome Classification Criteria. 2023;75:1687–702. https://doi.org/10.1002/art.42624 PMID:37635643
  4. The Criteria Committee of the New York Heart Association. Nomenclature and criteria for diagnosis of diseases of the heart and great vessels. 9th ed. Boston, MA: Little, Brown and Co; 1994.
  5. Curtis KM, Nguyen AT, Tepper NK, et al. U.S. selected practice recommendations for contraceptive use, 2024. 2024;73(No. RR-3):1–77.
  6. Panel on Treatment of HIV During Pregnancy and Prevention of Perinatal Transmission. Recommendations for the use of antiretroviral drugs during pregnancy and interventions to reduce perinatal HIV transmission in the United States. Washington, DC: US Department of Health and Human Services; 2023. https://clinicalinfo.hiv.gov/en/guidelines/perinatal/recommendations-arv-drugs-pregnancy-overview
  7. Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of antiretroviral agents in adults and adolescents with HIV. Washington, DC: US Department of Health and Human Services; 2023. https://clinicalinfo.hiv.gov/sites/default/files/guidelines/documents/adult-adolescent-arv/guidelines-adult-adolescent-arv.pdf

Appendix B: Classifications for Intrauterine Devices

Classifications for intrauterine devices (IUDs) are for the copper (380 mm2) and levonorgestrel (13.5 mg, 19.5 mg, or 52 mg) IUDs (Box B1) (Table B1). IUDs do not protect against sexually transmitted infections (STIs), including HIV infection, and patients using IUDs should be counseled that consistent and correct use of external (male) latex condoms reduces the risk for STIs, including HIV infection (1). Use of internal (female) condoms can provide protection from transmission of STIs, although data are limited (1). Patients also should be counseled that pre-exposure prophylaxis, when taken as prescribed, is highly effective for preventing HIV infection (2).

Return to your place in the textBOX B1. Categories for classifying intrauterine devices

U.S. MEC 1 = A condition for which there is no restriction for the use of the contraceptive method

U.S. MEC 2 = A condition for which the advantages of using the method generally outweigh the theoretical or proven risks

U.S. MEC 3 = A condition for which the theoretical or proven risks usually outweigh the advantages of using the method

U.S. MEC 4 = A condition that represents an unacceptable health risk if the contraceptive method is used

Abbreviation: U.S. MEC = U.S. Medical Eligibility Criteria for Contraceptive Use.



TABLE B1. Classifications for intrauterine devices, including the copper intrauterine device and levonorgestrel intrauterine device
Condition Category Clarification/Evidence/Comment
Cu-IUD LNG-IUD
Personal Characteristics and Reproductive History
Pregnancy 4 4 Clarification: The IUD is not indicated during pregnancy and should not be used because of the risk for serious pelvic infection and septic spontaneous abortion.
Age
a. Menarche to <20 years 2 2 Comment: Concern exists both about the risk for expulsion from nulliparity and for STIs from sexual behavior in younger age groups (see U.S. SPR for recommendations on STI screening before IUD placement (https://www.cdc.gov/contraception/hcp/usspr) (3).
b. ≥20 years 1 1
Parity
a. Nulliparous 2 2 Evidence: Data conflict about whether IUD use is associated with infertility among nulliparous women, although well-conducted studies suggest no increased risk (412).
b. Parous 1 1
Postpartum (including
cesarean delivery,
breastfeeding, or
nonbreastfeeding)
a. <10 minutes after delivery of the placenta 2 2 Clarification: Postpartum placement of IUDs is safe and does not appear to increase health risks associated with IUD use such as infection. Higher rates of expulsion during the postpartum period should be considered as they relate to effectiveness, along with patient access to interval placement (i.e., not related to pregnancy) when expulsion rates are lower.
Clarification (breastfeeding): Breastfeeding provides important health benefits for breastfeeding parent and infant. The U.S. Dietary Guidelines for Americans and American Academy of Pediatrics recommend that infants be exclusively breastfed for about the first 6 months with continued breastfeeding while introducing appropriate complementary foods for 1 year or longer (13) or up to age 2 years or longer (14).
Evidence: Studies suggest that immediate postplacental (<10 minutes) and early postpartum (10 minutes up until 72 hours) placement of Cu-IUDs and LNG-IUDs is associated with increased risk for expulsion compared with interval placement (i.e., not related to pregnancy). A meta-analysis found an increased risk for expulsion with immediate postplacental placement (8.6%; range = 0%–31.9%) and early postpartum placement (25.1%; range = 3.5%–46.7%) compared with interval placement (1.6%; range = 0%–4.8%) (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516). Although immediate postplacental placement at the time of cesarean delivery might have increased risk for expulsion compared with interval placement, risk appears lower than that for placement at the time of vaginal delivery. Evidence for infection, perforation, and removals for pain or bleeding are limited; however, these events are rare (1567).
Evidence (breastfeeding): Two RCTs found conflicting results on breastfeeding outcomes when LNG-IUDs were initiated immediately postpartum compared with 6–8 weeks postpartum. Initiation of LNG-IUDs immediately postpartum had no other harmful effect on infant health, growth, or development (19,68). Breastfeeding women using IUDs do not have an increased risk for certain IUD-related adverse events including expulsion, infection, pain, or bleeding compared with nonbreastfeeding women. The risk for perforation is increased independently among breastfeeding women and among women ≤36 weeks postpartum, compared with nonpostpartum women; however, the absolute risk for perforation remains low (1567,69).
Comment: Risk factors for breastfeeding difficulties include previous breastfeeding difficulties, certain medical conditions, certain perinatal complications, and preterm birth. For all breastfeeding persons, with or without risk factors for breastfeeding difficulties, discussions about contraception should include information about risks, benefits, and alternatives.
b. 10 minutes after delivery of the placenta to <4 weeks 2 2 Clarification: Postpartum placement of IUDs is safe and does not appear to increase health risks associated with IUD use such as infection. Higher rates of expulsion during the postpartum period should be considered as they relate to effectiveness, along with patient access to interval placement (i.e., not related to pregnancy) when expulsion rates are lower.
Clarification (breastfeeding): Breastfeeding provides important health benefits for breastfeeding parent and infant. The U.S. Dietary Guidelines for Americans and American Academy of Pediatrics recommend that infants be exclusively breastfed for about the first 6 months with continued breastfeeding while introducing appropriate complementary foods for 1 year or longer (13) or up to age 2 years or longer (14).
Evidence: Studies suggest that immediate postplacental (<10 minutes) and early postpartum (10 minutes up until 72 hours) placement of Cu-IUDs and LNG-IUDs is associated with increased risk for expulsion compared with interval placement (i.e., not related to pregnancy). A meta-analysis found an increased risk for expulsion with immediate postplacental placement (8.6%; range = 0%–31.9%) and early postpartum placement (25.1%; range = 3.5%–46.7%) compared with interval placement (1.6%; range = 0%–4.8%) (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516). Although immediate postplacental placement at the time of cesarean delivery might have increased risk for expulsion compared with interval placement, risk appears lower than that for placement at the time of vaginal delivery. Evidence for infection, perforation, and removals for pain or bleeding are limited; however, these events are rare (1567).
Evidence (breastfeeding): Two RCTs found conflicting results on breastfeeding outcomes when LNG-IUDs were initiated immediately postpartum compared with 6–8 weeks postpartum. Initiation of LNG-IUDs immediately postpartum had no other harmful effect on infant health, growth, or development (19,68). Breastfeeding women using IUDs do not have an increased risk for certain IUD-related adverse events including expulsion, infection, pain, or bleeding compared with nonbreastfeeding women. The risk for perforation is increased independently among breastfeeding women and among women ≤36 weeks postpartum, compared with nonpostpartum women; however, the absolute risk for perforation remains low (1567,69).
Comment: Risk factors for breastfeeding difficulties include previous breastfeeding difficulties, certain medical conditions, certain perinatal complications, and preterm birth. For all breastfeeding persons, with or without risk factors for breastfeeding difficulties, discussions about contraception should include information about risks, benefits, and alternatives.
c. ≥4 weeks 1 1 Clarification: Postpartum placement of IUDs is safe and does not appear to increase health risks associated with IUD use such as infection. Higher rates of expulsion during the postpartum period should be considered as they relate to effectiveness, along with patient access to interval placement (i.e., not related to pregnancy) when expulsion rates are lower.
Clarification (breastfeeding): Breastfeeding provides important health benefits for breastfeeding parent and infant. The U.S. Dietary Guidelines for Americans and American Academy of Pediatrics recommend that infants be exclusively breastfed for about the first 6 months with continued breastfeeding while introducing appropriate complementary foods for 1 year or longer (13) or up to age 2 years or longer (14).
Evidence (breastfeeding): Initiation of LNG-IUDs at 4 weeks postpartum or later demonstrated no detrimental effect on breastfeeding outcomes and no harmful effect on infant health, growth, or development (19,68). Breastfeeding women using IUDs do not have an increased risk for certain IUD-related adverse events including expulsion, infection, pain, or bleeding compared with nonbreastfeeding women. The risk for perforation is increased independently among breastfeeding women and among women ≤36 weeks postpartum, compared with nonpostpartum women; however, the absolute risk for perforation remains low (1567,69).
Comment: Risk factors for breastfeeding difficulties include previous breastfeeding difficulties, certain medical conditions, certain perinatal complications, and preterm birth. For all breastfeeding persons, with or without risk factors for breastfeeding difficulties, discussions about contraception should include information about risks, benefits, and alternatives.
d. Postpartum sepsis 4 4 Comment: Theoretical concern exists that postpartum placement of an IUD in a person with recent chorioamnionitis or current endometritis might be associated with increased complications.
Postabortion
(spontaneous or induced)
a. First trimester abortion Clarification: IUDs may be placed immediately after abortion completion.
Evidence: Risk for complications from immediate versus delayed placement of an IUD after abortion did not differ. Expulsion was greater when an IUD was placed after a second trimester procedural abortion than when placed after a first trimester procedural abortion. Safety or expulsion for postabortion placement of an LNG-IUD did not differ from that of a Cu-IUD (70).
  i. Procedural (surgical) 1 1
  ii. Medication 1 1
  iii. Spontaneous abortion with no intervention 1 1
b. Second trimester abortion
  i. Procedural (surgical) 2 2
  ii. Medication 2 2
  iii. Spontaneous abortion with no intervention 2 2
c. Immediate postseptic abortion 4 4 Comment: Placement of an IUD might substantially worsen the condition.
Past ectopic pregnancy 1 1 Comment: The absolute risk for ectopic pregnancy is extremely low because of the high effectiveness of IUDs. However, when a person becomes pregnant during IUD use, the relative likelihood of ectopic pregnancy increases substantially.
History of pelvic surgery
(see recommendations for Postpartum [including cesarean delivery])
1 1
Smoking
a. Age <35 years 1 1
b. Age ≥35 years
  i. <15 cigarettes per day 1 1
  ii. ≥15 cigarettes per day 1 1
Obesity
a. BMI ≥30 kg/m2 1 1
b. Menarche to <18 years and BMI ≥30 kg/m2 1 1
History of bariatric surgery
This condition is associated with
increased risk for adverse health
events as a result
of pregnancy (Box 3).
a. Restrictive procedures: decrease storage capacity of the stomach (vertical banded gastroplasty, laparoscopic adjustable gastric band, or laparoscopic sleeve gastrectomy) 1 1
b. Malabsorptive procedures: decrease absorption of nutrients and calories by shortening the functional length of the small intestine (Roux-en-Y gastric bypass or biliopancreatic diversion) 1 1
Surgery
a. Minor surgery without immobilization 1 1
b. Major surgery
  i. Without prolonged immobilization 1 1
  ii. With prolonged immobilization 1 1 Evidence: No direct evidence was identified on risk for thrombosis with POC use among those undergoing major surgery (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
Cardiovascular Disease
Multiple risk factors for atherosclerotic cardiovascular disease (e.g., older age, smoking, diabetes, hypertension, low HDL, high LDL, or high triglyceride levels) 1 2
Hypertension
Systolic blood pressure
≥160 mm Hg or diastolic blood
pressure ≥100 mm Hg are
associated with increased risk
for adverse health events as a
result of pregnancy (Box 3).
a. Adequately controlled hypertension 1 1 Clarification: For all categories of hypertension, classifications are based on the assumption that no other risk factors for cardiovascular disease exist. When multiple risk factors do exist, risk for cardiovascular disease might increase substantially. A single reading of blood pressure level is not sufficient to classify a person as hypertensive.
b. Elevated blood pressure levels
(properly taken measurements)
Clarification: For all categories of hypertension, classifications are based on the assumption that no other risk factors for cardiovascular disease exist. When multiple risk factors do exist, risk for cardiovascular disease might increase substantially. A single reading of blood pressure level is not sufficient to classify a person as hypertensive.
Comment: Theoretical concern exists about the effect of LNG on lipids. Use of Cu-IUDs has no restrictions.
  i. Systolic 140–159 mm Hg or diastolic 90–99 mm Hg 1 1
  ii. Systolic ≥160 mm Hg or diastolic ≥100 mm Hg 1 2
c. Vascular disease 1 2 Clarification: For all categories of hypertension, classifications are based on the assumption that no other risk factors for cardiovascular disease exist. When multiple risk factors do exist, risk for cardiovascular disease might increase substantially. A single reading of blood pressure level is not sufficient to classify a person as hypertensive.
Comment: Theoretical concern exists about the effect of LNG on lipids. Use of Cu-IUDs has no restrictions.
History of high blood pressure during pregnancy (when current blood pressure is measurable and normal) 1 1
Deep venous thrombosis/
Pulmonary embolism

This condition is associated with
increased risk for adverse health
events as a result
of pregnancy (Box 3).
a. Current or history of DVT/PE, receiving anticoagulant therapy (therapeutic dose) (e.g., acute DVT/PE or long-term therapeutic dose) 2 2 Clarification (Cu-IUD): Persons using anticoagulant therapy are at risk for gynecologic complications of therapy, such as heavy or prolonged bleeding. Cu-IUDs might worsen bleeding.
Clarification (LNG-IUD): Persons using anticoagulant therapy are at risk for gynecologic complications of therapy, such as heavy or prolonged bleeding. LNG-IUDs can be of benefit in preventing or treating this complication. When a contraceptive method is used as a therapy, rather than solely to prevent pregnancy, the risk/benefit ratio might differ and should be considered on a case-by-case basis.
Evidence: Limited evidence was identified on use of POCs or Cu-IUDs among women with acute DVT/PE receiving anticoagulant therapy (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516). In one study among women with a history of acute VTE currently receiving therapeutic anticoagulant therapy (i.e., rivaroxaban or enoxaparin/vitamin K antagonist [warfarin or acenocoumarol]), the incidence of recurrent VTE was similar among estrogen users (CHC or estrogen-only pills), POC users, and women not on hormonal therapy (71). Limited evidence suggests that placement of a Cu-IUD or LNG-IUD does not increase risk for bleeding complications in women receiving anticoagulant therapy (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
b. History of DVT/PE, receiving
anticoagulant therapy
(prophylactic dose)
Clarification (Cu-IUD): Persons using anticoagulant therapy are at risk for gynecologic complications of therapy, such as heavy or prolonged bleeding. Cu-IUDs might worsen bleeding.
Clarification (LNG-IUD): Persons using anticoagulant therapy are at risk for gynecologic complications of therapy, such as heavy or prolonged bleeding. LNG-IUDs can be of benefit in preventing or treating this complication. When a contraceptive method is used as a therapy, rather than solely to prevent pregnancy, the risk/benefit ratio might differ and should be considered on a case-by-case basis.
Evidence: Limited evidence suggests that placement of the LNG-IUD does not increase risk for bleeding complications in women receiving anticoagulant therapy (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
  i. Higher risk for recurrent DVT/PE (one or more risk factors) 2 2
• Thrombophilia (e.g., factor V Leiden mutation; prothrombin gene mutation; protein S, protein C, and antithrombin deficiencies; or antiphospholipid syndrome)
• Active cancer (metastatic, receiving therapy, or within 6 months after clinical remission), excluding nonmelanoma skin cancer
• History of recurrent DVT/PE
  ii. Lower risk for recurrent DVT/PE (no risk factors) 2 2
c. History of DVT/PE, not
receiving anticoagulant therapy
  i. Higher risk for recurrent DVT/PE (one or more risk factors) 1 2
• History of estrogen-associated DVT/PE
• Pregnancy-associated DVT/PE
• Idiopathic DVT/PE
• Thrombophilia (e.g., factor V Leiden mutation; prothrombin gene mutation; protein S, protein C, and antithrombin deficiencies; or antiphospholipid syndrome)
• Active cancer (metastatic, receiving therapy, or within 6 months after clinical remission), excluding nonmelanoma skin cancer
• History of recurrent DVT/PE
  ii. Lower risk for recurrent DVT/PE (no risk factors) 1 2
d. Family history (first-degree relatives) 1 1
Thrombophilia (e.g., factor V Leiden mutation; prothrombin gene mutation; protein S, protein C, and antithrombin deficiencies; or antiphospholipid syndrome)
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 2 Clarification: Routine screening in the general population before contraceptive initiation is not recommended.
Clarification: If a person has current or history of DVT/PE, see recommendations for DVT/PE.
Clarification: Classification of antiphospholipid syndrome includes presence of a clinical feature (e.g., thrombosis or obstetric morbidity) and persistently abnormal antiphospholipid antibody test on two or more occasions at least 12 weeks apart (72).
Evidence: Limited evidence was identified on LNG-IUD use among persons with thrombophilia. Among women with factor V Leiden mutation, one study found that women using LNG-IUD had similar risk for venous thrombosis as those not using hormonal contraception (73). No evidence was identified on POC use among persons with prothrombin gene mutation, protein S deficiency, protein C deficiency, antithrombin deficiency, or antiphospholipid syndrome (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
Superficial venous disorders
a. Varicose veins 1 1
b. Superficial venous thrombosis (acute or history) 1 1
Current and history of ischemic heart disease
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 Initiation Continuation Comment: Theoretical concern exists about the effect of LNG on lipids. Use of Cu-IUDs has no restrictions.
2 3
Stroke (history of cerebrovascular accident)
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 2 Comment: Theoretical concern exists about the effect of LNG on lipids. Use of Cu-IUDs has no restrictions.
Valvular heart disease
Complicated valvular heart disease
is associated with increased risk for
adverse health events as a result of
pregnancy (Box 3).
Comment: According to the American Heart Association, administration of prophylactic antibiotics solely to prevent endocarditis is not recommended for patients who undergo genitourinary tract procedures, including placement or removal of IUDs (74).
a. Uncomplicated 1 1
b. Complicated (pulmonary hypertension, risk for atrial fibrillation, or history of subacute bacterial endocarditis) 1 1
Peripartum cardiomyopathy
This condition is associated with
increased risk for adverse health
events as a result of
pregnancy (Box 3).
Evidence: No direct evidence exists on the safety of IUDs among women with peripartum cardiomyopathy. Limited indirect evidence from noncomparative studies did not demonstrate any cases of arrhythmia or infective endocarditis in women with cardiac disease who used IUDs (75).
Comment: IUD placement might induce cardiac arrhythmias in healthy persons; persons with peripartum cardiomyopathy have a high incidence of cardiac arrhythmias.
a. Normal or mildly impaired
cardiac function (New York Heart
Association Functional Class I or II:
no limitation of activities or slight,
mild limitation of activity) (76)
  i. <6 months 2 2
  ii. ≥6 months 2 2
b. Moderately or severely impaired cardiac function (New York Heart Association Functional Class III or IV: marked limitation of activity or should be at complete rest) (76) 2 2
Renal Disease
Chronic kidney disease
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
Initiation Continuation Initiation Continuation
a. Current nephrotic syndrome 1 1 2 2 Comment: A person might have CKD without current nephrotic syndrome, but might have other conditions often associated with CKD (e.g., diabetes, hypertension, SLE). See recommendations for other conditions if they apply.
b. Hemodialysis 1 1 2 2 Evidence: No comparative studies were identified on the safety of IUD use among persons with CKD on hemodialysis (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516). One case report of LNG-IUD use in a person with CKD on hemodialysis reported improved abnormal uterine bleeding and anemia (77).
Comment: A person might have CKD without hemodialysis, but might have other conditions often associated with CKD (e.g., diabetes, hypertension, and SLE). See recommendations for other conditions if they apply.
c. Peritoneal dialysis 2 1 2 2 Evidence: No comparative studies were identified on IUD use among persons with CKD on peritoneal dialysis (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516). Four case reports of IUD use among women with CKD on peritoneal dialysis identified one case of peritoneal allergic reaction (78), three cases of peritonitis (7880) and one case of TOA (78).
Comment: A person might have CKD without peritoneal dialysis, but might have other conditions often associated with CKD (e.g., diabetes, hypertension, and SLE). See recommendations for other conditions if they apply.
Rheumatic Diseases
Systemic lupus erythematosus
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
Initiation Continuation
a. Positive (or unknown) antiphospholipid antibodies 1 1 2 Clarification: Persons with SLE are at increased risk for ischemic heart disease, stroke, and VTE. Categories assigned to such conditions in U.S. MEC should be the same for persons with SLE who have these conditions. For all subconditions of SLE, classifications are based on the assumption that no other risk factors for cardiovascular disease are present; these classifications must be modified in the presence of such risk factors (8199).
Evidence: No direct evidence was identified on POC use among persons with SLE with antiphospholipid antibodies (100) (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
b. Severe thrombocytopenia 3 2 2 Clarification: Persons with SLE are at increased risk for ischemic heart disease, stroke, and VTE. Categories assigned to such conditions in U.S. MEC should be the same for persons with SLE who have these conditions. For all subconditions of SLE, classifications are based on the assumption that no other risk factors for cardiovascular disease are present; these classifications must be modified in the presence of such risk factors (8199).
Clarification: Severe thrombocytopenia increases the risk for bleeding. The category should be assessed according to the severity of thrombocytopenia and its clinical manifestations. In persons with very severe thrombocytopenia who are at risk for spontaneous bleeding, consultation with a specialist and certain pretreatments might be warranted.
Evidence: The LNG-IUD might be a useful treatment for menorrhagia in women with severe thrombocytopenia (94).
c. Immunosuppressive therapy 2 1 2 Clarification: Persons with SLE are at increased risk for ischemic heart disease, stroke, and VTE. Categories assigned to such conditions in U.S. MEC should be the same for persons with SLE who have these conditions. For all subconditions of SLE, classifications are based on the assumption that no other risk factors for cardiovascular disease are present; these classifications must be modified in the presence of such risk factors (8199).
d. None of the above 1 1 2 Clarification: Persons with SLE are at increased risk for ischemic heart disease, stroke, and VTE. Categories assigned to such conditions in U.S. MEC should be the same for persons with SLE who have these conditions. For all subconditions of SLE, classifications are based on the assumption that no other risk factors for cardiovascular disease are present; these classifications must be modified in the presence of such risk factors (8199).
Rheumatoid arthritis Initiation Continuation Initiation Continuation
a. Not receiving immunosuppressive therapy 1 1 1 1
b. Receiving immunosuppressive therapy 2 1 2 1
Neurologic Conditions
Headaches
a. Nonmigraine (mild or severe) 1 1
b. Migraine
  i. Without aura (includes menstrual migraine) 1 1 Evidence: No studies directly examined the risk for stroke among women with migraine using LNG-IUDs (101). Limited evidence demonstrated that women using LNG-IUDs do not have an increased risk for ischemic stroke compared with women not using hormonal contraceptives (102).
Comment: Menstrual migraine is a subtype of migraine without aura. For more information see the International Headache Society’s International Classification of Headache Disorders, 3rd ed. (https://ichd-3.org) (103).
  ii. With aura 1 1
Epilepsy
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 1
Multiple sclerosis
a. Without prolonged immobility 1 1
b. With prolonged immobility 1 1
Depressive Disorders
Depressive disorders 1 1 Clarification: If a person is receiving psychotropic medications or St. John’s wort, see recommendations for Drug Interactions.
Evidence: The frequency of psychiatric hospitalizations for women with bipolar disorder or depression did not significantly differ among women using DMPA, LNG-IUD, Cu-IUD, or sterilization (104).
Reproductive Tract Infections and Disorders
Vaginal bleeding patterns Initiation Continuation
a. Irregular pattern without heavy bleeding 1 1 1
b. Heavy or prolonged bleeding (includes regular and irregular patterns) 2 1 2 Clarification: Unusually heavy bleeding should raise suspicion of a serious underlying condition.
Evidence: Evidence from studies examining the treatment effects of the LNG-IUD among women with heavy or prolonged bleeding reported no increase in adverse effects and found the LNG-IUD to be beneficial in treating menorrhagia (105112).
Unexplained vaginal bleeding (suspicious for serious condition) before evaluation Initiation Continuation Initiation Continuation Clarification: If pregnancy or an underlying pathological condition (e.g., pelvic malignancy) is suspected, it must be evaluated and the category adjusted after evaluation. The IUD does not need to be removed before evaluation.
4 2 4 2
Endometriosis 2 1 Evidence: LNG-IUD use among women with endometriosis decreased dysmenorrhea, pelvic pain, and dyspareunia (113117).
Benign ovarian tumors (including cysts) 1 1
Severe dysmenorrhea 2 1 Comment: Dysmenorrhea might intensify with Cu-IUD use. LNG-IUD use has been associated with reduction of dysmenorrhea.
Gestational trophoblastic disease
This condition is associated with
increased risk for adverse health
events as a result
of pregnancy (Box 3).
a. Suspected gestational trophoblastic
disease (immediate postevacuation)
Clarification: For all subconditions of gestational trophoblastic disease, classifications are based on the assumption that persons are under close medical supervision because of the need for monitoring of β-hCG levels for appropriate disease surveillance.
Evidence: Limited evidence suggests that women using an IUD after uterine evacuation for a molar pregnancy are not at greater risk for postmolar trophoblastic disease than are women using other methods of contraception (118).
Comment: The risk for expulsion immediately postevacuation for gestational trophoblastic disease is unknown. Expulsion is greater after IUD placement immediately postevacuation for a spontaneous or induced abortion in the second trimester compared with IUD placement after a first trimester abortion.
  i. Uterine size first trimester 1 1
  ii. Uterine size second trimester 2 2
b. Confirmed gestational trophoblastic disease (after initial evacuation and during monitoring) Initiation Continuation Initiation Continuation
  i. Undetectable or nonpregnant β-hCG levels 1 1 1 1 Clarification: For all subconditions of gestational trophoblastic disease, classifications are based on the assumption that persons are under close medical supervision because of the need for monitoring of β-hCG levels for appropriate disease surveillance.
Evidence: Limited evidence suggests that women using an IUD after uterine evacuation for a molar pregnancy are not at greater risk for postmolar trophoblastic disease than are women using other methods of contraception (118).
Comment: Once β-hCG levels have decreased to nonpregnant levels, the risk for disease progression is likely to be very low.
  ii. Decreasing β-hCG levels 2 1 2 1 Clarification: For all subconditions of gestational trophoblastic disease, classifications are based on the assumption that persons are under close medical supervision because of the need for monitoring of β-hCG levels for appropriate disease surveillance.
Clarification: For persons at higher risk for disease progression, the benefits of effective contraception must be weighed against the potential need for early IUD removal.
Evidence: Limited evidence suggests that women using an IUD after uterine evacuation for a molar pregnancy are not at greater risk for postmolar trophoblastic disease than are women using other methods of contraception (118).
  iii. Persistently elevated β-hCG levels or malignant disease, with no evidence or suspicion of intrauterine disease 2 1 2 1 Clarification: For all subconditions of gestational trophoblastic disease, classifications are based on the assumption that persons are under close medical supervision because of the need for monitoring of β-hCG levels for appropriate disease surveillance.
Evidence: Limited evidence suggests that women using an IUD after uterine evacuation for a molar pregnancy are not at greater risk for postmolar trophoblastic disease than are women using other methods of contraception (118).
  iv. Persistently elevated β-hCG levels or malignant disease, with evidence or suspicion of intrauterine disease 4 2 4 2 Clarification: For all subconditions of gestational trophoblastic disease, classifications are based on the assumption that persons are under close medical supervision because of the need for monitoring of β-hCG levels for appropriate disease surveillance.
Evidence: Limited evidence suggests that women using an IUD after uterine evacuation for a molar pregnancy are not at greater risk for postmolar trophoblastic disease than are women using other methods of contraception (118).
Comment: For persons with suspected or confirmed intrauterine disease, an IUD should not be placed because of theoretical risk for perforation, infection, and hemorrhage. For persons who already have an IUD in place, individual circumstance along with the benefits of effective contraception must be weighed against theoretical risks of either removal or continuation of the IUD.
Cervical ectropion 1 1
Cervical intraepithelial neoplasia 1 2 Comment: Theoretical concern exists that LNG-IUDs might enhance progression of cervical intraepithelial neoplasia.
Cervical cancer (awaiting treatment) Initiation Continuation Initiation Continuation Comment: Concern exists about the increased risk for infection and bleeding at placement. The IUD most likely will need to be removed at the time of treatment but until then, the person is at risk for pregnancy.
4 2 4 2
Breast disease
Breast cancer is associated with
increased risk for adverse health
events as a result
of pregnancy (Box 3).
a. Undiagnosed mass 1 2 Clarification (LNG-IUD): Evaluation of mass should be pursued as early as possible.
b. Benign breast disease 1 1
c. Family history of cancer 1 1
d. Breast cancer Comment: Breast cancer is a hormonally sensitive tumor. Concerns about progression of the disease might be less with LNG-IUDs than with COCs or higher-dose POCs.
  i. Current 1 4
  ii. Past and no evidence of current disease for 5 years 1 3
Endometrial hyperplasia 1 1 Evidence: Among women with endometrial hyperplasia, no adverse health events occurred with LNG-IUD use; most women experienced disease regression (119).
Endometrial cancer
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
Initiation Continuation Initiation Continuation Comment: Concern exists about the increased risk for infection, perforation, and bleeding at placement. The IUD most likely will need to be removed at the time of treatment, but until then, the person is at risk for pregnancy.
4 2 4 2
Ovarian cancer
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 1 Comment: Persons with ovarian cancer who undergo fertility-sparing treatment and need contraception can use an IUD.
Uterine fibroids 2 2 Evidence: Among women with uterine fibroids using an LNG-IUD, most experienced improvements in serum levels of hemoglobin, hematocrit, and ferritin and in menstrual blood loss (120). Rates of LNG-IUD expulsion were higher in women with uterine fibroids (11%) than in women without fibroids (0%–3%); these findings were either not statistically significant or significance testing was not conducted (120). Rates of expulsion found in noncomparative studies ranged from 0%–20% (120).
Comment: Persons with heavy or prolonged bleeding should be assigned the category for that condition.
Anatomical abnormalities
a. Distorted uterine cavity (any congenital or acquired uterine abnormality distorting the uterine cavity in a manner that is incompatible with IUD placement) 4 4 Comment: An anatomical abnormality that distorts the uterine cavity might preclude proper IUD placement.
b. Other abnormalities (including cervical stenosis or cervical lacerations) not distorting the uterine cavity or interfering with IUD placement 2 2
Pelvic inflammatory disease Initiation Continuation Initiation Continuation Clarification (continuation): Treat the PID using appropriate antibiotics. The IUD usually does not need to be removed if the person wants to continue using it. Continued use of an IUD depends on the person’s informed choice and current risk factors for STIs and PID.
Evidence: Among IUD users treated for PID, clinical course did not differ regardless of whether the IUD was removed or left in place (121).
a. Current PID 4 2 4 2
b. Past PID Comment: IUDs do not protect against STIs, including HIV infection, or PID. In persons at low risk for STIs, IUD placement poses little risk for PID.
  i. With subsequent pregnancy 1 1 1 1
  ii. Without subsequent pregnancy 2 2 2 2
Sexually transmitted infections Initiation Continuation Initiation Continuation
a. Current purulent cervicitis or chlamydial infection or gonococcal infection 4 2 4 2 Clarification (continuation): Treat the STI using appropriate antibiotics. The IUD usually does not need to be removed if the person wants to continue using it. Continued use of an IUD depends on the person’s informed choice and current risk factors for STIs and PID.
Evidence: Among women who had an IUD placed, the absolute risk for subsequent PID was low among women with STI at the time of placement but greater than among women with no STI at the time of IUD placement (122128).
b. Vaginitis (including Trichomonas vaginalis and bacterial vaginosis) 2 2 2 2
c. Other factors related to STIs 2 2 2 2 Clarification (initiation): Most persons do not require additional STI screening at the time of IUD placement. If a person with risk factors for STIs has not been screened for gonorrhea and chlamydia according to CDC STI treatment guidelines (1), screening may be performed at the time of IUD placement and placement should not be delayed.
Evidence: Women who undergo same-day STI screening and IUD placement have low incidence rates of PID. Algorithms for predicting PID among women with risk factors for STIs have poor predictive value. Risk for PID among women with risk factors for STIs is low (129).
HIV
High risk for HIV infection Initiation Continuation Initiation Continuation Clarification: Many persons at high risk for HIV infection are also at risk for other STIs (see recommendations for Sexually transmitted infections in U.S. MEC and recommendations on STI screening before IUD placement in U.S. SPR (https://www.cdc.gov/contraception/hcp/usspr) (3).
Evidence: High-quality evidence from one RCT, along with low-quality evidence from two observational studies, suggested no increased risk for HIV acquisition with Cu-IUD use. No studies were identified for LNG-IUDs (130132).
1 1 1 1
HIV infection
For persons with HIV infection
who are not clinically well or not
receiving ARV therapy, this
condition is associated with
increased risk for adverse health
events as a result of
pregnancy (Box 3).
Evidence: Among IUD users, limited evidence demonstrates a low risk for PID among HIV-infected women using IUDs and no higher risk for pelvic infectious complications in HIV-infected than in HIV-noninfected women or among women with varying degrees of HIV severity. IUD use did not adversely affect progression of HIV infection during 6–45 months of follow-up or when compared with hormonal contraceptive use among HIV-infected women. Furthermore, IUD use among HIV-infected women was not associated with increased risk for transmission to sex partners or with increased genital viral shedding (133).
a. Clinically well receiving ARV therapy 1 1 1 1
b. Not clinically well or not receiving ARV therapy 2 1 2 1
Other Infections
Schistosomiasis
Schistosomiasis with fibrosis of
the liver is associated with
increased risk for adverse health
events as a result of
pregnancy (Box 3).
a. Uncomplicated 1 1
b. Fibrosis of the liver (if severe, see recommendations for Cirrhosis) 1 1
Tuberculosis
This condition is associated with
increased risk for adverse health
events as a result of pregnancy
(Box 3).
Initiation Continuation Initiation Continuation
a. Nonpelvic 1 1 1 1
b. Pelvic 4 3 4 3 Comment: Placement of an IUD might substantially worsen the condition.
Malaria 1 1
Endocrine Conditions
Diabetes
Insulin-dependent diabetes
diabetes with nephropathy,
retinopathy, or neuropathy;
diabetes with other vascular
disease; or diabetes of >20 years’
duration are associated with
increased risk for adverse health
events as a result of pregnancy
(Box 3).
a. History of gestational disease 1 1
b. Nonvascular disease Evidence: Limited evidence on the use of the LNG-IUD among women with insulin-dependent or non–insulin-dependent diabetes suggests that these methods have little effect on short-term or long-term diabetes control (e.g., glycosylated hemoglobin levels), hemostatic markers, or lipid profile (134,135).
  i. Non-insulin dependent 1 2
  ii. Insulin dependent 1 2
c. Nephropathy, retinopathy, or neuropathy 1 2
d. Other vascular disease or diabetes of >20 years’ duration 1 2
Thyroid disorders
a. Simple goiter 1 1
b. Hyperthyroid 1 1
c. Hypothyroid 1 1
Gastrointestinal Conditions
Inflammatory bowel disease (ulcerative colitis or Crohn’s disease) 1 1 Evidence: Although two case reports described three women with IBD who experienced exacerbation of disease 5 days–25 months after LNG-IUD placement (136), no comparative studies have examined the safety of IUD use among women with IBD (136).
Gallbladder disease
a. Asymptomatic 1 2
b. Symptomatic
  i. Current 1 2
  ii. Treated by cholecystectomy 1 2
  iii. Medically treated 1 2
History of cholestasis
a. Pregnancy related 1 1
b. Past COC related 1 2 Comment: Concern exists that history of COC related cholestasis might predict subsequent cholestasis with LNG use. Whether risk exists with use of LNG-IUD is unclear.
Viral hepatitis
a. Acute or flare 1 1 Evidence: No direct evidence was identified on IUD use among persons with viral hepatitis (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
b. Chronic 1 1 Evidence: No direct evidence was identified on IUD use among persons with viral hepatitis (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
Cirrhosis
Decompensated cirrhosis is
associated with increased risk for
adverse health events as a result of
pregnancy (Box 3).
a. Compensated (normal liver function) 1 1 Evidence: No direct evidence was identified on IUD use among persons with cirrhosis (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
b. Decompensated (impaired liver function) 1 2 Evidence: No direct evidence was identified on IUD use among persons with cirrhosis (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
Comment: Hepatic metabolism of exogenous hormones might be impaired in persons with liver dysfunction, which could lead to increased progestin levels in circulation and progestin-related side effects and adverse events, which might vary by dose and formulation. Any progestin-related hepatotoxicity might be less tolerated in persons with existing liver dysfunction.
Liver tumors
Hepatocellular adenoma and
malignant liver tumors are
associated with increased risk for
adverse health events as a result
of pregnancy (Box 3).
a. Benign
  i. Focal nodular hyperplasia 1 2 Evidence: Limited evidence suggests that progestin use does not influence either progression or regression of focal nodular hyperplasia (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
  ii. Hepatocellular adenoma 1 2 Evidence: Limited evidence suggests that hepatocellular adenomas generally regress or remain stable during progestin use (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
b. Malignant (hepatocellular carcinoma) 1 3 Evidence: No direct evidence was identified on IUD use among persons with hepatocellular carcinoma (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
Respiratory Conditions
Cystic fibrosis
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 1 Clarification: Persons with cystic fibrosis are at increased risk for diabetes, liver disease, gallbladder disease, and VTE (particularly related to use of central venous catheters) and are frequently prescribed antibiotics. Categories assigned to such conditions in U.S. MEC should be the same for persons with cystic fibrosis who have these conditions. For cystic fibrosis, classifications are based on the assumption that no other conditions are present; these classifications must be modified in the presence of such conditions.
Hematologic Conditions
Thalassemia 2 1 Comment: Concern exists about an increased risk for blood loss with Cu-IUDs.
Sickle cell disease
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
2 1 Comment: Concern exists about an increased risk for blood loss with Cu-IUDs.
Iron deficiency anemia 2 1 Comment: Concern exists about an increased risk for blood loss with Cu-IUDs.
Solid Organ Transplantation
Solid organ transplantation
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
Initiation Continuation Initiation Continuation Evidence: Limited evidence suggests that LNG-IUD use among solid organ transplantation recipients does not increase risk for pelvic infections or decrease contraceptive effectiveness over time or compared with persons without solid organ transplantation No evidence was identified for Cu-IUD (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
a. No graft failure 1 1 1 1
b. Graft failure 2 1 2 1
Drug Interactions
Antiretrovirals used for prevention (PrEP) or treatment of HIV Initiation Continuation Initiation Continuation Clarification: No known interaction exists between ARV therapy and IUD use. However, for persons with HIV infection, IUD placement is classified as category 2 if the person is not clinically well or not receiving ARV therapy. Otherwise, both placement and continuation are classified as category 1 (see recommendations for HIV infection). For persons at high risk for HIV infection, IUDs are category 1 for initiation and continuation (see recommendations for High risk for HIV infection).
a. Nucleoside reverse
transcriptase inhibitors
(NRTIs)
  i. Abacavir (ABC) 1/2 1 1/2 1
  ii. Tenofovir (TDF) 1/2 1 1/2 1
  iii. Zidovudine (AZT) 1/2 1 1/2 1
  iv. Lamivudine (3TC) 1/2 1 1/2 1
  v. Didanosine (DDI) 1/2 1 1/2 1
  vi. Emtricitabine (FTC) 1/2 1 1/2 1
  vii. Stavudine (D4T) 1/2 1 1/2 1
b. Nonnucleoside reverse
transcriptase inhibitors (NNRTIs)
  i. Efavirenz (EFV) 1/2 1 1/2 1
  ii. Etravirine (ETR) 1/2 1 1/2 1
  iii. Nevirapine (NVP) 1/2 1 1/2 1
  iv. Rilpivirine (RPV) 1/2 1 1/2 1
c. Ritonavir-boosted protease
inhibitors
  i. Ritonavir-boosted atazanavir (ATV/r) 1/2 1 1/2 1
  ii. Ritonavir-boosted darunavir (DRV/r) 1/2 1 1/2 1
  iii. Ritonavir-boosted fosamprenavir (FPV/r) 1/2 1 1/2 1
  iv. Ritonavir-boosted lopinavir (LPV/r) 1/2 1 1/2 1
  v. Ritonavir-boosted saquinavir (SQV/r) 1/2 1 1/2 1
  vi. Ritonavir-boosted tipranavir (TPV/r) 1/2 1 1/2 1
d. Protease inhibitors without
ritonavir
  i. Atazanavir (ATV) 1/2 1 1/2 1
  ii. Fosamprenavir (FPV) 1/2 1 1/2 1
  iii. Indinavir (IDV) 1/2 1 1/2 1
  iv. Nelfinavir (NFV) 1/2 1 1/2 1
e. CCR5 co-receptor antagonists
  i. Maraviroc (MVC) 1/2 1 1/2 1
f. HIV integrase strand transfer
inhibitors
  i. Raltegravir (RAL) 1/2 1 1/2 1
  ii. Dolutegravir (DTG) 1/2 1 1/2 1
  iii. Elvitegravir (EVG) 1/2 1 1/2 1
g. Fusion inhibitors
  i. Enfuvirtide 1/2 1 1/2 1
Anticonvulsant therapy
a. Certain anticonvulsants (phenytoin, carbamazepine, barbiturates, primidone, topiramate, and oxcarbazepine) 1 1 Evidence: Limited evidence suggests use of certain anticonvulsants does not interfere with the contraceptive effectiveness of the LNG-IUD (137,138).
b. Lamotrigine 1 1 Evidence: No drug interactions have been reported among women with epilepsy who are receiving lamotrigine and using the LNG-IUD (138,139).
Antimicrobial therapy
a. Broad-spectrum antibiotics 1 1
b. Antifungals 1 1
c. Antiparasitics 1 1
d. Rifampin or rifabutin therapy 1 1 Evidence: One cross-sectional survey found that rifabutin had no impact on the effectiveness of the LNG-IUD (137).
Psychotropic medications
a. Selective serotonin reuptake inhibitors (SSRIs) 1 1 Comment: For many common psychotropic agents, limited or no theoretical concern exists for clinically significant drug interactions when co-administered with hormonal contraceptives. However, either no or very limited data exist examining potential interactions for these classes of medications.
St. John’s wort 1 1

Abbreviations: ARV = antiretroviral; BMI = body mass index; CHC = combined hormonal contraceptive; CKD = chronic kidney disease; COC = combined oral contraceptive; Cu-IUD = copper intrauterine device; DMPA = depot medroxyprogesterone acetate; DVT = deep venous thrombosis; hCG = human chorionic gonadotropin; HDL = high-density lipoprotein; IBD = inflammatory bowel disease; IUD = intrauterine device; LDL = low-density lipoprotein; LNG = levonorgestrel; LNG-IUD = levonorgestrel intrauterine device; NA = not applicable; PE = pulmonary embolism; PID = pelvic inflammatory disease; POC = progestin-only contraceptive; PrEP = pre-exposure prophylaxis; RCT = randomized clinical trial; SLE = systemic lupus erythematous; STI = sexually transmitted infection; TOA = tubo-ovarian abscess; U.S. MEC = U.S. Medical Eligibility Criteria for Contraceptive Use; U.S. SPR = U.S. Selected Practice Recommendations for Contraceptive Use; VTE = venous thromboembolism.

References

  1. Workowski KA, Bachmann LH, Chan PA, et al. Sexually transmitted infections treatment guidelines, 2021. 2021;70(No. RR-4):1–187. https://doi.org/10.15585/mmwr.rr7004a1 PMID:34292926
  2. CDC. US Public Health Service preexposure prophylaxis for the prevention of HIV infection in the United States—2021 update: a clinical practice guideline. Atlanta, GA: US Department of Health and Human Services, CDC; 2021. https://www.cdc.gov/hiv/pdf/risk/prep/cdc-hiv-prep-guidelines-2021.pdf
  3. Curtis KM, Nguyen AT, Tepper NK, et al. U.S. selected practice recommendations for contraceptive use, 2024. 2024;73(No. RR-3):1–77.
  4. Cramer DW, Schiff I, Schoenbaum SC, et al. Tubal infertility and the intrauterine device. 1985;312:941–7. https://doi.org/10.1056/NEJM198504113121502 PMID:3974684
  5. Daling JR, Weiss NS, Metch BJ, et al. Primary tubal infertility in relation to the use of an intrauterine device. 1985;312:937–41. https://doi.org/10.1056/NEJM198504113121501 PMID:3974683
  6. Daling JR, Weiss NS, Voigt LF, McKnight B, Moore DE. The intrauterine device and primary tubal infertility. 1992;326:203–4. https://doi.org/10.1056/NEJM199201163260314 PMID:1727559
  7. Delbarge W, Bátár I, Bafort M, et al. Return to fertility in nulliparous and parous women after removal of the GyneFix intrauterine contraceptive system. 2002;7:24–30. https://doi.org/10.1080/ejc.7.1.24.30 PMID:12041861
  8. Doll H, Vessey M, Painter R. Return of fertility in nulliparous women after discontinuation of the intrauterine device: comparison with women discontinuing other methods of contraception. 2001;108:304–14. https://doi.org/10.1111/j.1471-0528.2001.00075.x PMID:11281473
  9. Hubacher D, Lara-Ricalde R, Taylor DJ, Guerra-Infante F, Guzmán-Rodríguez R. Use of copper intrauterine devices and the risk of tubal infertility among nulligravid women. 2001;345:561–7. https://doi.org/10.1056/NEJMoa010438 PMID:11529209
  10. Skjeldestad FE, Bratt H. Return of fertility after use of IUDs (Nova-T, MLCu250 and MLCu375). 1987;3:139–45. https://doi.org/10.1007/BF01890702 PMID:3630823
  11. Urbach DR, Marrett LD, Kung R, Cohen MM. Association of perforation of the appendix with female tubal infertility. 2001;153:566–71. https://doi.org/10.1093/aje/153.6.566 PMID:11257064
  12. Wilson JC. A prospective New Zealand study of fertility after removal of copper intrauterine contraceptive devices for conception and because of complications: a four-year study. 1989;160:391–6. https://doi.org/10.1016/0002-9378(89)90455-9 PMID:2916624
  13. US Department of Agriculture; US Department of Health and Human Services. Dietary guidelines for Americans, 2020–2025. 9th ed. Washington, DC: US Department of Agriculture and US Department of Health and Human Services; 2020. https://www.dietaryguidelines.gov/sites/default/files/2021-03/Dietary_Guidelines_for_Americans-2020-2025.pdf
  14. Meek JY, Noble L; Section on Breastfeeding. Policy statement: breastfeeding and the use of human milk. 2022;150:e2022057988. https://doi.org/10.1542/peds.2022-057988 PMID:35921640
  15. Annus J, Brat T, Diethelm MP, et al. ; World Health Organization Special. Comparative multicentre trial of three IUDs inserted immediately following delivery of the placenta. 1980;22:9–18. https://doi.org/10.1016/0010-7824(80)90112-2 PMID:7418410
  16. Apelo RA, Waszak CS. Postpartum IUD insertions in Manila, Philippines. 1985;1:319–28. https://doi.org/10.1007/BF01849307 PMID:3842222
  17. Baldwin MK, Edelman AB, Lim JY, Nichols MD, Bednarek PH, Jensen JT. Intrauterine device placement at 3 versus 6 weeks postpartum: a randomized trial. 2016;93:356–63. https://doi.org/10.1016/j.contraception.2015.12.006 PMID:26686914
  18. Bonilla Rosales F, Aguilar Zamudio ME, Cázares Montero ML, Hernández Ortiz ME, Luna Ruiz MA. [Factors for expulsion of intrauterine device Tcu380A applied immediately postpartum and after a delayed period]. 2005;43:5–10. PMID:15998475
  19. Braniff K, Gomez E, Muller R. A randomised clinical trial to assess satisfaction with the levonorgestrel-releasing intrauterine system inserted at caesarean section compared to postpartum placement. 2015;55:279–83. https://doi.org/10.1111/ajo.12335 PMID:26053465
  20. Bryant AG, Kamanga G, Stuart GS, Haddad LB, Meguid T, Mhango C. Immediate postpartum versus 6-week postpartum intrauterine device insertion: a feasibility study of a randomized controlled trial. 2013;17:72–9. PMID:24069753
  21. Caliskan E, Oztürk N, Dilbaz BO, Dilbaz S. Analysis of risk factors associated with uterine perforation by intrauterine devices. 2003;8:150–5. https://doi.org/10.1080/ejc.8.3.150.155 PMID:14667326
  22. Celen S, Möröy P, Sucak A, Aktulay A, Danişman N. Clinical outcomes of early postplacental insertion of intrauterine contraceptive devices. 2004;69:279–82. https://doi.org/10.1016/j.contraception.2003.12.004 PMID:15033401
  23. Çelen Ş, Sucak A, Yıldız Y, Danışman N. Immediate postplacental insertion of an intrauterine contraceptive device during cesarean section. 2011;84:240–3. https://doi.org/10.1016/j.contraception.2011.01.006 PMID:21843687
  24. Chen BA, Reeves MF, Hayes JL, Hohmann HL, Perriera LK, Creinin MD. Postplacental or delayed insertion of the levonorgestrel intrauterine device after vaginal delivery: a randomized controlled trial. 2010;116:1079–87. https://doi.org/10.1097/AOG.0b013e3181f73fac PMID:20966692
  25. Chen JH, Wu SC, Shao WQ, et al. The comparative trial of TCu 380A IUD and progesterone-releasing vaginal ring used by lactating women. 1998;57:371–9. https://doi.org/10.1016/S0010-7824(98)00043-2 PMID:9693396
  26. Cohen R, Sheeder J, Arango N, Teal SB, Tocce K. Twelve-month contraceptive continuation and repeat pregnancy among young mothers choosing postdelivery contraceptive implants or postplacental intrauterine devices. 2016;93:178–83. https://doi.org/10.1016/j.contraception.2015.10.001 PMID:26475368
  27. Dahlke JD, Terpstra ER, Ramseyer AM, Busch JM, Rieg T, Magann EF. Postpartum insertion of levonorgestrel—intrauterine system at three time periods: a prospective randomized pilot study. 2011;84:244–8. https://doi.org/10.1016/j.contraception.2011.01.007 PMID:21843688
  28. Dias T, Abeykoon S, Kumarasiri S, Gunawardena C, Padeniya T, D’Antonio F. Use of ultrasound in predicting success of intrauterine contraceptive device insertion immediately after delivery. 2015;46:104–8. https://doi.org/10.1002/uog.14733 PMID:25418016
  29. El-Shafei MMMA, Hassan EO, El-Boghdad L, El-Lakkany N. Postpartum and postabortion intrauterine device insertion unmet needs of safe reproductive health: three years experience of Mansoura University Hospital. 2000;26:253–62.
  30. Elsedeek MS. Puerperal and menstrual bleeding patterns with different types of contraceptive device fitted during elective cesarean delivery. 2012;116:31–4. https://doi.org/10.1016/j.ijgo.2011.07.036 PMID:22036512
  31. Elsedeek MS. Five-year follow-up of two types of contraceptive device fitted during elective cesarean delivery. 2015;130:179–82. https://doi.org/10.1016/j.ijgo.2015.02.031 PMID:25957802
  32. Eroğlu K, Akkuzu G, Vural G, et al. Comparison of efficacy and complications of IUD insertion in immediate postplacental/early postpartum period with interval period: 1 year follow-up. 2006;74:376–81. https://doi.org/10.1016/j.contraception.2006.07.003 PMID:17046378
  33. Gueye M, Gaye YF, Diouf AA, et al. [Trancesarean intra-uterine device. Pilot study performed at Dakar teaching hospital]. 2013;42:585–90. https://doi.org/10.1016/j.jgyn.2013.06.003 PMID:23850420
  34. Gupta S, Malik S, Sinha R, Shyamsunder S, Mittal MK. Association of the position of the Copper T 380A as determined by the ultrasonography following its insertion in the immediate postpartum period with the subsequent complications: an observational study. 2014;64:349–53. https://doi.org/10.1007/s13224-014-0532-5 PMID:25368459
  35. Hagbard L, Ingemanson CA, Sorbe B. Early postpartum insertion of copper IUD. 1978;17:355–63. https://doi.org/10.1016/0010-7824(78)90081-1 PMID:648157
  36. Hayes JL, Cwiak C, Goedken P, Zieman M. A pilot clinical trial of ultrasound-guided postplacental insertion of a levonorgestrel intrauterine device. 2007;76:292–6. https://doi.org/10.1016/j.contraception.2007.06.003 PMID:17900440
  37. Jatlaoui TC, Marcus M, Jamieson DJ, Goedken P, Cwiak C. Postplacental intrauterine device insertion at a teaching hospital. 2014;89:528–33. https://doi.org/10.1016/j.contraception.2013.10.008 PMID:24565735
  38. Jatlaoui TC, Whiteman MK, Jeng G, et al. Intrauterine device expulsion after postpartum placement: a systematic review and meta-analysis. 2018;132:895–905. https://doi.org/10.1097/AOG.0000000000002822 PMID:30204688
  39. Kumar S, Sethi R, Balasubramaniam S, et al. Women’s experience with postpartum intrauterine contraceptive device use in India. 2014;11:32. https://doi.org/10.1186/1742-4755-11-32 PMID:24755312
  40. Laes E, Lehtovirta P, Weintraub D, Pyörälä T, Luukkainen T. Early puerperal insertions of copper-T-200. 1975;11:289–95. https://doi.org/10.1016/0010-7824(75)90037-2 PMID:1116368
  41. Lara Ricalde R, Menocal Tobías G, Ramos Pérez C, Velázquez Ramírez N. [Random comparative study between intrauterine device Multiload Cu375 and TCu 380a inserted in the postpartum period]. 2006;74:306–11. PMID:16970116
  42. Lavin P, Bravo C, Waszak C. Comparison of T Cu 200 and Progestasert IUDs. 1983;4:143–7. PMID:12338635
  43. Lavin P, Waszak C, Bravo C. Preliminary report on a postpartum CuT 200 study, Santiago, Chile. 1983;21:71–5. https://doi.org/10.1016/0020-7292(83)90073-5 PMID:6133798
  44. Lester F, Kakaire O, Byamugisha J, et al. Intracesarean insertion of the Copper T380A versus 6 weeks postcesarean: a randomized clinical trial. 2015;91:198–203. https://doi.org/10.1016/j.contraception.2014.12.002 PMID:25499587
  45. Letti Müller AL, Lopes Ramos JG, Martins-Costa SH, et al. Transvaginal ultrasonographic assessment of the expulsion rate of intrauterine devices inserted in the immediate postpartum period: a pilot study. 2005;72:192–5. https://doi.org/10.1016/j.contraception.2005.03.014 PMID:16102554
  46. Levi E, Cantillo E, Ades V, Banks E, Murthy A. Immediate postplacental IUD insertion at cesarean delivery: a prospective cohort study. 2012;86:102–5. https://doi.org/10.1016/j.contraception.2011.11.019 PMID:22264666
  47. Levi EE, Stuart GS, Zerden ML, Garrett JM, Bryant AG. Intrauterine device placement during cesarean delivery and continued use 6 months postpartum. 2015;126:5–11. https://doi.org/10.1097/AOG.0000000000000882 PMID:26241250
  48. Mishra S. Evaluation of safety, efficacy, and expulsion of post-placental and intra-cesarean insertion of intrauterine contraceptive devices (PPIUCD). 2014;64:337–43. https://doi.org/10.1007/s13224-014-0550-3 PMID:25368457
  49. Morrison C, Waszak C, Katz K, Diabaté F, Mate EM. Clinical outcomes of two early postpartum IUD insertion programs in Africa. 1996;53:17–21. https://doi.org/10.1016/0010-7824(95)00254-5 PMID:8631184
  50. Nelson AL, Chen S, Eden R. Intraoperative placement of the Copper T-380 intrauterine devices in women undergoing elective cesarean delivery: a pilot study. 2009;80:81–3. https://doi.org/10.1016/j.contraception.2009.01.014 PMID:19501220
  51. Newton J, Harper M, Chan KK. Immediate post-placental insertion of intrauterine contraceptive devices. 1977;2:272–4. https://doi.org/10.1016/S0140-6736(77)90955-2 PMID:69881
  52. Prema K, Gayathri TL, Philips FS. Comparative study of early postpartum, postabortal and interval insertion of Cu T 200 mm2 device. 1978;28:946–8. PMID:571822
  53. Puzey M. Mirena at caesarean section. 2005;10:164–7. https://doi.org/10.1080/13625180500233851 PMID:16318963
  54. Ragab A, Hamed HO, Alsammani MA, et al. Expulsion of Nova-T380, Multiload 375, and Copper-T380A contraceptive devices inserted during cesarean delivery. 2015;130:174–8. https://doi.org/10.1016/j.ijgo.2015.03.025 PMID:25975871
  55. Shukla M, Qureshi S; Chandrawati. Post-placental intrauterine device insertion—a five year experience at a tertiary care centre in north India. 2012;136:432–5. PMID:23041736
  56. Singal S, Bharti R, Dewan R, et al. Clinical outcome of postplacental Copper T 380A insertion in women delivering by caesarean section. 2014;8:OC01–04. PMID:25386484
  57. Stuart GS, Bryant AG, O’Neill E, Doherty IA. Feasibility of postpartum placement of the levonorgestrel intrauterine system more than 6 h after vaginal birth. 2012;85:359–62. https://doi.org/10.1016/j.contraception.2011.08.005 PMID:22067759
  58. Stuart GS, Lesko CR, Stuebe AM, Bryant AG, Levi EE, Danvers AI. A randomized trial of levonorgestrel intrauterine system insertion 6 to 48 h compared to 6 weeks after vaginal delivery; lessons learned. 2015;91:284–8. https://doi.org/10.1016/j.contraception.2014.12.009 PMID:25553871
  59. Thiery M, Van Kets H, Van der Pas H. Immediate post-placental IUD insertion: the expulsion problem. 1985;31:331–49. https://doi.org/10.1016/0010-7824(85)90002-2 PMID:4006467
  60. Van Der Pas MT, Delbeke L, Van Dets H. Comparative performance of two copper-wired IUDs (ML Cu 250 and T Cu 200: immediate postpartum and interval insertion. 1980;1:27–35. PMID:12261715
  61. Welkovic S, Costa LO, Faúndes A, de Alencar Ximenes R, Costa CF. Post-partum bleeding and infection after post-placental IUD insertion. 2001;63:155–8. https://doi.org/10.1016/S0010-7824(01)00180-9 PMID:11368989
  62. Whitaker AK, Endres LK, Mistretta SQ, Gilliam ML. Postplacental insertion of the levonorgestrel intrauterine device after cesarean delivery vs. delayed insertion: a randomized controlled trial. 2014;89:534–9. https://doi.org/10.1016/j.contraception.2013.12.007 PMID:24457061
  63. Woo CJ, Alamgir H, Potter JE. Women’s experiences after Planned Parenthood’s exclusion from a family planning program in Texas. 2016;93:298–302. https://doi.org/10.1016/j.contraception.2015.12.004 PMID:26680757
  64. Wu SC; Research Group on Failure Causes and Prevention Measures of Intrauterine Device. [Efficacy of intrauterine device TCu380A when inserted in four different periods]. 2009;44:431–5. PMID:19953943
  65. Xu J, Yang X, Gu X, et al. Comparison between two techniques used in immediate postplacental insertion of TCu 380A intrauterine device: 36-month follow-up. 1999;10:156–62. PMID:12349462
  66. Xu J, Zhuang L, Yu G. [Comparison of two techniques used in immediate postplacental insertion of TCu 380A intrauterine device: 12 month follow-up of 910 cases]. 1997;32:354–7. PMID:9596916
  67. Xu JX, Rivera R, Dunson TR, et al. A comparative study of two techniques used in immediate postplacental insertion (IPPI) of the Copper T-380A IUD in Shanghai, People’s Republic of China. 1996;54:33–8. https://doi.org/10.1016/0010-7824(96)00117-5 PMID:8804806
  68. Phillips SJ, Tepper NK, Kapp N, Nanda K, Temmerman M, Curtis KM. Progestogen-only contraceptive use among breastfeeding women: a systematic review. 2016;94:226–52. https://doi.org/10.1016/j.contraception.2015.09.010 PMID:26410174
  69. Berry-Bibee EN, Tepper NK, Jatlaoui TC, Whiteman MK, Jamieson DJ, Curtis KM. The safety of intrauterine devices in breastfeeding women: a systematic review. 2016;94:725–38. https://doi.org/10.1016/j.contraception.2016.07.006 PMID:27421765
  70. Steenland MW, Tepper NK, Curtis KM, Kapp N. Intrauterine contraceptive insertion postabortion: a systematic review. 2011;84:447–64. https://doi.org/10.1016/j.contraception.2011.03.007 PMID:22018119
  71. Martinelli I, Lensing AW, Middeldorp S, et al. Recurrent venous thromboembolism and abnormal uterine bleeding with anticoagulant and hormone therapy use. 2016;127:1417–25. https://doi.org/10.1182/blood-2015-08-665927 PMID:26696010
  72. Barbhaiya M, Zuily S, Naden R, et al. ; ACR/EULAR APS Classification Criteria Collaborators. The 2023 ACR/EULAR Antiphospholipid Syndrome Classification Criteria. 2023;75:1687–702. https://doi.org/10.1002/art.42624 PMID:37635643
  73. Bergendal A, Persson I, Odeberg J, et al. Association of venous thromboembolism with hormonal contraception and thrombophilic genotypes. 2014;124:600–9. https://doi.org/10.1097/AOG.0000000000000411 PMID:25162263
  74. Wilson W, Taubert KA, Gewitz M, et al. ; American Heart Association Rheumatic Fever, Endocarditis, and Kawasaki Disease Committee; American Heart Association Council on Cardiovascular Disease in the Young; American Heart Association Council on Clinical Cardiology; American Heart Association Council on Cardiovascular Surgery and Anesthesia; Quality of Care and Outcomes Research Interdisciplinary Working Group. Prevention of infective endocarditis: guidelines from the American Heart Association: a guideline from the American Heart Association Rheumatic Fever, Endocarditis, and Kawasaki Disease Committee, Council on Cardiovascular Disease in the Young, and the Council on Clinical Cardiology, Council on Cardiovascular Surgery and Anesthesia, and the Quality of Care and Outcomes Research Interdisciplinary Working Group. 2007;116:1736–54. https://doi.org/10.1161/CIRCULATIONAHA.106.183095 PMID:17446442
  75. Tepper NK, Paulen ME, Marchbanks PA, Curtis KM. Safety of contraceptive use among women with peripartum cardiomyopathy: a systematic review. 2010;82:95–101. https://doi.org/10.1016/j.contraception.2010.02.004 PMID:20682147
  76. The Criteria Committee of the New York Heart Association. Nomenclature and criteria for diagnosis of diseases of the heart and great vessels. 9th ed. Boston, MA: Little, Brown and Co; 1994.
  77. Fedele L, Gammaro L, Bianchi S. Levonorgestrel-releasing intrauterine device for the treatment of menometrorrhagia in a woman on hemodialysis. 1999;341:541. https://doi.org/10.1056/NEJM199908123410718 PMID:10447446
  78. Chen Y, Li Z, Mo L, et al. Eosinophilia in peritoneal effluent due to a levonorgestrel-releasing intrauterine system in a woman on peritoneal dialysis. 2017;37:349–50. https://doi.org/10.3747/pdi.2016.00154 PMID:28512169
  79. Korzets A, Chagnac A, Ori Y, Zevin D, Levi J. Pneumococcal peritonitis complicating CAPD—was the indwelling intrauterine device to blame? 1991;35:24–5. PMID:2007293
  80. Plaza MM. Intrauterine device-related peritonitis in a patient on CAPD. 2002;22:538–9. https://doi.org/10.1177/089686080202200420 PMID:12322833
  81. Bernatsky S, Clarke A, Ramsey-Goldman R, et al. Hormonal exposures and breast cancer in a sample of women with systemic lupus erythematosus. 2004;43:1178–81. https://doi.org/10.1093/rheumatology/keh282 PMID:15226516
  82. Bernatsky S, Ramsey-Goldman R, Gordon C, et al. Factors associated with abnormal Pap results in systemic lupus erythematosus. 2004;43:1386–9. https://doi.org/10.1093/rheumatology/keh331 PMID:15280571
  83. Chopra N, Koren S, Greer WL, et al. Factor V Leiden, prothrombin gene mutation, and thrombosis risk in patients with antiphospholipid antibodies. 2002;29:1683–8. PMID:12180730
  84. Esdaile JM, Abrahamowicz M, Grodzicky T, et al. Traditional Framingham risk factors fail to fully account for accelerated atherosclerosis in systemic lupus erythematosus. 2001;44:2331–7. https://doi.org/10.1002/1529-0131(200110)44:10<2331::AID-ART395>3.0.CO;2-I PMID:11665973
  85. Julkunen HA, Kaaja R, Friman C. Contraceptive practice in women with systemic lupus erythematosus. 1993;32:227–30. https://doi.org/10.1093/rheumatology/32.3.227 PMID:8448613
  86. Manzi S, Meilahn EN, Rairie JE, et al. Age-specific incidence rates of myocardial infarction and angina in women with systemic lupus erythematosus: comparison with the Framingham Study. 1997;145:408–15. https://doi.org/10.1093/oxfordjournals.aje.a009122 PMID:9048514
  87. McAlindon T, Giannotta L, Taub N, D’Cruz D, Hughes G. Environmental factors predicting nephritis in systemic lupus erythematosus. 1993;52:720–4. https://doi.org/10.1136/ard.52.10.720 PMID:8257208
  88. McDonald J, Stewart J, Urowitz MB, Gladman DD. Peripheral vascular disease in patients with systemic lupus erythematosus. 1992;51:56–60. https://doi.org/10.1136/ard.51.1.56 PMID:1540039
  89. Mintz G, Gutiérrez G, Delezé M, Rodríguez E. Contraception with progestagens in systemic lupus erythematosus. 1984;30:29–38. https://doi.org/10.1016/0010-7824(84)90076-3 PMID:6434228
  90. Petri M. Musculoskeletal complications of systemic lupus erythematosus in the Hopkins Lupus Cohort: an update. 1995;8:137–45. https://doi.org/10.1002/art.1790080305 PMID:7654797
  91. Petri M. Lupus in Baltimore: evidence-based ‘clinical pearls’ from the Hopkins Lupus Cohort. 2005;14:970–3. https://doi.org/10.1191/0961203305lu2230xx PMID:16425579
  92. Sánchez-Guerrero J, Uribe AG, Jiménez-Santana L, et al. A trial of contraceptive methods in women with systemic lupus erythematosus. 2005;353:2539–49. https://doi.org/10.1056/NEJMoa050817 PMID:16354890
  93. Sarabi ZS, Chang E, Bobba R, et al. Incidence rates of arterial and venous thrombosis after diagnosis of systemic lupus erythematosus. 2005;53:609–12. https://doi.org/10.1002/art.21314 PMID:16082635
  94. Schaedel ZE, Dolan G, Powell MC. The use of the levonorgestrel-releasing intrauterine system in the management of menorrhagia in women with hemostatic disorders. 2005;193:1361–3. https://doi.org/10.1016/j.ajog.2005.05.002 PMID:16202726
  95. Somers E, Magder LS, Petri M. Antiphospholipid antibodies and incidence of venous thrombosis in a cohort of patients with systemic lupus erythematosus. 2002;29:2531–6. PMID:12465147
  96. Urowitz MB, Bookman AA, Koehler BE, Gordon DA, Smythe HA, Ogryzlo MA. The bimodal mortality pattern of systemic lupus erythematosus. 1976;60:221–5. https://doi.org/10.1016/0002-9343(76)90431-9 PMID:1251849
  97. Julkunen HA. Oral contraceptives in systemic lupus erythematosus: side-effects and influence on the activity of SLE. 1991;20:427–33. https://doi.org/10.3109/03009749109096822 PMID:1771400
  98. Jungers P, Dougados M, Pélissier C, et al. Influence of oral contraceptive therapy on the activity of systemic lupus erythematosus. 1982;25:618–23. https://doi.org/10.1002/art.1780250603 PMID:7092961
  99. Petri M, Kim MY, Kalunian KC, et al. ; OC-SELENA Trial. Combined oral contraceptives in women with systemic lupus erythematosus. 2005;353:2550–8. https://doi.org/10.1056/NEJMoa051135 PMID:16354891
  100. Culwell KR, Curtis KM, Del Carmen Cravioto M. Safety of contraceptive method use among women with systemic lupus erythematosus: a systematic review. 2009;114:341–53. https://doi.org/10.1097/AOG.0b013e3181ae9c64 PMID:19622996
  101. Tepper NK, Whiteman MK, Zapata LB, Marchbanks PA, Curtis KM. Safety of hormonal contraceptives among women with migraine: asystematic review. 2016;94:630–40. https://doi.org/10.1016/j.contraception.2016.04.016 PMID:27153744
  102. Tepper NK, Whiteman MK, Marchbanks PA, James AH, Curtis KM. Progestin-only contraception and thromboembolism: asystematic review. 2016;94:678–700. https://doi.org/10.1016/j.contraception.2016.04.014 PMID:27153743
  103. Headache Classification Committee of the International Headache Society (IHS) The International Classification of Headache Disorders. 3rd ed. Cephalalgia 2018;38:1–211. https://www.ichd-3.org/wp-content/uploads/2018/01/The-International-Classification-of-Headache-Disorders-3rd-Edition-2018.pdf
  104. Pagano HP, Zapata LB, Berry-Bibee EN, Nanda K, Curtis KM. Safety of hormonal contraception and intrauterine devices among women with depressive and bipolar disorders: a systematic review. 2016;94:641–9. https://doi.org/10.1016/j.contraception.2016.06.012 PMID:27364100
  105. Barrington JW, Arunkalaivanan AS, Abdel-Fattah M. Comparison between the levonorgestrel intrauterine system (LNG-IUS) and thermal balloon ablation in the treatment of menorrhagia. 2003;108:72–4. https://doi.org/10.1016/S0301-2115(02)00408-6 PMID:12694974
  106. Gupta B, Mittal S, Misra R, Deka D, Dadhwal V. Levonorgestrel-releasing intrauterine system vs. transcervical endometrial resection for dysfunctional uterine bleeding. 2006;95:261–6. https://doi.org/10.1016/j.ijgo.2006.07.004 PMID:16999960
  107. Hurskainen R, Teperi J, Rissanen P, et al. Quality of life and cost-effectiveness of levonorgestrel-releasing intrauterine system versus hysterectomy for treatment of menorrhagia: a randomised trial. [see comment]. 2001;357:273–7. https://doi.org/10.1016/S0140-6736(00)03615-1 PMID:11214131
  108. Istre O, Trolle B. Treatment of menorrhagia with the levonorgestrel intrauterine system versus endometrial resection. 2001;76:304–9. https://doi.org/10.1016/S0015-0282(01)01909-4 PMID:11476777
  109. Koh SC, Singh K. The effect of levonorgestrel-releasing intrauterine system use on menstrual blood loss and the hemostatic, fibrinolytic/inhibitor systems in women with menorrhagia. 2007;5:133–8. https://doi.org/10.1111/j.1538-7836.2006.02243.x PMID:17010149
  110. Lethaby AE, Cooke I, Rees M. Progesterone/progestogen releasing intrauterine systems versus either placebo or any other medication for heavy menstrual bleeding. 2000;(2):CD002126. PMID:10796865
  111. Magalhães J, Aldrighi JM, de Lima GR. Uterine volume and menstrual patterns in users of the levonorgestrel-releasing intrauterine system with idiopathic menorrhagia or menorrhagia due to leiomyomas. 2007;75:193–8. https://doi.org/10.1016/j.contraception.2006.11.004 PMID:17303488
  112. Stewart A, Cummins C, Gold L, Jordan R, Phillips W. The effectiveness of the levonorgestrel-releasing intrauterine system in menorrhagia: a systematic review. 2001;108:74–86. https://doi.org/10.1111/j.1471-0528.2001.00020.x PMID:11213008
  113. Fedele L, Bianchi S, Zanconato G, Portuese A, Raffaelli R. Use of a levonorgestrel-releasing intrauterine device in the treatment of rectovaginal endometriosis. 2001;75:485–8. https://doi.org/10.1016/S0015-0282(00)01759-3 PMID:11239528
  114. Lockhat FBE, Emembolu J, Konje JC. The effect of a levonorgestrel intrauterine system (LNG-IUS) on symptomatic endometriosis. 2002;77(Suppl 1):S24. https://doi.org/10.1016/S0015-0282(01)03086-2
  115. Petta CA, Ferriani RA, Abrao MS, et al. Randomized clinical trial of a levonorgestrel-releasing intrauterine system and a depot GnRH analogue for the treatment of chronic pelvic pain in women with endometriosis. 2005;20:1993–8. https://doi.org/10.1093/humrep/deh869 PMID:15790607
  116. Vercellini P, Aimi G, Panazza S, De Giorgi O, Pesole A, Crosignani PG. A levonorgestrel-releasing intrauterine system for the treatment of dysmenorrhea associated with endometriosis: a pilot study. 1999;72:505–8. https://doi.org/10.1016/S0015-0282(99)00291-5 PMID:10519624
  117. Vercellini P, Frontino G, De Giorgi O, Aimi G, Zaina B, Crosignani PG. Comparison of a levonorgestrel-releasing intrauterine device versus expectant management after conservative surgery for symptomatic endometriosis: a pilot study. 2003;80:305–9. https://doi.org/10.1016/S0015-0282(03)00608-3 PMID:12909492
  118. Gaffield ME, Kapp N, Curtis KM. Combined oral contraceptive and intrauterine device use among women with gestational trophoblastic disease. 2009;80:363–71. https://doi.org/10.1016/j.contraception.2009.03.022 PMID:19751859
  119. Whiteman MK, Zapata LB, Tepper NK, Marchbanks PA, Curtis KM. Use of contraceptive methods among women with endometrial hyperplasia: a systematic review. 2010;82:56–63. https://doi.org/10.1016/j.contraception.2010.02.005 PMID:20682143
  120. Zapata LB, Whiteman MK, Tepper NK, Jamieson DJ, Marchbanks PA, Curtis KM. Intrauterine device use among women with uterine fibroids: a systematic review. 2010;82:41–55. https://doi.org/10.1016/j.contraception.2010.02.011 PMID:20682142
  121. Tepper NK, Steenland MW, Gaffield ME, Marchbanks PA, Curtis KM. Retention of intrauterine devices in women who acquire pelvic inflammatory disease: a systematic review. 2013;87:655–60. https://doi.org/10.1016/j.contraception.2012.08.011 PMID:23040135
  122. Faúndes A, Telles E, Cristofoletti ML, Faúndes D, Castro S, Hardy E. The risk of inadvertent intrauterine device insertion in women carriers of endocervical Chlamydia trachomatis. 1998;58:105–9. https://doi.org/10.1016/S0010-7824(98)00064-X PMID:9773265
  123. Ferraz do Lago R, Simões JA, Bahamondes L, Camargo RP, Perrotti M, Monteiro I. Follow-up of users of intrauterine device with and without bacterial vaginosis and other cervicovaginal infections. 2003;68:105–9. https://doi.org/10.1016/S0010-7824(03)00109-4 PMID:12954522
  124. Morrison CS, Sekadde-Kigondu C, Miller WC, Weiner DH, Sinei SK. Use of sexually transmitted disease risk assessment algorithms for selection of intrauterine device candidates. 1999;59:97–106. https://doi.org/10.1016/S0010-7824(99)00006-2 PMID:10361624
  125. Pap-Akeson M, Solheim F, Thorbert G, Akerlund M. Genital tract infections associated with the intrauterine contraceptive device can be reduced by inserting the threads into the uterine cavity. 1992;99:676–9. https://doi.org/10.1111/j.1471-0528.1992.tb13854.x PMID:1390474
  126. Sinei SK, Schulz KFLP, Lamptey PR, et al. Preventing IUCD-related pelvic infection: the efficacy of prophylactic doxycycline at insertion. 1990;97:412–9. https://doi.org/10.1111/j.1471-0528.1990.tb01828.x PMID:2196934
  127. Skjeldestad FE, Halvorsen LE, Kahn H, Nordbø SA, Saake K. IUD users in Norway are at low risk for genital C. trachomatis infection. 1996;54:209–12. https://doi.org/10.1016/S0010-7824(96)00190-4 PMID:8922873
  128. Walsh TL, Bernstein GS, Grimes DA, Frezieres R, Bernstein L, Coulson AH; IUD Study Group. Effect of prophylactic antibiotics on morbidity associated with IUD insertion: results of a pilot randomized controlled trial. 1994;50:319–27. https://doi.org/10.1016/0010-7824(94)90019-1 PMID:7813220
  129. Jatlaoui TC, Simmons KB, Curtis KM. The safety of intrauterine contraception initiation among women with current asymptomatic cervical infections or at increased risk of sexually transmitted infections. 2016;94:701–12. https://doi.org/10.1016/j.contraception.2016.05.013 PMID:27263041
  130. Curtis KM, Hannaford PC, Rodriguez MI, Chipato T, Steyn PS, Kiarie JN. Hormonal contraception and HIV acquisition among women: an updated systematic review. 2020;46:8–16. https://doi.org/10.1136/bmjsrh-2019-200509 PMID:31919239
  131. Hannaford PC, Ti A, Chipato T, Curtis KM. Copper intrauterine device use and HIV acquisition in women: a systematic review. 2020;46:17–25. https://doi.org/10.1136/bmjsrh-2019-200512 PMID:31919240
  132. Tepper NK, Curtis KM, Cox S, Whiteman MK. Update to U.S. medical eligibility criteria for contraceptive use, 2016: updated recommendations for the use of contraception among women at high risk for HIV infection. 2020;69:405–10. https://doi.org/10.15585/mmwr.mm6914a3 PMID:32271729
  133. Tepper NK, Curtis KM, Nanda K, Jamieson DJ. Safety of intrauterine devices among women with HIV: a systematic review. 2016;94:713–24. https://doi.org/10.1016/j.contraception.2016.06.011 PMID:27343750
  134. Grigoryan OR, Grodnitskaya EE, Andreeva EN, Shestakova MV, Melnichenko GA, Dedov II. Contraception in perimenopausal women with diabetes mellitus. 2006;22:198–206. https://doi.org/10.1080/09513590600624317 PMID:16723306
  135. Rogovskaya S, Rivera R, Grimes DA, et al. Effect of a levonorgestrel intrauterine system on women with type 1 diabetes: a randomized trial. 2005;105:811–5. https://doi.org/10.1097/01.AOG.0000156301.11939.56 PMID:15802410
  136. Zapata LB, Paulen ME, Cansino C, Marchbanks PA, Curtis KM. Contraceptive use among women with inflammatory bowel disease: a systematic review. 2010;82:72–85. https://doi.org/10.1016/j.contraception.2010.02.012 PMID:20682145
  137. Bounds W, Guillebaud J. Observational series on women using the contraceptive Mirena concurrently with anti-epileptic and other enzyme-inducing drugs. 2002;28:78–80. https://doi.org/10.1783/147118902101195992 PMID:12396777
  138. Gaffield ME, Culwell KR, Lee CR. The use of hormonal contraception among women taking anticonvulsant therapy. 2011;83:16–29. https://doi.org/10.1016/j.contraception.2010.06.013 PMID:21134499
  139. Reimers A, Helde G, Brodtkorb E. Ethinyl estradiol, not progestogens, reduces lamotrigine serum concentrations. 2005;46:1414–7. https://doi.org/10.1111/j.1528-1167.2005.10105.x PMID:16146436

Appendix C: Classifications for Progestin-Only Contraceptives

Classifications for progestin-only contraceptives (POCs) include those for progestin-only implants (68 mg etonogestrel), progestin-only injectables (depot medroxyprogesterone acetate [DMPA], 150 mg intramuscular [DMPA-IM] or 104 mg subcutaneous [DMPA-SC]), and progestin-only pills (POPs) (containing norethindrone, norgestrel, or drospirenone [DRSP]) (Box C1) (Table C1). DMPA-SC can be administered by a health care provider or through self-administration. Recommendations in this report and U.S. Selected Practice Recommendations for Contraceptive Use, 2024 (1) for provider-administered DMPA (IM or SC) also apply to self-administered DMPA-SC. POCs do not protect against sexually transmitted infections (STIs), including HIV infection, and patients using POCs should be counseled that consistent and correct use of external (male) latex condoms reduces the risk for STIs, including HIV infection (2). Use of internal (female) condoms can provide protection from transmission of STIs, although data are limited (2). Patients also should be counseled that pre-exposure prophylaxis, when taken as prescribed, is highly effective for preventing HIV infection (3).

Return to your place in the textBOX C1. Categories for classifying progestin-only contraceptives

U.S. MEC 1 = A condition for which there is no restriction for the use of the contraceptive method

U.S. MEC 2 = A condition for which the advantages of using the method generally outweigh the theoretical or proven risks

U.S. MEC 3 = A condition for which the theoretical or proven risks usually outweigh the advantages of using the method

U.S. MEC 4 = A condition that represents an unacceptable health risk if the contraceptive method is used

Abbreviation: U.S. MEC = U.S. Medical Eligibility Criteria for Contraceptive Use.



TABLE C1. Classifications for progestin-only contraceptives, including implants, depot medroxyprogesterone acetate, and progestin-only pills
Condition Category Clarification/Evidence/Comment
Implant DMPA POP
Personal Characteristics and Reproductive History
Pregnancy NA NA NA Clarification: Use of POCs is not required. No known harm to the patient, the course of pregnancy, or the fetus occurs if POCs are inadvertently used during pregnancy. However, the relation between DMPA use during pregnancy and its effects on the fetus remains unclear.
Age Evidence: Most studies have found that women lose BMD during DMPA use but recover BMD after discontinuation (4). Limited evidence demonstrates a weak association with fracture. However, one large study suggests that women who choose DMPA might be at higher risk for fracture before initiation (5). It is unclear whether adult women with long durations of DMPA use can regain BMD to baseline levels before entering menopause and whether adolescents can reach peak bone mass after discontinuation of DMPA. The relation between these changes in BMD during the reproductive years and future fracture risk is unknown. Studies generally find no effect of POCs other than DMPA on BMD (452).
a. Menarche to <18 years 1 2 1
b. 18–45 years 1 1 1
c. >45 years 1 2 1
Parity
a. Nulliparous 1 1 1
b. Parous 1 1 1
Breastfeeding
a. <21 days postpartum 2 2 2 Clarification (breastfeeding): Breastfeeding provides important health benefits for breastfeeding parent and infant. The U.S. Dietary Guidelines for Americans and American Academy of Pediatrics recommend that infants be exclusively breastfed for about the first 6 months with continued breastfeeding while introducing appropriate complementary foods for 1 year or longer (53) or up to age 2 years or longer (54).
Evidence (breastfeeding): Two small, RCTs found no adverse impact on breastfeeding with initiation of etonogestrel implants within 48 hours postpartum. Other studies found that initiation of POPs, injectables, and implants at ≤6 weeks postpartum compared with nonhormonal use had no detrimental effect on breastfeeding outcomes or infant health, growth, and development in the first year postpartum. In general, these studies are of poor quality, lack standard definitions of breastfeeding or outcome measures, and have not included premature or ill infants (55,56).
Evidence: Limited evidence suggests that DMPA use might further elevate risk for VTE among postpartum women compared with non-use (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
Comment: Risk factors for breastfeeding difficulties include previous breastfeeding difficulties, certain medical conditions, certain perinatal complications, and preterm birth. For all breastfeeding persons, with or without breastfeeding difficulties, discussions about contraception should include information about risks, benefits, and alternatives.
b. 21 to <30 days postpartum Clarification (breastfeeding): Breastfeeding provides important health benefits for breastfeeding parent and infant. The U.S. Dietary Guidelines for Americans and American Academy of Pediatrics recommend that infants be exclusively breastfed for about the first 6 months with continued breastfeeding while introducing appropriate complementary foods for 1 year or longer (53) or up to age 2 years or longer (54).
Evidence (breastfeeding): Two small, RCTs found no adverse impact on breastfeeding with initiation of etonogestrel implants within 48 hours postpartum. Other studies found that initiation of POPs, injectables, and implants at ≤6 weeks postpartum compared with nonhormonal use had no detrimental effect on breastfeeding outcomes or infant health, growth, and development in the first year postpartum. In general, these studies are of poor quality, lack standard definitions of breastfeeding or outcome measures, and have not included premature or ill infants (55,56).
Evidence: Limited evidence suggests that DMPA use might further elevate risk for VTE among postpartum women compared with nonuse (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
Comment: Risk factors for breastfeeding difficulties include previous breastfeeding difficulties, certain medical conditions, certain perinatal complications, and preterm birth. For all breastfeeding persons, with or without difficulties, discussions about contraception should include information about risks, benefits, and alternatives.
  i. With other risk factors for VTE (e.g., age ≥35 years, previous VTE, thrombophilia, immobility, transfusion at delivery, peripartum cardiomyopathy, BMI ≥30 kg/m2, postpartum hemorrhage, postcesarean delivery, preeclampsia, or smoking) 2 2 2
  ii. Without other risk factors for VTE 2 2 2
c. 30–42 days postpartum Clarification (breastfeeding): Breastfeeding provides important health benefits for breastfeeding parent and infant. The U.S. Dietary Guidelines for Americans and American Academy of Pediatrics recommend that infants be exclusively breastfed for about the first 6 months with continued breastfeeding while introducing appropriate complementary foods for 1 year or longer (53) or up to age 2 years or longer (54).
Evidence (breastfeeding): Two small, RCTs found no adverse impact on breastfeeding with initiation of etonogestrel implants within 48 hours postpartum. Other studies found that initiation of POPs, injectables, and implants at ≤6 weeks postpartum compared with nonhormonal use had no detrimental effect on breastfeeding outcomes or infant health, growth, and development in the first year postpartum. In general, these studies are of poor quality, lack standard definitions of breastfeeding or outcome measures, and have not included premature or ill infants (55,56).
Evidence: Limited evidence suggests that DMPA use might further elevate risk for VTE among postpartum women compared with nonuse (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
Comment: Risk factors for breastfeeding difficulties include previous breastfeeding difficulties, certain medical conditions, certain perinatal complications, and preterm birth. For all breastfeeding persons, with or without breastfeeding difficulties, discussions about contraception should include information about risks, benefits, and alternatives.
  i. With other risk factors for VTE (e.g., age ≥35 years, previous VTE, thrombophilia, immobility, transfusion at delivery, peripartum cardiomyopathy, BMI ≥30 kg/m2, postpartum hemorrhage, postcesarean delivery, preeclampsia, or smoking) 1 2 1
  ii. Without other risk factors for VTE 1 1 1
d. >42 days postpartum 1 1 1 Clarification (breastfeeding): Breastfeeding provides important health benefits for breastfeeding parent and infant. The U.S. Dietary Guidelines for Americans and American Academy of Pediatrics recommend that infants be exclusively breastfed for about the first 6 months with continued breastfeeding while introducing appropriate complementary foods for 1 year or longer (53) or up to age 2 years or longer (54).
Evidence: Overall, studies found that initiation of POPs, injectables, and implants at >6 weeks postpartum compared with nonhormonal use had no detrimental effect on breastfeeding outcomes or infant health, growth, and development in the first year postpartum. In general, these studies are of poor quality, lack standard definitions of breastfeeding or outcome measures, and have not included premature or ill infants (56).
Comment: Risk factors for breastfeeding difficulties include previous breastfeeding difficulties, certain medical conditions, certain perinatal complications, and preterm birth. For all breastfeeding persons, with or without breastfeeding difficulties, discussions about contraception should include information about risks, benefits, and alternatives.
Postpartum
(nonbreastfeeding)
a. <21 days postpartum 1 2 1 Evidence: Limited evidence suggests that DMPA use might further elevate risk for VTE among postpartum women compared with nonuse (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
b. 21–42 days postpartum
  i. With other risk factors for VTE (e.g., age ≥35 years, previous VTE, thrombophilia, immobility, transfusion at delivery, peripartum cardiomyopathy, BMI ≥30 kg/m2, postpartum hemorrhage, postcesarean delivery, preeclampsia, or smoking) 1 2 1 Evidence: Limited evidence suggests that DMPA use might further elevate risk for VTE among postpartum women compared with nonuse (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
  ii. Without other risk factors for VTE 1 1 1
c. >42 days postpartum 1 1 1
Postabortion
(spontaneous or
induced)
a. First trimester abortion Clarification: POCs may be started immediately after abortion completion or at time of medication abortion initiation.
Clarification (DMPA): After a first trimester medication abortion that did not include mifepristone, there is no restriction for the use of DMPA (category 1). After a first trimester medication abortion that included mifepristone, there is no restriction for use of DMPA after abortion completion (category 1) and benefits generally outweigh risks with DMPA use immediately at time of medication abortion initiation (category 2). Concurrent administration of DMPA with mifepristone might slightly decrease medication abortion effectiveness and increase risk for ongoing pregnancy. Risk for ongoing pregnancy with concurrent administration of DMPA with mifepristone should be considered along with personal preference and access to follow-up abortion and contraceptive care.
Evidence: Limited evidence suggests decreased first trimester medication abortion effectiveness with concurrent administration of DMPA with mifepristone (immediate) versus DMPA administration after abortion completion (delayed). In one study, the risk for ongoing pregnancy, while overall low, was higher with immediate (3.6%) versus delayed (0.9%) DMPA administration (difference 2.7%; 90% CI = 0.4–5.6%) (57). This difference was not seen with other progestin-only methods (58). Evidence suggests that there is no increased risk for adverse events when POCs are initiated after first trimester procedural or medication abortion (immediately or delayed) (58) (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
  i. Procedural (surgical) 1 1 1
  ii. Medication 1 1/2 1
  iii. Spontaneous abortion with no intervention 1 1 1
b. Second trimester abortion Clarification: POCs may be started immediately after abortion completion or at time of medication abortion initiation.
  i. Procedural (surgical) 1 1 1
  ii. Medication 1 1 1
  iii. Spontaneous abortion with no intervention 1 1 1
c. Immediate postseptic abortion 1 1 1 Clarification: POCs may be started immediately after abortion completion or at time of medication abortion initiation.
Past ectopic pregnancy 1 1 2 Comment: POP users have a higher absolute rate of ectopic pregnancy than do users of other POCs but still lower than those using no method.
History of pelvic surgery 1 1 1
Smoking
a. Age <35 years 1 1 1
b. Age ≥35 years
  i. <15 cigarettes per day 1 1 1
  ii. ≥15 cigarettes per day 1 1 1
Obesity
a. BMI ≥30 kg/m2 1 1 1
b. Menarche to <18 years and BMI ≥30 kg/m2 1 2 1 Evidence: Among adult women, generally no association has been found between baseline weight and weight gain among DMPA users compared with nonusers. Evidence is mixed for adolescent DMPA users, with certain studies observing greater weight gain among users with obesity compared with those without obesity but other studies demonstrating no association; methodologic differences across studies might account for the differences in findings. Data on other POC methods and other adverse outcomes including weight gain are limited (5976).
History of bariatric
surgery

This condition is associated
with increased risk for
adverse health events as a
result of pregnancy (Box 3).
a. Restrictive procedures: decrease storage capacity of the stomach (vertical banded gastroplasty, laparoscopic adjustable gastric band, or laparoscopic sleeve gastrectomy) 1 1 1 Evidence: Limited evidence demonstrated no substantial decrease in effectiveness of oral contraceptives among women who underwent laparoscopic placement of an adjustable gastric band (77).
b. Malabsorptive procedures: decrease absorption of nutrients and calories by shortening the functional length of the small intestine (Roux-en-Y gastric bypass or biliopancreatic diversion) 1 1 3 Evidence: Limited evidence demonstrated no substantial decrease in effectiveness of oral contraceptives among women who underwent a biliopancreatic diversion; however, evidence from pharmacokinetic studies suggested conflicting results regarding oral contraceptive effectiveness among women who underwent a jejunoileal bypass (77).
Comment: Bariatric surgical procedures involving a malabsorptive component have the potential to decrease oral contraceptive effectiveness, perhaps further decreased by postoperative complications such as long-term diarrhea, vomiting, or both.
Surgery
a. Minor surgery without immobilization 1 1 1
b. Major surgery
  i. Without prolonged immobilization 1 1 1
  ii. With prolonged immobilization 1 2 1 Evidence: No direct evidence was identified on risk for thrombosis with POC use among those undergoing major surgery. Use of DMPA, which has been associated with a higher risk for venous thrombosis compared with nonuse (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516), might further elevate risk for thrombosis among persons with prolonged immobilization after major surgery.
Cardiovascular Disease
Multiple risk factors for atherosclerotic cardiovascular disease (e.g., older age, smoking, diabetes, hypertension, low HDL, high LDL, or high triglyceride levels) 2 3 2 Clarification: When multiple major risk factors exist, risk for cardiovascular disease might increase substantially. Certain POCs might increase the risk for thrombosis, although this increase is substantially less than with COCs. The effects of DMPA might persist for some time after discontinuation.
Clarification: The recommendations apply to known pre-existing medical conditions or characteristics. Few if any screening tests are needed before initiation of contraception. See U.S. SPR (https://www.cdc.gov/contraception/hcp/usspr) (1).
Hypertension
Systolic blood pressure
≥160 mm Hg or diastolic
blood pressure ≥100 mm
Hg are associated with
increased risk for adverse
health events as a
result of pregnancy (Box 3).
a. Adequately controlled hypertension 1 2 1 Clarification: For all categories of hypertension, classifications are based on the assumption that no other risk factors exist for cardiovascular disease. When multiple risk factors do exist, risk for cardiovascular disease might increase substantially. A single reading of blood pressure level is not sufficient to classify a person as hypertensive.
Clarification: Persons adequately treated for hypertension are at lower risk for acute myocardial infarction and stroke than are untreated persons. Although no data exist, POC users with adequately controlled and monitored hypertension should be at lower risk for acute myocardial infarction and stroke than are untreated hypertensive POC users.
b. Elevated blood pressure
levels (properly taken
measurements)
Clarification: For all categories of hypertension, classifications are based on the assumption that no other risk factors exist for cardiovascular disease. When multiple risk factors do exist, risk for cardiovascular disease might increase substantially. A single reading of blood pressure level is not sufficient to classify a person as hypertensive.
Evidence: Limited evidence suggests that among women with hypertension, those who used POPs or progestin-only injectables had a small increased risk for cardiovascular events compared with women who did not use these methods (78).
  i. Systolic 140–159 mm Hg or diastolic 90–99 mm Hg 1 2 1
  ii. Systolic ≥160 mm Hg or diastolic ≥100 mm Hg 2 3 2
c. Vascular disease 2 3 2 Clarification: For all categories of hypertension, classifications are based on the assumption that no other risk factors exist for cardiovascular disease. When multiple risk factors do exist, risk for cardiovascular disease might increase substantially. A single reading of blood pressure level is not sufficient to classify a person as hypertensive.
Comment: Concern exists about hypoestrogenic effects and reduced HDL levels, particularly among users of DMPA. However, little concern exists about these effects with regard to POPs. The effects of DMPA might persist for some time after discontinuation.
History of high blood pressure during pregnancy (when current blood pressure is measurable and normal) 1 1 1
Deep venous thrombosis/
Pulmonary embolism

This condition is associated
with increased risk for
adverse health events as a
result of pregnancy (Box 3).
a. Current or history of DVT/PE, receiving anticoagulant therapy (therapeutic dose) (e.g., acute DVT/PE or long-term therapeutic dose) 2 2 2 Clarification: Persons using anticoagulant therapy are at risk for gynecologic complications of therapy, such as heavy or prolonged bleeding and hemorrhagic ovarian cysts. POCs can be of benefit in preventing or treating these complications; benefits might vary by POC dose and formulation. When a contraceptive method is used as a therapy, rather than solely to prevent pregnancy, the risk/benefit ratio might differ and should be considered on a case-by-case basis.
Evidence: Limited evidence was identified on use of POCs among women with acute DVT/PE receiving anticoagulant therapy (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516 In one study among women with a history of acute VTE currently receiving therapeutic anticoagulant therapy (i.e., rivaroxaban or enoxaparin/vitamin K antagonist [warfarin or acenocoumarol]), the incidence of recurrent VTE was similar among estrogen users (CHC or estrogen-only pills), POC users, and women not on hormonal therapy (79).
Limited evidence suggests that intramuscular injections of DMPA in women receiving chronic anticoagulation therapy do not pose a significant risk for hematoma at the injection site or increase the risk for heavy or irregular vaginal bleeding (80).
b. History of DVT/PE,
receiving anticoagulant
therapy (prophylactic dose)
Clarification: Persons using anticoagulant therapy are at risk for gynecologic complications of therapy, such as heavy or prolonged bleeding and hemorrhagic ovarian cysts. POCs can be of benefit in preventing or treating these complications; benefits might vary by POC dose and formulation. When a contraceptive method is used as a therapy, rather than solely to prevent pregnancy, the risk/benefit ratio might differ and should be considered on a case-by-case basis.
Evidence: Limited evidence was identified on use of POCs among women with acute DVT/PE receiving anticoagulant therapy (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516). Use of DMPA, which has been associated with a higher risk for venous thrombosis compared with nonuse (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516), might further elevate risk for thrombosis among persons with a history of DVT/PE and at higher risk for recurrent DVT/PE.
Limited evidence suggests that intramuscular injections of DMPA in women receiving chronic anticoagulation therapy do not pose a significant risk for hematoma at the injection site or increase the risk for heavy or irregular vaginal bleeding (80).
  i. Higher risk for recurrent DVT/PE (one or more risk factors) 2 3 2
Thrombophilia (e.g., factor V Leiden mutation; prothrombin gene mutation; protein S, protein C, and antithrombin deficiencies; or antiphospholipid syndrome)
• Active cancer (metastatic, receiving therapy, or within 6 months after clinical remission), excluding nonmelanoma skin cancer
• History of recurrent DVT/PE
  ii. Lower risk for recurrent DVT/PE (no risk factors) 2 2 2
c. History of DVT/PE, not
receiving anticoagulant
therapy
  i. Higher risk for recurrent DVT/PE (one or more risk factors) 2 3 2 Evidence: Use of DMPA, which has been associated with a higher risk for venous thrombosis compared with nonuse (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516), might further elevate risk for thrombosis among persons with a history of DVT/PE and at higher risk for recurrent DVT/PE.
• History of estrogen-associated DVT/PE
• Pregnancy-associated DVT/PE
• Idiopathic DVT/PE
• Thrombophilia (e.g., factor V Leiden mutation; prothrombin gene mutation; protein S, protein C, and antithrombin deficiencies; or antiphospholipid syndrome)
• Active cancer (metastatic,
receiving therapy, or within 6 months after clinical remission), excluding nonmelanoma skin
cancer
• History of recurrent DVT/PE
  ii. Lower risk for recurrent DVT/PE (no risk factors) 2 2 2
d. Family history
(first-degree relatives)
1 1 1
Thrombophilia (e.g., factor V Leiden mutation; prothrombin gene mutation; protein S, protein C, and antithrombin deficiencies; or antiphospholipid syndrome)
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
2 3 2 Clarification: Routine screening in the general population before contraceptive initiation is not recommended.
Clarification: If a person has current or history of DVT/PE, see recommendations for DVT/PE.
Clarification: Classification of antiphospholipid syndrome includes presence of a clinical feature (e.g., thrombosis or obstetric morbidity) and persistently abnormal antiphospholipid antibody test on two or more occasions at least 12 weeks apart (81).
Evidence: Among women with factor V Leiden mutation, one study found that women using POCs had an increased risk for venous thrombosis compared with non-users without the mutation, with the highest relative risk for DMPA users (82). Women with prothrombin gene mutation using POCs did not have an increased risk for venous thrombosis compared with nonusers without the mutation (82). No evidence was identified on POC use among persons with protein S deficiency, protein C deficiency, antithrombin deficiency, or antiphospholipid syndrome (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
Superficial venous
disorders
a. Varicose veins 1 1 1
b. Superficial venous thrombosis (acute or history) 1 2 1 Evidence: No direct evidence was identified on risk for thrombosis with POC use among persons with superficial venous thrombosis (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516). Persons with superficial venous thrombosis are at higher risk for venous thrombosis than the general population (83). Use of DMPA, which has been associated with a higher risk for venous thrombosis compared with non-use (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516), might further elevate risk for thrombosis among persons with acute or history of superficial venous thrombosis.
Current and history of ischemic heart disease
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
Initiation Continuation Initiation Continuation Comment: Concern exists about hypoestrogenic effects and reduced HDL levels, particularly among users of DMPA. However, little concern exists about these effects with regard to POPs. The effects of DMPA might persist for some time after discontinuation.
2 3 3 2 3
Stroke (history of cerebrovascular accident)
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
Initiation Continuation Initiation Continuation Comment: Concern exists about hypoestrogenic effects and reduced HDL levels, particularly among users of DMPA. However, little concern exists about these effects with regard to POPs. The effects of DMPA might persist for some time after discontinuation.
2 3 3 2 3
Valvular heart disease
Complicated valvular heart
disease is associated with
increased risk for adverse
health events as a result of
pregnancy (Box 3).
a. Uncomplicated 1 1 1
b. Complicated (pulmonary hypertension, risk for atrial fibrillation, or history of subacute bacterial endocarditis) 1 2 1 Evidence: No direct evidence was identified on risk for thrombosis with POC use among persons with valvular heart disease (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516). Use of DMPA, which has been associated with a higher risk for venous thrombosis compared with nonuse (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516), might further elevate risk for thrombosis among persons with complicated valvular heart disease.
Peripartum
cardiomyopathy

This condition is associated
with increased risk for
adverse health events as a
result of pregnancy (Box 3).
Evidence: No direct evidence was identified on the safety of POC use among persons with peripartum cardiomyopathy (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516). Limited indirect evidence from noncomparative studies of women with cardiac disease demonstrated few cases of hypertension, thromboembolism, and heart failure in women with cardiac disease using POPs and DMPA (84). Use of DMPA, which has been associated with a higher risk for venous thrombosis compared with nonuse (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516), might further elevate risk for thrombosis among persons with peripartum cardiomyopathy.
Comment: Progestin-only implants might induce cardiac arrhythmias in healthy persons; persons with peripartum cardiomyopathy have a high incidence of cardiac arrhythmias.
a. Normal or mildly
impaired cardiac function
(New York Heart
Association Functional
Class I or II: no limitation
of activities or slight, mild
limitation of activity) (85)
  i. <6 months 1 2 1
  ii. ≥6 months 1 2 1
b. Moderately or severely impaired cardiac function (New York Heart Association Functional Class III or IV: marked limitation of activity or should be at complete rest) (85) 2 3 2
Renal Disease
Chronic kidney disease
This condition is associated
with increased risk for
adverse health events as a
result of pregnancy (Box 3).
a. Current nephrotic syndrome 2 3 2
DRSP POP with known hyperkalemia: 4
Clarification (DRSP POP): Persons with known hyperkalemia should not use DRSP POPs because of the risk for worsening hyperkalemia (category 4). For persons with CKD without known hyperkalemia (category 2), consider checking serum potassium level during first cycle of DRSP POPs.
Evidence: No direct evidence was identified on POC use among persons with CKD with current nephrotic syndrome (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516). Persons with severe CKD or nephrotic syndrome are at higher risk for thrombosis than the general population (8690). Use of DMPA, which has been associated with increased risk for thrombosis compared with nonuse (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516), might further elevate risk for thrombosis among those with CKD with current nephrotic syndrome. Persons with severe CKD have a higher prevalence of fracture than the general population (9193). Use of DMPA, which has been associated with small changes in bone mineral density (4) might further elevate risk for fracture among persons with CKD with current nephrotic syndrome.
Comment: A person might have CKD without current nephrotic syndrome, but might have other conditions often associated with CKD (e.g., diabetes, hypertension, SLE). See recommendations for other conditions if they apply.
b. Hemodialysis 2 3 2
DRSP POP with known hyperkalemia: 4
Clarification (DRSP POP): Persons with known hyperkalemia should not use DRSP POPs because of the risk for worsening hyperkalemia (category 4). For persons with CKD without known hyperkalemia (category 2), consider checking serum potassium level during first cycle of DRSP POPs.
Evidence: No direct evidence was identified on POC use among persons with CKD on hemodialysis (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516). Persons with CKD on dialysis are at higher risk for thrombosis than the general population (94-96). Use of DMPA, which has been associated with increased risk for thrombosis compared with nonuse (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516), might further elevate risk for thrombosis among those with CKD on dialysis. Persons with CKD on dialysis have a higher prevalence of fracture than the general population (9799). Use of DMPA, which has been associated with small changes in bone mineral density (4), might further elevate risk for fracture among persons with CKD on dialysis.
Comment: A person might have CKD without hemodialysis, but might have other conditions often associated with CKD (e.g., diabetes, hypertension, SLE). See recommendations for other conditions if they apply.
c. Peritoneal dialysis 2 3 2
DRSP POP with known hyperkalemia: 4
Clarification (DRSP POP): Persons with known hyperkalemia should not use DRSP POPs because of the risk for worsening hyperkalemia (category 4). For persons with CKD without known hyperkalemia (category 2), consider checking serum potassium level during first cycle of DRSP POPs.
Evidence: No direct evidence was identified on POC use among persons with CKD on peritoneal dialysis (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516). Persons with CKD on dialysis are at higher risk for thrombosis than the general population (9496). Use of DMPA, which has been associated with increased risk for thrombosis compared with nonuse (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516), might further elevate risk for thrombosis among those with CKD on dialysis. Persons with CKD on dialysis have a higher prevalence of fracture than the general population (9799). Use of DMPA, which has been associated with small changes in bone mineral density (4), might further elevate risk for fracture among persons with CKD on dialysis.
Comment: A person might have CKD without peritoneal dialysis but might have other conditions often associated with CKD (e.g., diabetes, hypertension, and SLE). See recommendations for other conditions if they apply.
Rheumatic Diseases
Systemic lupus
erythematosus

This condition is
associated with increased
risk for adverse health events
as a result of
pregnancy (Box 3).
Initiation Continuation
a. Positive (or unknown) antiphospholipid antibodies 2 3 3 2 Clarification: Persons with SLE are at increased risk for ischemic heart disease, stroke, and VTE. Categories assigned to such conditions in U.S. MEC should be the same for persons with SLE who have these conditions. For all subconditions of SLE, classifications are based on the assumption that no other risk factors for cardiovascular disease are present; these classifications must be modified in the presence of such risk factors (100118).
Evidence: No direct evidence was identified on POC use among persons with SLE with antiphospholipid antibodies (119) (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516). Persons with SLE with antiphospholipid antibodies are at higher risk for thrombosis than the general population (120,121). Use of DMPA, which has been associated with a higher risk for venous thrombosis compared with nonuse (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516), might further elevate risk for thrombosis among persons with SLE with antiphospholipid antibodies.
b. Severe thrombocytopenia 2 3 2 2 Clarification: Persons with SLE are at increased risk for ischemic heart disease, stroke, and VTE. Categories assigned to such conditions in U.S. MEC should be the same for persons with SLE who have these conditions. For all subconditions of SLE, classifications are based on the assumption that no other risk factors for cardiovascular disease are present; these classifications must be modified in the presence of such risk factors (100118).
Comment: Severe thrombocytopenia increases the risk for bleeding. POCs might be useful in treating heavy or prolonged bleeding in persons with severe thrombocytopenia. However, given the increased or erratic bleeding that might be seen on initiation of DMPA and its irreversibility for 11–13 weeks after administration, initiation of this method in persons with severe thrombocytopenia should be done with caution.
c. Immunosuppressive therapy 2 2 2 2 Clarification: Persons with SLE are at increased risk for ischemic heart disease, stroke, and VTE. Categories assigned to such conditions in U.S. MEC should be the same for persons with SLE who have these conditions. For all subconditions of SLE, classifications are based on the assumption that no other risk factors for cardiovascular disease are present; these classifications must be modified in the presence of such risk factors (100118).
d. None of the above 2 2 2 2 Clarification: Persons with SLE are at increased risk for ischemic heart disease, stroke, and VTE. Categories assigned to such conditions in U.S. MEC should be the same for persons with SLE who have these conditions. For all subconditions of SLE, classifications are based on the assumption that no other risk factors for cardiovascular disease are present; these classifications must be modified in the presence of such risk factors (100118).
Rheumatoid arthritis
a. Not receiving immunosuppressive therapy 1 2 1 Evidence: Limited evidence demonstrates no consistent pattern of improvement or worsening of rheumatoid arthritis with use of oral contraceptives, progesterone, or estrogen (122).
b. Receiving immunosuppressive therapy 1 2/3 1 Clarification (DMPA): DMPA use among persons receiving long-term corticosteroid therapy with a history of, or with risk factors for, nontraumatic fractures is classified as category 3. Otherwise, DMPA use for persons with rheumatoid arthritis is classified as category 2.
Evidence: Limited evidence demonstrates no consistent pattern of improvement or worsening of rheumatoid arthritis with use of oral contraceptives, progesterone, or estrogen (122).
Neurologic Conditions
Headaches
a. Nonmigraine (mild or severe) 1 1 1
b. Migraine Evidence: No studies directly examined the risk for stroke among women with migraine using POCs (123). Limited evidence demonstrated that women using POPs, DMPA, or implants do not have an increased risk for ischemic stroke compared with nonusers (124).
Comment: Menstrual migraine is a subtype of migraine without aura. For more information, see the International Headache Society’s International Classification of Headache Disorders, 3rd ed. (https://ichd-3.org) (125).
  i. Without aura (includes menstrual migraine) 1 1 1
  ii. With aura 1 1 1
Epilepsy
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 1 1 Clarification: If a person is taking anticonvulsants, see recommendations for Drug Interactions. Certain anticonvulsants lower POC effectiveness.
Multiple sclerosis Evidence: Limited evidence demonstrates that use of COCs or oral contraceptives (type not specified) among women with multiple sclerosis does not worsen the clinical course of disease (126).
Comment: Persons with multiple sclerosis might have compromised bone health from disease-related disability, immobility, and use of corticosteroids. Use of DMPA, which has been associated with small changes in BMD, might be of concern.
a. Without prolonged immobility 1 2 1
b. With prolonged immobility 1 2 1
Depressive Disorders
Depressive disorders 1 1 1 Clarification: If a person is taking psychotropic medications or St. John’s wort, see recommendations for Drug Interactions.
Evidence: The frequency of psychiatric hospitalizations for women with bipolar disorder or depression did not significantly differ among women using DMPA, LNG-IUD, Cu-IUD, or sterilization (127).
Reproductive Tract Infections and Disorders
Vaginal bleeding patterns
a. Irregular pattern without heavy bleeding 2 2 2 Comment: Irregular menstrual bleeding patterns are common among healthy persons. POC use frequently induces an irregular bleeding pattern. Implant use might induce irregular bleeding patterns, especially during the first 3–6 months, although these patterns might persist longer.
b. Heavy or prolonged bleeding (includes regular and irregular patterns) 2 2 2 Clarification: Unusually heavy bleeding should raise the suspicion of a serious underlying condition.
Unexplained vaginal bleeding (suspicious for serious condition) before evaluation 3 3 2 Clarification: If pregnancy or an underlying pathological condition (e.g., pelvic malignancy) is suspected, it must be evaluated and the category adjusted after evaluation.
Comment: POCs might cause irregular bleeding patterns, which might mask symptoms of underlying pathologic conditions. The effects of DMPA might persist for some time after discontinuation.
Endometriosis 1 1 1
Benign ovarian tumors (including cysts) 1 1 1
Severe dysmenorrhea 1 1 1
Gestational trophoblastic
disease

This condition is
associated with increased
risk for adverse health
events as a result of
pregnancy (Box 3).
Clarification: For all subconditions of gestational trophoblastic disease, classifications are based on the assumption that persons are under close medical supervision because of the need for monitoring of β-hCG levels for appropriate disease surveillance.
a. Suspected gestational
trophoblastic disease
(immediate
postevacuation)
  i. Uterine size first trimester 1 1 1
  ii. Uterine size second trimester 1 1 1
b. Confirmed gestational
trophoblastic disease
(after initial evacuation
and during monitoring)
  i. Undetectable or nonpregnant β–hCG levels 1 1 1
  ii. Decreasing β–hCG levels 1 1 1
  iii. Persistently elevated β-hCG levels or malignant disease, with no evidence or suspicion of intrauterine disease 1 1 1
  iv. Persistently elevated β-hCG levels or malignant disease, with evidence or suspicion of intrauterine disease 1 1 1
Cervical ectropion 1 1 1
Cervical intraepithelial neoplasia 2 2 1 Evidence: Among women with persistent human papillomavirus infection, long-term DMPA use (≥5 years) might increase the risk for carcinoma in situ and invasive carcinoma (128).
Cervical cancer (awaiting treatment) 2 2 1 Comment: Theoretical concern exists that POC use might affect prognosis of the existing disease. While awaiting treatment, POCs may be used. In general, treatment of this condition can render a person infertile.
Breast disease
Breast cancer is associated
with increased risk for
adverse health events as a
result of pregnancy
(Box 3).
a. Undiagnosed mass 2 2 2 Clarification: Evaluation of mass should be pursued as early as possible.
b. Benign breast disease 1 1 1
c. Family history of cancer 1 1 1
d. Breast cancer
  i. Current 4 4 4 Comment: Breast cancer is a hormonally sensitive tumor, and the prognosis for persons with current or recent breast cancer might worsen with POC use.
  ii. Past and no evidence of current disease for 5 years 3 3 3
Endometrial hyperplasia 1 1 1
Endometrial cancer
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 1 1 Comment: While awaiting treatment, POCs may be used. In general, treatment of this condition renders a person infertile.
Ovarian cancer
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 1 1 Comment: While awaiting treatment, POCs may be used. In general, treatment of this condition renders a person infertile.
Uterine fibroids 1 1 1 Comment: POCs do not appear to cause growth of uterine fibroids.
Pelvic inflammatory disease Comment: Whether POCs, like COCs, reduce the risk for PID among persons with STIs is unknown; however, they do not protect against HIV infection or lower genital tract STIs.
a. Current PID 1 1 1
b. Past PID
  i. With subsequent pregnancy 1 1 1
  ii. Without subsequent pregnancy 1 1 1
Sexually transmitted
infections
a. Current purulent cervicitis or chlamydial infection or gonococcal infection 1 1 1
b. Vaginitis (including Trichomonas vaginalis and bacterial vaginosis) 1 1 1
c. Other factors related to STIs 1 1 1
HIV
High risk for HIV infection 1 1 1 Evidence: High-quality evidence from one RCT observed no statistically significant differences in HIV acquisition between DMPA-IM versus Cu-IUD, DMPA-IM versus LNG implant, and Cu-IUD versus LNG implant. Of the low-to-moderate-quality evidence from 14 observational studies, certain studies suggested a possible increased risk for HIV infection with progestin-only injectable use, which was most likely due to unmeasured confounding. Low-quality evidence from three observational studies did not suggest an increased HIV infection risk for implant users. No studies of sufficient quality were identified for POPs (129131).
HIV infection
For persons with HIV infection who are not clinically well or not receiving ARV therapy, this condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 1 1 Clarification: Drug interactions might exist between hormonal contraceptives and ARV drugs (see recommendations for Drug Interactions).
Evidence: Overall, evidence does not support an association between POC use and progression of HIV infection. Limited direct evidence on an association between POC use and transmission of HIV to noninfected partners, as well as studies measuring genital viral shedding as a proxy for infectivity, have had mixed results. Studies measuring whether hormonal contraceptive methods affect plasma HIV viral load generally have found no effect (132134).
Other Infections
Schistosomiasis
Schistosomiasis with
fibrosis of the liver is
associated with increased
risk for adverse health
events as a result of
pregnancy (Box 3).
a. Uncomplicated 1 1 1 Evidence: Among women with uncomplicated schistosomiasis, limited evidence demonstrated that DMPA use had no adverse effects on liver function (135).
b. Fibrosis of the liver (if severe, see recommendations for Cirrhosis) 1 1 1
Tuberculosis
This condition is
associated with increased
risk for adverse health
events as a result of
pregnancy (Box 3).
Clarification: If a person is taking rifampin, see recommendations for Drug Interactions. Rifampin is likely to decrease the effectiveness of certain POCs.
a. Nonpelvic 1 1 1
b. Pelvic 1 1 1
Malaria 1 1 1
Endocrine Conditions
Diabetes
Insulin-dependent
diabetes; diabetes with
nephropathy, retinopathy
or neuropathy; diabetes
with other vascular
disease; or diabetes of
>20 years’ duration are
associated with increased
risk for adverse health
events as a result of
pregnancy (Box 3).
a. History of gestational disease 1 1 1 Evidence: POCs had no adverse effects on serum lipid levels in women with a history of gestational diabetes in two small studies (136,137). Limited evidence is inconsistent about the development of noninsulin-dependent diabetes among users of POCs with a history of gestational diabetes (138141).
b. Nonvascular disease Evidence: Among women with insulin-dependent or noninsulin-dependent diabetes, limited evidence on use of POCs (POPs, DMPA, and LNG implant) suggests that these methods have little effect on short-term or long-term diabetes control (e.g., glycosylated hemoglobin levels), hemostatic markers, or lipid profile (142145).
  i. Non-insulin dependent 2 2 2
  ii. Insulin dependent 2 2 2
c. Nephropathy, retinopathy or neuropathy 2 3 2 Comment: Concern exists about hypoestrogenic effects and reduced HDL levels, particularly among users of DMPA. The effects of DMPA might persist for some time after discontinuation. Certain POCs might increase the risk for thrombosis, although this increase is substantially less than with COCs.
d. Other vascular disease or diabetes of >20 years’ duration 2 3 2 Comment: Concern exists about hypoestrogenic effects and reduced HDL levels, particularly among users of DMPA. The effects of DMPA might persist for some time after discontinuation. Certain POCs might increase the risk for thrombosis, although this increase is substantially less than with COCs.
Thyroid disorders
a. Simple goiter 1 1 1
b. Hyperthyroid 1 1 1
c. Hypothyroid 1 1 1
Gastrointestinal Conditions
Inflammatory bowel disease (ulcerative colitis or Crohn’s disease) 1 2 2 Evidence: Risk for disease relapse among women with IBD using oral contraceptives (most studies did not specify formulation) did not increase significantly from that for nonusers (146).
Comment: Absorption of POPs among persons with IBD might be reduced if the person has substantial malabsorption caused by severe disease or small bowel surgery.
Women with IBD have a higher prevalence of osteoporosis and osteopenia than the general population. Use of DMPA, which has been associated with small changes in BMD, might be of concern.
Gallbladder disease
a. Asymptomatic 2 2 2
b. Symptomatic
  i. Current 2 2 2
  ii. Treated by cholecystectomy 2 2 2
  iii. Medically treated 2 2 2
History of cholestasis
a. Pregnancy related 1 1 1
b. Past COC related 2 2 2 Comment: Theoretical concern exists that a history of COC-related cholestasis might predict subsequent cholestasis with POC use. However, this has not been documented.
Viral hepatitis
a. Acute or flare 1 1 1 Evidence: No direct evidence was identified on POC use among persons with viral hepatitis (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
b. Chronic 1 1 1 Evidence: No evidence was identified on POC use among persons with viral hepatitis (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
Cirrhosis
Decompensated cirrhosis is
associated with increased risk
for adverse health events as a
result of pregnancy (Box 3).
a. Compensated (normal liver function) 1 1 1 Evidence: No direct evidence was identified on POC use among persons with cirrhosis (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
b. Decompensated (impaired liver function) 2 3 2 Evidence: No direct evidence was identified on POC use among persons with cirrhosis. (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516). DMPA use has been associated with a higher risk for venous thrombosis compared with nonuse (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
Comment: Hepatic metabolism of exogenous hormones might be impaired in persons with liver dysfunction, which could lead to increased progestin levels in circulation and progestin-related side effects and adverse events (e.g., thrombosis), which might vary by dose and formulation. Any progestin-related hepatotoxicity might be less tolerated in persons with existing liver dysfunction.
Liver tumors
Hepatocellular adenoma
and malignant liver tumors
are associated with increased
risk for adverse health
events as a result
of pregnancy (Box 3).
a. Benign
  i. Focal nodular hyperplasia 2 2 2 Evidence: Limited evidence suggests that progestin use does not influence either progression or regression of focal nodular hyperplasia (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
  ii. Hepatocellular adenoma 2 3 2 Evidence: Limited evidence suggests that hepatocellular adenomas generally regress or remain stable during progestin use (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
b. Malignant (hepatocellular carcinoma) 3 3 3 Evidence: No direct evidence was identified on POC use among persons with hepatocellular carcinoma (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
Respiratory Conditions
Cystic fibrosis
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 2 1 Clarification: Persons with cystic fibrosis are at increased risk for diabetes, liver disease, gallbladder disease, and VTE (particularly related to use of central venous catheters) and are frequently prescribed antibiotics. Categories assigned to such conditions in U.S. MEC should be the same for persons with cystic fibrosis who have these conditions. For cystic fibrosis, classifications are based on the assumption that no other conditions are present; these classifications must be modified in the presence of such conditions.
Clarification: Certain drugs to treat cystic fibrosis (e.g., lumacaftor) might reduce effectiveness of hormonal contraceptives, including oral, injectable, transdermal, and implantable contraceptives.
Evidence: Limited evidence suggests that use of COCs or oral contraceptives (type not specified) among women with cystic fibrosis is not associated with worsening of disease severity. Very limited evidence suggests that cystic fibrosis does not impair the effectiveness of hormonal contraception (147).
Comment: Persons with cystic fibrosis have a higher prevalence of osteopenia, osteoporosis, and fragility fractures than the general population. Use of DMPA, which has been associated with small changes in BMD, might be of concern.
Hematologic Conditions
Thalassemia 1 1 1
Sickle cell disease
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 2/3 1 Clarification (DMPA): The category should be assessed according to the severity of the condition and risk for thrombosis.
Evidence: Limited evidence suggests that POC use does not increase risk for thrombosis among persons with sickle cell disease (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516). Persons with sickle cell disease are at higher risk for stroke and venous thrombosis than the general population (148151). Use of DMPA, which has been associated with a higher risk for venous thrombosis compared with nonuse (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516), might further elevate risk for thrombosis among persons with sickle cell disease. POC might be beneficial in reducing clinical symptoms (e.g., pain crises) (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
Iron deficiency anemia 1 1 1 Comment: Changes in the menstrual pattern associated with POC use have little effect on hemoglobin levels.
Solid Organ Transplantation
Solid organ
transplantation

This condition is
associated with increased
risk for adverse health
events as a result of
pregnancy (Box 3).
a. No graft failure 2 2/3 2 Clarification (DMPA): DMPA use among persons receiving long-term immunosuppressive therapy with a history of, or risk factors for, nontraumatic fractures is classified as category 3. Otherwise, DMPA use for persons with solid organ transplantation is classified as category 2.
Evidence: One study observed no differences in transplant-related adverse outcomes (e.g., infection, graft failure, and graft rejection) or occurrence of pregnancy between transplant recipients using the implant and those using no hormonal method (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516). No direct evidence was identified on bone health or fracture with use of POCs, including DMPA, among persons with solid organ transplantation. Persons with solid organ transplantation have a higher prevalence of osteoporosis and fracture than the general population, especially in the early posttransplantation period (152). Use of DMPA, which has been associated with small changes in bone mineral density compared with nonuse (4) might further elevate risk for fracture among persons with solid organ transplantation.
b. Graft failure 2 2/3 2
Drug Interactions
Antiretrovirals used for
prevention (PrEP) or
treatment of HIV infection
Comment: These recommendations generally are for ARV agents used alone. However, most persons receiving ARV are using multiple drugs in combination. In general, whether interactions between ARVs and hormonal contraceptives differ when ARVs are given alone or in combination is unknown.
See the following guidelines for the most up-to-date recommendations on drug-drug interactions between hormonal contraception and antiretrovirals: 1) Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions to Reduce Perinatal HIV Transmission in the United States (https://clinicalinfo.hiv.gov/en/guidelines/perinatal/prepregnancy-counseling-childbearing-age-overview?view=full#table-3) (153) and 2) Guidelines for the Use of Antiretroviral Agents in Adults and Adolescents with HIV (https://clinicalinfo.hiv.gov/en/guidelines/hiv-clinical-guidelines-adult-and-adolescent-arv/drug-interactions-overview?view=full) (154).
a. Nucleoside reverse
transcriptase inhibitors
(NRTIs)
  i. Abacavir (ABC) 1 1 1 Evidence: NRTIs do not appear to have significant risk for interactions with hormonal contraceptive methods (155160).
  ii. Tenofovir (TDF) 1 1 1
  iii. Zidovudine (AZT) 1 1 1
  iv. Lamivudine (3TC) 1 1 1
  v. Didanosine (DDI) 1 1 1
  vi. Emtricitabine (FTC) 1 1 1
  vii. Stavudine (D4T) 1 1 1
b. Nonnucleoside reverse
transcriptase inhibitors
(NNRTIs)
  i. Efavirenz (EFV) 2 1 2 Clarification: Evidence suggests drug interactions between EFV and certain hormonal contraceptives. These interactions might reduce the effectiveness of the hormonal contraceptive.
Evidence: One study found that women using etonogestrel implants with EFV had a higher pregnancy rate than women not using ARVs, although confidence intervals overlapped and absolute pregnancy rates were still lower than for other hormonal methods; another study found that etonogestrel levels were decreased and 5% of women had presumptive ovulation while using etonogestrel implants with EFV (161,162). Three studies of women using LNG implants demonstrated increased pregnancy rates for women using EFV-containing ARV therapy compared with no ARV use, although absolute pregnancy rates were still lower than for other hormonal methods in one study (162164); another study of LNG implant users found no difference in pregnancy rates with EFV compared with no EFV (165). No significant effects were found on pregnancy rates, DMPA levels, EFV levels, or HIV disease progression in women using DMPA and EFV compared with DMPA alone (162,165169). No significant effects were found on HIV disease progression in women using LNG implants and EFV compared with no ARVs (164). No data have assessed effectiveness of contraceptive implants during later years of use when progestin concentrations are lower and risk for failure from drug interactions might be greater.
  ii. Etravirine (ETR) 1 1 1
  iii. Nevirapine (NVP) 1 1 1 Evidence: Five studies found no significant increase in pregnancy rates among women using implants and NVP compared with implants alone (162165,170). Four studies found no significant increase in pregnancy rates among women using DMPA or other contraceptive injectables and NVP compared with DMPA or other contraceptive injectables alone (162,165,168,171). One study found no ovulations or changes in DMPA concentrations (166). No effect was found on HIV disease progression with use of NVP and DMPA or LNG implants (164,166,168170,172). No data have assessed effectiveness of contraceptive implants during later years of use when progestin concentrations are lower and risk for failure from drug interactions might be greater.
  iv. Rilpivirine (RPV) 1 1 1
c. Ritonavir-boosted
protease inhibitors
  i. Ritonavir-boosted atazanavir (ATV/r) 2 1 2 Clarification: Theoretically, drug interactions might occur between certain ritonavir-boosted protease inhibitors and certain hormonal contraceptives that might reduce the effectiveness of the hormonal contraceptive. Any potential effect on contraceptive effectiveness is likely to be lower with DMPA than with other POCs because of the higher dose of DMPA.
Evidence: One pharmacokinetic study demonstrated increased progestin concentrations with use of POPs and ATV/r compared with POPs alone (173).
  ii. Ritonavir-boosted darunavir (DRV/r) 2 1 2 Clarification: Theoretically, drug interactions might occur between certain ritonavir-boosted protease inhibitors and certain hormonal contraceptives that might reduce the effectiveness of the hormonal contraceptive. Any potential effect on contraceptive effectiveness is likely to be lower with DMPA than with other POCs because of the higher dose of DMPA.
  iii. Ritonavir-boosted fosamprenavir (FPV/r) 2 1 2 Clarification: Theoretically, drug interactions might occur between certain ritonavir-boosted protease inhibitors and certain hormonal contraceptives that might reduce the effectiveness of the hormonal contraceptive. Any potential effect on contraceptive effectiveness is likely to be lower with DMPA than with other POCs because of the higher dose of DMPA.
  iv. Ritonavir-boosted lopinavir (LPV/r) 1 1 1 Evidence: One study demonstrated no pregnancies, no ovulations, no change in LPV/r level, and no change in HIV disease progression in women using DMPA (174); another study found a small increase in pregnancy rate in women using DMPA with LPV/r compared with no ARV therapy, however confidence intervals overlapped (162). Two studies found no increased risk for pregnancy in women using implants (162,163). Two studies found contraceptive hormones increased in women using LPV/r with DMPA or etonogestrel implants (161,174).
  v. Ritonavir-boosted saquinavir (SQV/r) 2 1 2 Clarification: Theoretically, drug interactions might occur between certain ritonavir-boosted protease inhibitors and certain hormonal contraceptives that might reduce the effectiveness of the hormonal contraceptive. Any potential effect on contraceptive effectiveness is likely to be lower with DMPA than with other POCs because of the higher dose of DMPA.
  vi. Ritonavir-boosted tipranavir (TPV/r) 2 1 2 Clarification: Theoretically, drug interactions might occur between certain ritonavir-boosted protease inhibitors and certain hormonal contraceptives that might reduce the effectiveness of the hormonal contraceptive. Any potential effect on contraceptive effectiveness is likely to be lower with DMPA than with other POCs because of the higher dose of DMPA.
d. Protease inhibitors
without ritonavir
  i. Atazanavir (ATV) 1 1 1 Comment: When ATV is administered with cobicistat, theoretical concern exists for a drug interaction with hormonal contraceptives. Cobicistat is an inhibitor of CYP3A and CYP2D6 and could theoretically increase contraceptive hormone levels. However, its effects on CYP enzymes and drug levels might vary when combined with other ARVs.
  ii. Fosamprenavir (FPV) 2 2 2 Clarification: Theoretical concern exists that interactions between FPV and hormonal contraceptives leading to decreased levels of FPV might diminish effectiveness of the ARV drug. The drug interaction likely involves CYP3A4 pathways; POCs have less effect on CYP3A4 enzymes than CHCs.
  iii. Indinavir (IDV) 1 1 1
  iv. Nelfinavir (NFV) 2 1 2 Clarification: Theoretically, drug interactions might occur between certain protease inhibitors and certain hormonal contraceptives that might reduce the effectiveness of the hormonal contraceptive. Any potential effect on contraceptive effectiveness is likely to be lower with DMPA than with other POCs because of the higher dose of DMPA. Concern exists that interactions between NFV and POCs might decrease NFV levels.
Evidence: One study found no pregnancies, no ovulations, no change in DMPA concentrations and no change in HIV disease progression with use of DMPA and NFV compared with DMPA alone; NFV concentrations were decreased with concomitant DMPA use (166,168).
e. CCR5 co-receptor
antagonists
  i. Maraviroc (MVC) 1 1 1
f. HIV integrase strand transfer
inhibitors
  i. Raltegravir (RAL) 1 1 1
  ii. Dolutegravir (DTG) 1 1 1
  iii. Elvitegravir (EVG) 1 1 1 Comment: When EVG is administered with cobicistat, theoretical concern exists for a drug interaction with hormonal contraceptives. Cobicistat is an inhibitor of CYP3A and CYP2D6 and could theoretically increase contraceptive hormone levels. However, its effects on CYP enzymes and drug levels might vary when combined with other ARVs.
g. Fusion inhibitors
  i. Enfuvirtide 1 1 1
Anticonvulsant therapy
a. Certain anticonvulsants (phenytoin, carbamazepine, barbiturates, primidone, topiramate, and oxcarbazepine) 2 1 3 Clarification: Although the interaction of certain anticonvulsants with POPs and etonogestrel implants is not harmful, it is likely to reduce the effectiveness of POPs and etonogestrel implants. Whether increasing the hormone dose of POPs alleviates this concern remains unclear. Use of other contraceptives should be encouraged for persons who are long-term users of any of these drugs. Use of DMPA is a category 1 because its effectiveness is not decreased by use of certain anticonvulsants.
Evidence: Use of certain anticonvulsants might decrease the effectiveness of POCs (175178).
b. Lamotrigine 1 1 1 Evidence: No drug interactions have been reported among women with epilepsy receiving lamotrigine and POCs (178,179).
Antimicrobial therapy
a. Broad-spectrum antibiotics 1 1 1
b. Antifungals 1 1 1
c. Antiparasitics 1 1 1
d. Rifampin or rifabutin therapy 2 1 3 Clarification: Although the interaction of rifampin or rifabutin with POPs and etonogestrel implants is not harmful, it is likely to reduce the effectiveness of POPs and etonogestrel implants. Use of other contraceptives should be encouraged for persons who are long-term users of any of these drugs. Use of DMPA is a category 1 because its effectiveness is not decreased by use of rifampin or rifabutin. Whether increasing the hormone dose of POPs alleviates this concern remains unclear.
Psychotropic medications Comment: For many common psychotropic agents, limited or no theoretical concern exits for clinically significant drug interactions when co-administered with hormonal contraceptives. However, either no or very limited data exist examining potential interactions for these classes of medications.
a. Selective serotonin reuptake inhibitors (SSRIs) 1 1 1 Evidence: No evidence specifically examined the use of POCs with SSRIs. Limited clinical and pharmacokinetic data do not demonstrate concern for SSRIs decreasing the effectiveness of oral contraceptives. Limited evidence suggests that for women taking SSRIs, the use of hormonal contraceptives was not associated with differences in effectiveness of the SSRI for treatment or in adverse events when compared with women not taking hormonal contraceptives (180).
Comment: Drugs that are inhibitors of CYP3A4 or CYP2C9 theoretically have the potential to increase levels of contraceptive steroid, which might increase adverse events. Fluvoxamine is an SSRI known to be a moderate inhibitor of both 3A4 and 2C9; however, no clinical or pharmacokinetic studies were identified to explore potential drug-drug interactions.
St. John’s wort 2 1 2 Evidence: No evidence specifically examined the use of POCs with St. John’s wort. Although clinical data are limited, studies with pharmacokinetic and pharmacodynamics outcomes raise concern that St. John’s wort might decrease effectiveness of hormonal contraceptives, including increased risk for breakthrough bleeding and ovulation and increased metabolism of estrogen and progestin. Any interactions might be dependent on the dose of St. John’s wort, and the concentration of active ingredients across types of St. John’s wort preparations might vary (181).
Comment: Any potential effect on contraceptive effectiveness is likely to be lower with DMPA than with other POCs because of the higher dose of DMPA.

Abbreviations: ARV = antiretroviral; BMD = bone mineral density; BMI = body mass index; CHC = combined hormonal contraceptive; CKD = chronic kidney disease; COC = combined oral contraceptive; Cu-IUD = copper intrauterine device; CYP = cytochrome P450; DMPA = depot medroxyprogesterone acetate; DRSP = drospirenone; DVT = deep venous thrombosis; hCG = human chorionic gonadotropin; HDL = high-density lipoprotein; IBD = inflammatory bowel disease; IM = intramuscular; LDL = low-density lipoprotein; LNG = levonorgestrel; LNG-IUD = levonorgestrel intrauterine device; NA = not applicable; PE = pulmonary embolism; PID = pelvic inflammatory disease; POC = progestin-only contraceptive; POP = progestin-only pill; PrEP = pre-exposure prophylaxis; RCT = randomized clinical trial; SLE = systemic lupus erythematosus; STI = sexually transmitted infection; U.S. MEC = U.S. Medical Eligibility Criteria for Contraceptive Use; U.S. SPR = U.S. Selected Practice Recommendations for Contraceptive Use; VTE = venous thromboembolism.

References

  1. Curtis KM, Nguyen AT, Tepper NK, et al. U.S. selected practice recommendations for contraceptive use, 2024. 2024;73(No. RR-3):1–77.
  2. Workowski KA, Bachmann LH, Chan PA, et al. Sexually transmitted infections treatment guidelines, 2021. 2021;70(No. RR-4):1–187. https://doi.org/10.15585/mmwr.rr7004a1 PMID:34292926
  3. CDC. US Public Health Service preexposure prophylaxis for the prevention of HIV infection in the United States—2021 update: a clinical practice guideline. Atlanta, GA: US Department of Health and Human Services, CDC; 2021. https://www.cdc.gov/hiv/pdf/risk/prep/cdc-hiv-prep-guidelines-2021.pdf
  4. Curtis KM, Martins SL. Progestogen-only contraception and bone mineral density: a systematic review. 2006;73:470–87. https://doi.org/10.1016/j.contraception.2005.12.010 PMID:16627031
  5. Lanza LL, McQuay LJ, Rothman KJ, et al. Use of depot medroxyprogesterone acetate contraception and incidence of bone fracture. 2013;121:593–600. https://doi.org/10.1097/AOG.0b013e318283d1a1 PMID:23635623
  6. Harel Z, Riggs S, Vaz R, Flanagan P, Harel D, Machan JT. Bone accretion in adolescents using the combined estrogen and progestin transdermal contraceptive method Ortho Evra: a pilot study. 2010;23:23–31. https://doi.org/10.1016/j.jpag.2009.04.008 PMID:19647454
  7. Kaunitz AM, Miller PD, Rice VM, Ross D, McClung MR. Bone mineral density in women aged 25–35 years receiving depot medroxyprogesterone acetate: recovery following discontinuation. 2006;74:90–9. https://doi.org/10.1016/j.contraception.2006.03.010 PMID:16860045
  8. Kaunitz AM, Arias R, McClung M. Bone density recovery after depot medroxyprogesterone acetate injectable contraception use. 2008;77:67–76. https://doi.org/10.1016/j.contraception.2007.10.005 PMID:18226668
  9. Kaunitz AM, Darney PD, Ross D, Wolter KD, Speroff L. Subcutaneous DMPA vs. intramuscular DMPA: a 2-year randomized study of contraceptive efficacy and bone mineral density. 2009;80:7–17. https://doi.org/10.1016/j.contraception.2009.02.005 PMID:19501210
  10. Lappe JM, Stegman MR, Recker RR. The impact of lifestyle factors on stress fractures in female Army recruits. 2001;12:35–42. https://doi.org/10.1007/s001980170155 PMID:11305081
  11. Lara-Torre E, Edwards CP, Perlman S, Hertweck SP. Bone mineral density in adolescent females using depot medroxyprogesterone acetate. 2004;17:17–21. https://doi.org/10.1016/j.jpag.2003.11.017 PMID:15010034
  12. Lopez LM, Chen M, Mullins S, Curtis KM, Helmerhorst FM. Steroidal contraceptives and bone fractures in women: evidence from observational studies. 2012;8:CD009849. PMID:22895991
  13. Lopez LM, Grimes DA, Schulz KF, Curtis KM. Steroidal contraceptives: effect on bone fractures in women. 2011; (7):CD006033. PMID:21735401
  14. Meier C, Brauchli YB, Jick SS, Kraenzlin ME, Meier CR. Use of depot medroxyprogesterone acetate and fracture risk. 2010;95:4909–16. https://doi.org/10.1210/jc.2010-0032 PMID:20685865
  15. Merki-Feld GS, Neff M, Keller PJ. A 2-year prospective study on the effects of depot medroxyprogesterone acetate on bone mass-response to estrogen and calcium therapy in individual users. 2003;67:79–86. https://doi.org/10.1016/S0010-7824(02)00460-2 PMID:12586317
  16. Monteiro-Dantas C, Espejo-Arce X, Lui-Filho JF, Fernandes AM, Monteiro I, Bahamondes L. A three-year longitudinal evaluation of the forearm bone density of users of etonogestrel- and levonorgestrel-releasing contraceptive implants. 2007;4:11. https://doi.org/10.1186/1742-4755-4-11 PMID:17997844
  17. Naessen T, Olsson SE, Gudmundson J. Differential effects on bone density of progestogen-only methods for contraception in premenopausal women. 1995;52:35–9. https://doi.org/10.1016/0010-7824(95)00121-P PMID:8521712
  18. Sanches L, Marchi NM, Castro S, Juliato CT, Villarroel M, Bahamondes L. Forearm bone mineral density in postmenopausal former users of depot medroxyprogesterone acetate. 2008;78:365–9. https://doi.org/10.1016/j.contraception.2008.07.013 PMID:18929732
  19. Scholes D, LaCroix AZ, Ichikawa LE, Barlow WE, Ott SM. Injectable hormone contraception and bone density: results from a prospective study. 2002;13:581–7. https://doi.org/10.1097/00001648-200209000-00015 PMID:12192229
  20. Scholes D, LaCroix AZ, Ichikawa LE, Barlow WE, Ott SM. Change in bone mineral density among adolescent women using and discontinuing depot medroxyprogesterone acetate contraception. 2005;159:139–44. https://doi.org/10.1001/archpedi.159.2.139 PMID:15699307
  21. Segall-Gutierrez P, Agarwal R, Ge M, Lopez C, Hernandez G, Stanczyk FZ. A pilot study examining short-term changes in bone mineral density among class 3 obese users of depot-medroxyprogesterone acetate. 2013;18:199–205. https://doi.org/10.3109/13625187.2013.774358 PMID:23530919
  22. Tang OS, Tang G, Yip PS, Li B. Further evaluation on long-term depot-medroxyprogesterone acetate use and bone mineral density: a longitudinal cohort study. 2000;62:161–4. https://doi.org/10.1016/S0010-7824(00)00168-2 PMID:11137068
  23. Vestergaard P, Rejnmark L, Mosekilde L. The effects of depot medroxyprogesterone acetate and intrauterine device use on fracture risk in Danish women. 2008;78:459–64. https://doi.org/10.1016/j.contraception.2008.07.014 PMID:19014791
  24. Viola AS, Castro S, Marchi NM, Bahamondes MV, Viola CF, Bahamondes L. Long-term assessment of forearm bone mineral density in postmenopausal former users of depot medroxyprogesterone acetate. 2011;84:122–7. https://doi.org/10.1016/j.contraception.2010.11.007 PMID:21757052
  25. Walsh JS, Eastell R, Peel NF. Depot medroxyprogesterone acetate use after peak bone mass is associated with increased bone turnover but no decrease in bone mineral density. 2010;93:697–701. https://doi.org/10.1016/j.fertnstert.2008.10.004 PMID:19013564
  26. Wetmore CM, Ichikawa L, LaCroix AZ, Ott SM, Scholes D. Association between caffeine intake and bone mass among young women: potential effect modification by depot medroxyprogesterone acetate use. 2008;19:519–27. https://doi.org/10.1007/s00198-007-0473-2 PMID:18004611
  27. Wong AY, Tang LC, Chin RK. Levonorgestrel-releasing intrauterine system (Mirena) and Depot medroxyprogesterone acetate (Depoprovera) as long-term maintenance therapy for patients with moderate and severe endometriosis: a randomised controlled trial. 2010;50:273–9. https://doi.org/10.1111/j.1479-828X.2010.01152.x PMID:20618247
  28. Yang KY, Kim YS, Ji YI, Jung MH. Changes in bone mineral density of users of the levonorgestrel-releasing intrauterine system. 2012;79:190–4. https://doi.org/10.1272/jnms.79.190 PMID:22791119
  29. Zhang MH, Zhang W, Zhang AD, Yang Y, Gai L. Effect of depot medroxyprogesterone acetate on bone mineral density in adolescent women. 2013;126:4043–7. https://doi.org/10.3760/cma.j.issn.0366-6999.20130885 PMID:24229671
  30. Bahamondes MV, Monteiro I, Castro S, Espejo-Arce X, Bahamondes L. Prospective study of the forearm bone mineral density of long-term users of the levonorgestrel-releasing intrauterine system. 2010;25:1158–64. https://doi.org/10.1093/humrep/deq043 PMID:20185512
  31. Banks E, Berrington A, Casabonne D. Overview of the relationship between use of progestogen-only contraceptives and bone mineral density. 2001;108:1214–21. https://doi.org/10.1111/j.1471-0528.2001.00296.x PMID:11843382
  32. Beerthuizen R, van Beek A, Massai R, Mäkäräinen L, Hout J, Bennink HC. Bone mineral density during long-term use of the progestagen contraceptive implant Implanon compared to a non-hormonal method of contraception. 2000;15:118–22. https://doi.org/10.1093/humrep/15.1.118 PMID:10611199
  33. Beksinska ME, Kleinschmidt I, Smit JA, Farley TM. Bone mineral density in adolescents using norethisterone enanthate, depot-medroxyprogesterone acetate or combined oral contraceptives for contraception. 2007;75:438–43. https://doi.org/10.1016/j.contraception.2007.02.001 PMID:17519149
  34. Beksinska ME, Kleinschmidt I, Smit JA, Farley TM. Bone mineral density in a cohort of adolescents during use of norethisterone enanthate, depot-medroxyprogesterone acetate or combined oral contraceptives and after discontinuation of norethisterone enanthate. 2009;79:345–9. https://doi.org/10.1016/j.contraception.2008.11.009 PMID:19341845
  35. Berenson AB, Breitkopf CR, Grady JJ, Rickert VI, Thomas A. Effects of hormonal contraception on bone mineral density after 24 months of use. 2004;103:899–906. https://doi.org/10.1097/01.AOG.0000117082.49490.d5 PMID:15121563
  36. Berenson AB, Rahman M, Breitkopf CR, Bi LX. Effects of depot medroxyprogesterone acetate and 20-microgram oral contraceptives on bone mineral density. 2008;112:788–99. https://doi.org/10.1097/AOG.0b013e3181875b78 PMID:18827121
  37. Busen NH, Britt RB, Rianon N. Bone mineral density in a cohort of adolescent women using depot medroxyprogesterone acetate for one to two years. 2003;32:257–9. https://doi.org/10.1016/S1054-139X(02)00567-0 PMID:12667729
  38. Caird LE, Reid-Thomas V, Hannan WJ, Gow S, Glasier AF. Oral progestogen-only contraception may protect against loss of bone mass in breast-feeding women. 1994;41:739–45. https://doi.org/10.1111/j.1365-2265.1994.tb02788.x PMID:7889609
  39. Clark MK, Sowers M, Levy B, Nichols S. Bone mineral density loss and recovery during 48 months in first-time users of depot medroxyprogesterone acetate. 2006;86:1466–74. https://doi.org/10.1016/j.fertnstert.2006.05.024 PMID:16996507
  40. Cromer BA, Lazebnik R, Rome E, et al. Double-blinded randomized controlled trial of estrogen supplementation in adolescent girls who receive depot medroxyprogesterone acetate for contraception. 2005;192:42–7. https://doi.org/10.1016/j.ajog.2004.07.041 PMID:15672001
  41. Cromer BA, Blair JM, Mahan JD, Zibners L, Naumovski Z. A prospective comparison of bone density in adolescent girls receiving depot medroxyprogesterone acetate (Depo-Provera), levonorgestrel (Norplant), or oral contraceptives. 1996;129:671–6. https://doi.org/10.1016/S0022-3476(96)70148-8 PMID:8917232
  42. Cromer BA, Bonny AE, Stager M, et al. Bone mineral density in adolescent females using injectable or oral contraceptives: a 24-month prospective study. 2008;90:2060–7. https://doi.org/10.1016/j.fertnstert.2007.10.070 PMID:18222431
  43. Cromer BA, Stager M, Bonny A, et al. Depot medroxyprogesterone acetate, oral contraceptives and bone mineral density in a cohort of adolescent girls. 2004;35:434–41. https://doi.org/10.1016/j.jadohealth.2004.07.005 PMID:15581522
  44. Cundy T, Ames R, Horne A, et al. A randomized controlled trial of estrogen replacement therapy in long-term users of depot medroxyprogesterone acetate. 2003;88:78–81. https://doi.org/10.1210/jc.2002-020874 PMID:12519833
  45. Cundy T, Cornish J, Evans MC, Roberts H, Reid IR. Recovery of bone density in women who stop using medroxyprogesterone acetate. 1994;308:247–8. https://doi.org/10.1136/bmj.308.6923.247 PMID:8111260
  46. Cundy T, Cornish J, Roberts H, Reid IR. Menopausal bone loss in long-term users of depot medroxyprogesterone acetate contraception. 2002;186:978–83. https://doi.org/10.1067/mob.2002.122420 PMID:12015524
  47. Di X, Li Y, Zhang C, Jiang J, Gu S. Effects of levonorgestrel-releasing subdermal contraceptive implants on bone density and bone metabolism. 1999;60:161–6. https://doi.org/10.1016/S0010-7824(99)00080-3 PMID:10640160
  48. Díaz S, Reyes MV, Zepeda A, et al. Norplant((R)) implants and progesterone vaginal rings do not affect maternal bone turnover and density during lactation and after weaning. 1999;14:2499–505. https://doi.org/10.1093/humrep/14.10.2499 PMID:10527977
  49. Gai L, Zhang J, Zhang H, Gai P, Zhou L, Liu Y. The effect of depot medroxyprogesterone acetate (DMPA) on bone mineral density (BMD) and evaluating changes in BMD after discontinuation of DMPA in Chinese women of reproductive age. 2011;83:218–22. https://doi.org/10.1016/j.contraception.2010.07.027 PMID:21310282
  50. Bahamondes L, Espejo-Arce X, Hidalgo MM, Hidalgo-Regina C, Teatin-Juliato C, Petta CA. A cross-sectional study of the forearm bone density of long-term users of levonorgestrel-releasing intrauterine system. 2006;21:1316–9. https://doi.org/10.1093/humrep/dei457 PMID:16373404
  51. Bahamondes L, Monteiro-Dantas C, Espejo-Arce X, et al. A prospective study of the forearm bone density of users of etonorgestrel- and levonorgestrel-releasing contraceptive implants. 2006;21:466–70. https://doi.org/10.1093/humrep/dei358 PMID:16253974
  52. Pitts SA, Feldman HA, Dorale A, Gordon CM. Bone mineral density, fracture, and vitamin D in adolescents and young women using depot medroxyprogesterone acetate. 2012;25:23–6. https://doi.org/10.1016/j.jpag.2011.07.014 PMID:22078997
  53. US Department of Agriculture; US Department of Health and Human Services. Dietary guidelines for Americans, 2020–2025. 9th ed. Washington, DC: US Department of Agriculture and US Department of Health and Human Services; 2020. https://www.dietaryguidelines.gov/sites/default/files/2021-03/Dietary_Guidelines_for_Americans-2020-2025.pdf
  54. Meek JY, Noble L; Section on Breastfeeding. Policy statement: breastfeeding and the use of human milk. 2022;150:e2022057988. https://doi.org/10.1542/peds.2022-057988 PMID:35921640
  55. Braga GC, Ferriolli E, Quintana SM, Ferriani RA, Pfrimer K, Vieira CS. Immediate postpartum initiation of etonogestrel-releasing implant: A randomized controlled trial on breastfeeding impact. 2015;92:536–42. https://doi.org/10.1016/j.contraception.2015.07.009 PMID:26209863
  56. Phillips SJ, Tepper NK, Kapp N, Nanda K, Temmerman M, Curtis KM. Progestogen-only contraceptive use among breastfeeding women: a systematic review. 2016;94:226–52. https://doi.org/10.1016/j.contraception.2015.09.010 PMID:26410174
  57. Raymond EG, Weaver MA, Louie KS, et al. Effects of depot medroxyprogesterone acetate injection timing on medical abortion efficacy and repeat pregnancy: a randomized controlled trial. 2016;128:739–45. https://doi.org/10.1097/AOG.0000000000001627 PMID:27607859
  58. Kim C, Nguyen AT, Berry-Bibee E, Ermias Y, Gaffield ME, Kapp N. Systemic hormonal contraception initiation after abortion: a systematic review and meta-analysis. 2021;103:291–304. https://doi.org/10.1016/j.contraception.2021.01.017 PMID:33548267
  59. Beksinska ME, Smit JA, Kleinschmidt I, Milford C, Farley TM. Prospective study of weight change in new adolescent users of DMPA, NET-EN, COCs, nonusers and discontinuers of hormonal contraception. 2010;81:30–4. https://doi.org/10.1016/j.contraception.2009.07.007 PMID:20004270
  60. Bender NM, Segall-Gutierrez P, Najera SO, Stanczyk FZ, Montoro M, Mishell DR Jr. Effects of progestin-only long-acting contraception on metabolic markers in obese women. 2013;88:418–25. https://doi.org/10.1016/j.contraception.2012.12.007 PMID:23410714
  61. Berenson AB, Rahman M. Changes in weight, total fat, percent body fat, and central-to-peripheral fat ratio associated with injectable and oral contraceptive use. 2009;200:329.e1–8. https://doi.org/10.1016/j.ajog.2008.12.052 PMID:19254592
  62. Bonny AE, Secic M, Cromer B. Early weight gain related to later weight gain in adolescents on depot medroxyprogesterone acetate. 2011;117:793–7. https://doi.org/10.1097/AOG.0b013e31820f387c PMID:21422849
  63. Bonny AE, Ziegler J, Harvey R, Debanne SM, Secic M, Cromer BA. Weight gain in obese and nonobese adolescent girls initiating depot medroxyprogesterone, oral contraceptive pills, or no hormonal contraceptive method. 2006;160:40–5. https://doi.org/10.1001/archpedi.160.1.40 PMID:16389209
  64. Clark MK, Dillon JS, Sowers M, Nichols S. Weight, fat mass, and central distribution of fat increase when women use depot-medroxyprogesterone acetate for contraception. 2005;29:1252–8. https://doi.org/10.1038/sj.ijo.0803023 PMID:15997247
  65. Gerlach LS, Saldaña SN, Wang Y, Nick TG, Spigarelli MG. Retrospective review of the relationship between weight change and demographic factors following initial depot medroxyprogesterone acetate injection in adolescents. 2011;33:182–7. https://doi.org/10.1016/j.clinthera.2011.02.008 PMID:21397330
  66. Jain J, Jakimiuk AJ, Bode FR, Ross D, Kaunitz AM. Contraceptive efficacy and safety of DMPA-SC. 2004;70:269–75. https://doi.org/10.1016/j.contraception.2004.06.011 PMID:15451329
  67. Kozlowski KJ, Rickert VI, Hendon A, Davis P. Adolescents and Norplant: preliminary findings of side effects. 1995;16:373–8. https://doi.org/10.1016/S1054-139X(94)00029-E PMID:7662687
  68. Le YL, Rahman M, Berenson AB. Early weight gain predicting later weight gain among depot medroxyprogesterone acetate users. 2009;114:279–84. https://doi.org/10.1097/AOG.0b013e3181af68b2 PMID:19622988
  69. Leiman G. Depo-medroxyprogesterone acetate as a contraceptive agent: its effect on weight and blood pressure. 1972;114:97–102. https://doi.org/10.1016/0002-9378(72)90296-7 PMID:4637044
  70. Lopez LM, Grimes DA, Chen M, et al. Hormonal contraceptives for contraception in overweight or obese women. 2013;4:CD008452. PMID:23633356
  71. Mangan SA, Larsen PG, Hudson S. Overweight teens at increased risk for weight gain while using depot medroxyprogesterone acetate. 2002;15:79–82. https://doi.org/10.1016/S1083-3188(01)00147-4 PMID:12057528
  72. Nyirati CM, Habash DL, Shaffer LE. Weight and body fat changes in postpartum depot-medroxyprogesterone acetate users. 2013;88:169–76. https://doi.org/10.1016/j.contraception.2012.10.016 PMID:23177262
  73. Pantoja M, Medeiros T, Baccarin MC, Morais SS, Bahamondes L, dos Santos Fernandes AM. Variations in body mass index of users of depot-medroxyprogesterone acetate as a contraceptive. 2010;81:107–11. https://doi.org/10.1016/j.contraception.2009.07.008 PMID:20103446
  74. Risser WL, Gefter LR, Barratt MS, Risser JM. Weight change in adolescents who used hormonal contraception. 1999;24:433–6. https://doi.org/10.1016/S1054-139X(98)00151-7 PMID:10401972
  75. Segall-Gutierrez P, Xiang AH, Watanabe RM, et al. Deterioration in cardiometabolic risk markers in obese women during depot medroxyprogesterone acetate use. 2012;85:36–41. https://doi.org/10.1016/j.contraception.2011.04.016 PMID:22067800
  76. Westhoff C, Jain JK, Milsom I, Ray A. Changes in weight with depot medroxyprogesterone acetate subcutaneous injection 104 mg/0.65 mL. 2007;75:261–7. https://doi.org/10.1016/j.contraception.2006.12.009 PMID:17362703
  77. Paulen ME, Zapata LB, Cansino C, Curtis KM, Jamieson DJ. Contraceptive use among women with a history of bariatric surgery: a systematic review. 2010;82:86–94. https://doi.org/10.1016/j.contraception.2010.02.008 PMID:20682146
  78. World Health Organization Collaborative Study of Cardiovascular Disease and Steroid Hormone Contraception. Cardiovascular disease and use of oral and injectable progestogen-only contraceptives and combined injectable contraceptives. Results of an international, multicenter, case-control study. 1998;57:315–24. https://doi.org/10.1016/S0010-7824(98)00041-9 PMID:9673838
  79. Martinelli I, Lensing AW, Middeldorp S, et al. Recurrent venous thromboembolism and abnormal uterine bleeding with anticoagulant and hormone therapy use. 2016;127:1417–25. https://doi.org/10.1182/blood-2015-08-665927 PMID:26696010
  80. Sönmezer M, Atabekoğlu C, Cengiz B, Dökmeci F, Cengiz SD. Depot-medroxyprogesterone acetate in anticoagulated patients with previous hemorrhagic corpus luteum. 2005;10:9–14. https://doi.org/10.1080/13625180400020952 PMID:16036292
  81. Barbhaiya M, Zuily S, Naden R, et al. ; ACR/EULAR APS Classification Criteria Collaborators. The 2023 ACR/EULAR Antiphospholipid Syndrome Classification Criteria. 2023;75:1687–702. https://doi.org/10.1002/art.42624 PMID:37635643
  82. Bergendal A, Persson I, Odeberg J, et al. Association of venous thromboembolism with hormonal contraception and thrombophilic genotypes. 2014;124:600–9. https://doi.org/10.1097/AOG.0000000000000411 PMID:25162263
  83. Leon L, Giannoukas AD, Dodd D, Chan P, Labropoulos N. Clinical significance of superficial vein thrombosis. 2005;29:10–7. https://doi.org/10.1016/j.ejvs.2004.09.021 PMID:15570265
  84. Tepper NK, Paulen ME, Marchbanks PA, Curtis KM. Safety of contraceptive use among women with peripartum cardiomyopathy: a systematic review. 2010;82:95–101. https://doi.org/10.1016/j.contraception.2010.02.004 PMID:20682147
  85. The Criteria Committee of the New York Heart Association. Nomenclature and criteria for diagnosis of diseases of the heart and great vessels. 9th ed. Boston, MA: Little, Brown and Co; 1994.
  86. Folsom AR, Lutsey PL, Astor BC, Wattanakit K, Heckbert SR, Cushman M; Atherosclerosis Risk in Communities Study. Chronic kidney disease and venous thromboembolism: a prospective study. 2010;25:3296–301. https://doi.org/10.1093/ndt/gfq179 PMID:20353958
  87. Kayali F, Najjar R, Aswad F, Matta F, Stein PD. Venous thromboembolism in patients hospitalized with nephrotic syndrome. 2008;121:226–30. https://doi.org/10.1016/j.amjmed.2007.08.042 PMID:18328307
  88. Mahmoodi BK, ten Kate MK, Waanders F, et al. High absolute risks and predictors of venous and arterial thromboembolic events in patients with nephrotic syndrome: results from a large retrospective cohort study. 2008;117:224–30. https://doi.org/10.1161/CIRCULATIONAHA.107.716951 PMID:18158362
  89. Singhal R, Brimble KS. Thromboembolic complications in the nephrotic syndrome: pathophysiology and clinical management. 2006;118:397–407. https://doi.org/10.1016/j.thromres.2005.03.030 PMID:15990160
  90. Wattanakit K, Cushman M, Stehman-Breen C, Heckbert SR, Folsom AR. Chronic kidney disease increases risk for venous thromboembolism. 2008;19:135–40. https://doi.org/10.1681/ASN.2007030308 PMID:18032796
  91. Naylor KL, McArthur E, Leslie WD, et al. The three-year incidence of fracture in chronic kidney disease. 2014;86:810–8. https://doi.org/10.1038/ki.2013.547 PMID:24429401
  92. Pimentel A, Ureña-Torres P, Zillikens MC, Bover J, Cohen-Solal M. Fractures in patients with CKD-diagnosis, treatment, and prevention: a review by members of the European Calcified Tissue Society and the European Renal Association of Nephrology Dialysis and Transplantation. 2017;92:1343–55. https://doi.org/10.1016/j.kint.2017.07.021 PMID:28964571
  93. Vilaca T, Salam S, Schini M, et al. Risks of hip and nonvertebral fractures in patients with CKD G3a-G5D: a systematic review and meta-analysis. 2020;76:521–32. https://doi.org/10.1053/j.ajkd.2020.02.450 PMID:32654892
  94. Molnar AO, Bota SE, McArthur E, et al. Risk and complications of venous thromboembolism in dialysis patients. 2018;33:874–80. PMID:28992258
  95. Tveit DP, Hypolite IO, Hshieh P, et al. Chronic dialysis patients have high risk for pulmonary embolism. 2002;39:1011–7. https://doi.org/10.1053/ajkd.2002.32774 PMID:11979344
  96. Wang IK, Shen TC, Muo CH, Yen TH, Sung FC. Risk of pulmonary embolism in patients with end-stage renal disease receiving long-term dialysis. 2017;32:1386–93. PMID:27448674
  97. Alem AM, Sherrard DJ, Gillen DL, et al. Increased risk of hip fracture among patients with end-stage renal disease. 2000;58:396–9. https://doi.org/10.1046/j.1523-1755.2000.00178.x PMID:10886587
  98. Ball AM, Gillen DL, Sherrard D, et al. Risk of hip fracture among dialysis and renal transplant recipients. 2002;288:3014–8. https://doi.org/10.1001/jama.288.23.3014 PMID:12479766
  99. Jadoul M, Albert JM, Akiba T, et al. Incidence and risk factors for hip or other bone fractures among hemodialysis patients in the Dialysis Outcomes and Practice Patterns Study. 2006;70:1358–66. https://doi.org/10.1038/sj.ki.5001754 PMID:16929251
  100. Bernatsky S, Clarke A, Ramsey-Goldman R, et al. Hormonal exposures and breast cancer in a sample of women with systemic lupus erythematosus. 2004;43:1178–81. https://doi.org/10.1093/rheumatology/keh282 PMID:15226516
  101. Bernatsky S, Ramsey-Goldman R, Gordon C, et al. Factors associated with abnormal Pap results in systemic lupus erythematosus. 2004;43:1386–9. https://doi.org/10.1093/rheumatology/keh331 PMID:15280571
  102. Chopra N, Koren S, Greer WL, et al. Factor V Leiden, prothrombin gene mutation, and thrombosis risk in patients with antiphospholipid antibodies. 2002;29:1683–8. PMID:12180730
  103. Esdaile JM, Abrahamowicz M, Grodzicky T, et al. Traditional Framingham risk factors fail to fully account for accelerated atherosclerosis in systemic lupus erythematosus. 2001;44:2331–7. https://doi.org/10.1002/1529-0131(200110)44:10<2331::AID-ART395>3.0.CO;2-I PMID:11665973
  104. Julkunen HA. Oral contraceptives in systemic lupus erythematosus: side-effects and influence on the activity of SLE. 1991;20:427–33. https://doi.org/10.3109/03009749109096822 PMID:1771400
  105. Julkunen HA, Kaaja R, Friman C. Contraceptive practice in women with systemic lupus erythematosus. 1993;32:227–30. https://doi.org/10.1093/rheumatology/32.3.227 PMID:8448613
  106. Jungers P, Dougados M, Pélissier C, et al. Influence of oral contraceptive therapy on the activity of systemic lupus erythematosus. 1982;25:618–23. https://doi.org/10.1002/art.1780250603 PMID:7092961
  107. Manzi S, Meilahn EN, Rairie JE, et al. Age-specific incidence rates of myocardial infarction and angina in women with systemic lupus erythematosus: comparison with the Framingham Study. 1997;145:408–15. https://doi.org/10.1093/oxfordjournals.aje.a009122 PMID:9048514
  108. McAlindon T, Giannotta L, Taub N, D’Cruz D, Hughes G. Environmental factors predicting nephritis in systemic lupus erythematosus. 1993;52:720–4. https://doi.org/10.1136/ard.52.10.720 PMID:8257208
  109. McDonald J, Stewart J, Urowitz MB, Gladman DD. Peripheral vascular disease in patients with systemic lupus erythematosus. 1992;51:56–60. https://doi.org/10.1136/ard.51.1.56 PMID:1540039
  110. Mintz G, Gutiérrez G, Delezé M, Rodríguez E. Contraception with progestagens in systemic lupus erythematosus. 1984;30:29–38. https://doi.org/10.1016/0010-7824(84)90076-3 PMID:6434228
  111. Petri M. Musculoskeletal complications of systemic lupus erythematosus in the Hopkins Lupus Cohort: an update. 1995;8:137–45. https://doi.org/10.1002/art.1790080305 PMID:7654797
  112. Petri M. Lupus in Baltimore: evidence-based ‘clinical pearls’ from the Hopkins Lupus Cohort. 2005;14:970–3. https://doi.org/10.1191/0961203305lu2230xx PMID:16425579
  113. Petri M, Kim MY, Kalunian KC, et al. ; OC-SELENA Trial. Combined oral contraceptives in women with systemic lupus erythematosus. 2005;353:2550–8. https://doi.org/10.1056/NEJMoa051135 PMID:16354891
  114. Sánchez-Guerrero J, Uribe AG, Jiménez-Santana L, et al. A trial of contraceptive methods in women with systemic lupus erythematosus. 2005;353:2539–49. https://doi.org/10.1056/NEJMoa050817 PMID:16354890
  115. Sarabi ZS, Chang E, Bobba R, et al. Incidence rates of arterial and venous thrombosis after diagnosis of systemic lupus erythematosus. 2005;53:609–12. https://doi.org/10.1002/art.21314 PMID:16082635
  116. Schaedel ZE, Dolan G, Powell MC. The use of the levonorgestrel-releasing intrauterine system in the management of menorrhagia in women with hemostatic disorders. 2005;193:1361–3. https://doi.org/10.1016/j.ajog.2005.05.002 PMID:16202726
  117. Somers E, Magder LS, Petri M. Antiphospholipid antibodies and incidence of venous thrombosis in a cohort of patients with systemic lupus erythematosus. 2002;29:2531–6. PMID:12465147
  118. Urowitz MB, Bookman AA, Koehler BE, Gordon DA, Smythe HA, Ogryzlo MA. The bimodal mortality pattern of systemic lupus erythematosus. 1976;60:221–5. https://doi.org/10.1016/0002-9343(76)90431-9 PMID:1251849
  119. Culwell KR, Curtis KM, Del Carmen Cravioto M. Safety of contraceptive method use among women with systemic lupus erythematosus: a systematic review. 2009;114:341–53. https://doi.org/10.1097/AOG.0b013e3181ae9c64 PMID:19622996
  120. Choojitarom K, Verasertniyom O, Totemchokchyakarn K, Nantiruj K, Sumethkul V, Janwityanujit S. Lupus nephritis and Raynaud’s phenomenon are significant risk factors for vascular thrombosis in SLE patients with positive antiphospholipid antibodies. 2008;27:345–51. https://doi.org/10.1007/s10067-007-0721-z PMID:17805483
  121. Wahl DG, Guillemin F, de Maistre E, Perret C, Lecompte T, Thibaut G. Risk for venous thrombosis related to antiphospholipid antibodies in systemic lupus erythematosus—a meta-analysis. 1997;6:467–73. https://doi.org/10.1177/096120339700600510 PMID:9229367
  122. Farr SL, Folger SG, Paulen ME, Curtis KM. Safety of contraceptive methods for women with rheumatoid arthritis: a systematic review. 2010;82:64–71. https://doi.org/10.1016/j.contraception.2010.02.003 PMID:20682144
  123. Tepper NK, Whiteman MK, Zapata LB, Marchbanks PA, Curtis KM. Safety of hormonal contraceptives among women with migraine: a systematic review. 2016;94:630–40. https://doi.org/10.1016/j.contraception.2016.04.016 PMID:27153744
  124. Tepper NK, Whiteman MK, Marchbanks PA, James AH, Curtis KM. Progestin-only contraception and thromboembolism: a systematic review. 2016;94:678–700. https://doi.org/10.1016/j.contraception.2016.04.014 PMID:27153743
  125. Headache Classification Committee of the International Headache Society (IHS) The International Classification of Headache Disorders. 3rd edition. Cephalalgia 2018;38:1–211. https://www.ichd-3.org/wp-content/uploads/2018/01/The-International-Classification-of-Headache-Disorders-3rd-Edition-2018.pdf
  126. Zapata LB, Oduyebo T, Whiteman MK, Houtchens MK, Marchbanks PA, Curtis KM. Contraceptive use among women with multiple sclerosis: a systematic review. 2016;94:612–20. https://doi.org/10.1016/j.contraception.2016.07.013 PMID:27452316
  127. Pagano HP, Zapata LB, Berry-Bibee EN, Nanda K, Curtis KM. Safety of hormonal contraception and intrauterine devices among women with depressive and bipolar disorders: a systematic review. 2016;94:641–9. https://doi.org/10.1016/j.contraception.2016.06.012 PMID:27364100
  128. Smith JS, Green J, Berrington de Gonzalez A, et al. Cervical cancer and use of hormonal contraceptives: a systematic review. 2003;361:1159–67. https://doi.org/10.1016/S0140-6736(03)12949-2 PMID:12686037
  129. Ahmed K, Baeten JM, Beksinska M, et al. ; Evidence for Contraceptive Options and HIV Outcomes (ECHO) Trial Consortium. HIV incidence among women using intramuscular depot medroxyprogesterone acetate, a copper intrauterine device, or a levonorgestrel implant for contraception: a randomized, multicentre, open-label trial. 2019;394:303–13. https://doi.org/10.1016/S0140-6736(19)31288-7 PMID:31204114
  130. Curtis KM, Hannaford PC, Rodriguez MI, Chipato T, Steyn PS, Kiarie JN. Hormonal contraception and HIV acquisition among women: an updated systematic review. 2020;46:8–16. https://doi.org/10.1136/bmjsrh-2019-200509 PMID:31919239
  131. Tepper NK, Curtis KM, Cox S, Whiteman MK. Update to U.S. medical eligibility criteria for contraceptive use, 2016: updated recommendations for the use of contraception among women at high risk for HIV infection. 2020;69:405–10. https://doi.org/10.15585/mmwr.mm6914a3 PMID:32271729
  132. Phillips SJ, Curtis KM, Polis CB. Effect of hormonal contraceptive methods on HIV disease progression: a systematic review. 2013;27:787–94. https://doi.org/10.1097/QAD.0b013e32835bb672 PMID:23135169
  133. Polis CB, Phillips SJ, Curtis KM. Hormonal contraceptive use and female-to-male HIV transmission: a systematic review of the epidemiologic evidence. 2013;27:493–505. https://doi.org/10.1097/QAD.0b013e32835ad539 PMID:23079808
  134. Phillips SJ, Polis CB, Curtis KM. The safety of hormonal contraceptives for women living with HIV and their sexual partners. 2016;93:11–6. https://doi.org/10.1016/j.contraception.2015.10.002 PMID:26515194
  135. Tagy AH, Saker ME, Moussa AA, Kolgah A. The effect of low-dose combined oral contraceptive pills versus injectable contraceptive (Depot Provera) on liver function tests of women with compensated bilharzial liver fibrosis. 2001;64:173–6. https://doi.org/10.1016/S0010-7824(01)00248-7 PMID:11704097
  136. Pyörälä T, Vähäpassi J, Huhtala M. The effect of lynestrenol and norethindrone on the carbohydrate and lipid metabolism in subjects with gestational diabetes. 1979;68:69–74. PMID:507743
  137. Rådberg T, Gustafson A, Skryten A, Karlsson K. Metabolic studies in gestational diabetic women during contraceptive treatment: effects on glucose tolerance and fatty acid composition of serum lipids. 1982;13:17–29. https://doi.org/10.1159/000299480 PMID:7035304
  138. Kjos SL, Peters RK, Xiang A, Thomas D, Schaefer U, Buchanan TA. Contraception and the risk of type 2 diabetes mellitus in Latina women with prior gestational diabetes mellitus. 1998;280:533–8. https://doi.org/10.1001/jama.280.6.533 PMID:9707143
  139. Nelson AL, Le MH, Musherraf Z, Vanberckelaer A. Intermediate-term glucose tolerance in women with a history of gestational diabetes: natural history and potential associations with breastfeeding and contraception. 2008;198:699.e1–8. https://doi.org/10.1016/j.ajog.2008.03.029 PMID:18439553
  140. Xiang AH, Kawakubo M, Buchanan TA, Kjos SL. A longitudinal study of lipids and blood pressure in relation to method of contraception in Latino women with prior gestational diabetes mellitus. 2007;30:1952–8. https://doi.org/10.2337/dc07-0180 PMID:17519432
  141. Xiang AH, Kawakubo M, Kjos SL, Buchanan TA. Long-acting injectable progestin contraception and risk of type 2 diabetes in Latino women with prior gestational diabetes mellitus. 2006;29:613–7. https://doi.org/10.2337/diacare.29.03.06.dc05-1940 PMID:16505515
  142. Diab KM, Zaki MM. Contraception in diabetic women: comparative metabolic study of Norplant, depot medroxyprogesterone acetate, low dose oral contraceptive pill and CuT380A. 2000;26:17–26. https://doi.org/10.1111/j.1447-0756.2000.tb01195.x PMID:10761326
  143. Lunt H, Brown LJ. Self-reported changes in capillary glucose and insulin requirements during the menstrual cycle. 1996;13:525–30. https://doi.org/10.1002/(SICI)1096-9136(199606)13:6<525::AID-DIA123>3.0.CO;2-D PMID:8799655
  144. Rådberg T, Gustafson A, Skryten A, Karlsson K. Oral contraception in diabetic women. A cross-over study on serum and high density lipoprotein (HDL) lipids and diabetes control during progestogen and combined estrogen/progestogen contraception. 1982;14:61–5. PMID:7040192
  145. Skouby SO, Mølsted-Pedersen L, Kühl C, Bennet P. Oral contraceptives in diabetic women: metabolic effects of four compounds with different estrogen/progestogen profiles. 1986;46:858–64. https://doi.org/10.1016/S0015-0282(16)49825-0 PMID:3781003
  146. Zapata LB, Paulen ME, Cansino C, Marchbanks PA, Curtis KM. Contraceptive use among women with inflammatory bowel disease: a systematic review. 2010;82:72–85. https://doi.org/10.1016/j.contraception.2010.02.012 PMID:20682145
  147. Whiteman MK, Oduyebo T, Zapata LB, Walker S, Curtis KM. Contraceptive safety among women with cystic fibrosis: a systematic review. 2016;94:621–9. https://doi.org/10.1016/j.contraception.2016.05.016 PMID:27287694
  148. Brunson A, Keegan T, Mahajan A, White R, Wun T. High incidence of venous thromboembolism recurrence in patients with sickle cell disease. 2019;94:862–70. https://doi.org/10.1002/ajh.25508 PMID:31074115
  149. Naik RP, Streiff MB, Haywood C Jr, Segal JB, Lanzkron S. Venous thromboembolism incidence in the Cooperative Study of Sickle Cell Disease. 2014;12:2010–6. https://doi.org/10.1111/jth.12744 PMID:25280124
  150. Noubiap JJ, Temgoua MN, Tankeu R, Tochie JN, Wonkam A, Bigna JJ. Sickle cell disease, sickle trait and the risk for venous thromboembolism: a systematic review and meta-analysis. 2018;16:27. https://doi.org/10.1186/s12959-018-0179-z PMID:30305805
  151. Ohene-Frempong K, Weiner SJ, Sleeper LA, et al. Cerebrovascular accidents in sickle cell disease: rates and risk factors. 1998;91:288–94. PMID:9414296
  152. Anastasilakis AD, Tsourdi E, Makras P, et al. Bone disease following solid organ transplantation: a narrative review and recommendations for management from The European Calcified Tissue Society. 2019;127:401–18. https://doi.org/10.1016/j.bone.2019.07.006 PMID:31299385
  153. Panel on Treatment of HIV During Pregnancy and Prevention of Perinatal Transmission. Recommendations for the use of antiretroviral drugs during pregnancy and interventions to reduce perinatal HIV transmission in the United States. Washington, DC: US Department of Health and Human Services; 2023. https://clinicalinfo.hiv.gov/en/guidelines/perinatal/recommendations-arv-drugs-pregnancy-overview
  154. Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of antiretroviral agents in adults and adolescents with HIV. Washington, DC: US Department of Health and Human Services; 2023. https://clinicalinfo.hiv.gov/sites/default/files/guidelines/documents/adult-adolescent-arv/guidelines-adult-adolescent-arv.pdf
  155. Aweeka FT, Rosenkranz SL, Segal Y, et al. ; NIAID AIDS Clinical Trials Group. The impact of sex and contraceptive therapy on the plasma and intracellular pharmacokinetics of zidovudine. 2006;20:1833–41. https://doi.org/10.1097/01.aids.0000244202.18629.36 PMID:16954724
  156. Kearney BP, Mathias A. Lack of effect of tenofovir disoproxil fumarate on pharmacokinetics of hormonal contraceptives. 2009;29:924–9. https://doi.org/10.1592/phco.29.8.924 PMID:19637945
  157. Todd CS, Deese J, Wang M, et al. ; FEM-PrEP Study Group. Sino-implant (II) continuation and effect of concomitant tenofovir disoproxil fumarate-emtricitabine use on plasma levonorgestrel concentrations among women in Bondo, Kenya. 2015;91:248–52. https://doi.org/10.1016/j.contraception.2014.10.008 PMID:25459097
  158. Murnane PM, Heffron R, Ronald A, et al. ; Partners PrEP Study Team. Pre-exposure prophylaxis for HIV-1 prevention does not diminish the pregnancy prevention effectiveness of hormonal contraception. 2014;28:1825–30. https://doi.org/10.1097/QAD.0000000000000290 PMID:24785951
  159. Kasonde M, Niska RW, Rose C, et al. Bone mineral density changes among HIV-uninfected young adults in a randomised trial of pre-exposure prophylaxis with tenofovir-emtricitabine or placebo in Botswana. 2014;9:e90111. https://doi.org/10.1371/journal.pone.0090111 PMID:24625530
  160. Callahan R, Nanda K, Kapiga S, et al. ; FEM-PrEP Study Group. Pregnancy and contraceptive use among women participating in the FEM-PrEP trial. 2015;68:196–203. https://doi.org/10.1097/QAI.0000000000000413 PMID:25590272
  161. Vieira CS, Bahamondes MV, de Souza RM, et al. Effect of antiretroviral therapy including lopinavir/ritonavir or efavirenz on etonogestrel-releasing implant pharmacokinetics in HIV-positive women. 2014;66:378–85. https://doi.org/10.1097/QAI.0000000000000189 PMID:24798768
  162. Patel RC, Onono M, Gandhi M, et al. Pregnancy rates in HIV-positive women using contraceptives and efavirenz-based or nevirapine-based antiretroviral therapy in Kenya: a retrospective cohort study. 2015;2:e474–82. https://doi.org/10.1016/S2352-3018(15)00184-8 PMID:26520927
  163. Perry SH, Swamy P, Preidis GA, Mwanyumba A, Motsa N, Sarero HN. Implementing the Jadelle implant for women living with HIV in a resource-limited setting: concerns for drug interactions leading to unintended pregnancies. 2014;28:791–3. https://doi.org/10.1097/QAD.0000000000000177 PMID:24401645
  164. Scarsi KK, Darin KM, Nakalema S, et al. Unintended pregnancies observed with combined use of the levonorgestrel contraceptive implant and efavirenz-based antiretroviral therapy: a three-arm pharmacokinetic evaluation over 48 weeks. 2016;62:675–82. https://doi.org/10.1093/cid/civ1001 PMID:26646680
  165. Pyra M, Heffron R, Mugo NR, et al. ; Partners in Prevention HSVHIV Transmission Study and Partners PrEP Study Teams. Effectiveness of hormonal contraception in HIV-infected women using antiretroviral therapy. 2015;29:2353–9. https://doi.org/10.1097/QAD.0000000000000827 PMID:26544706
  166. Cohn SE, Park JG, Watts DH, et al. ; ACTG A5093 Protocol Team. Depo-medroxyprogesterone in women on antiretroviral therapy: effective contraception and lack of clinically significant interactions. 2007;81:222–7. https://doi.org/10.1038/sj.clpt.6100040 PMID:17192768
  167. Nanda K, Amaral E, Hays M, Viscola MA, Mehta N, Bahamondes L. Pharmacokinetic interactions between depot medroxyprogesterone acetate and combination antiretroviral therapy. 2008;90:965–71. https://doi.org/10.1016/j.fertnstert.2007.07.1348 PMID:17880953
  168. Watts DH, Park JG, Cohn SE, et al. Safety and tolerability of depot medroxyprogesterone acetate among HIV-infected women on antiretroviral therapy: ACTG A5093. 2008;77:84–90. https://doi.org/10.1016/j.contraception.2007.10.002 PMID:18226670
  169. Polis CB, Nakigozi G, Ssempijja V, et al. Effect of injectable contraceptive use on response to antiretroviral therapy among women in Rakai, Uganda. 2012;86:725–30. https://doi.org/10.1016/j.contraception.2012.05.001 PMID:22717186
  170. Hubacher D, Liku J, Kiarie J, et al. Effect of concurrent use of anti-retroviral therapy and levonorgestrel sub-dermal implant for contraception on CD4 counts: a prospective cohort study in Kenya. 2013;16:18448. https://doi.org/10.7448/IAS.16.1.18448 PMID:23458102
  171. Myer L, Carter RJ, Katyal M, Toro P, El-Sadr WM, Abrams EJ. Impact of antiretroviral therapy on incidence of pregnancy among HIV-infected women in Sub-Saharan Africa: a cohort study. 2010;7:e1000229. https://doi.org/10.1371/journal.pmed.1000229 PMID:20161723
  172. Day S, Graham SM, Masese LN, et al. A prospective cohort study of the effect of depot medroxyprogesterone acetate on detection of plasma and cervical HIV-1 in women initiating and continuing antiretroviral therapy. 2014;66:452–6. https://doi.org/10.1097/QAI.0000000000000187 PMID:24798764
  173. DuBois BN, Atrio J, Stanczyk FZ, Cherala G. Increased exposure of norethindrone in HIV+ women treated with ritonavir-boosted atazanavir therapy. 2015;91:71–5. https://doi.org/10.1016/j.contraception.2014.08.009 PMID:25245190
  174. Luque AE, Cohn SE, Park JG, et al. Depot medroxyprogesterone acetate in combination with a twice-daily lopinavir-ritonavir-based regimen in HIV-infected women showed effective contraception and a lack of clinically significant interactions, with good safety and tolerability: results of the ACTG 5283 study. 2015;59:2094–101. https://doi.org/10.1128/AAC.04701-14 PMID:25624326
  175. Odlind V, Olsson SE. Enhanced metabolism of levonorgestrel during phenytoin treatment in a woman with Norplant implants. 1986;33:257–61. https://doi.org/10.1016/0010-7824(86)90018-1 PMID:3087695
  176. Schindlbeck C, Janni W, Friese K. Failure of Implanon contraception in a patient taking carbamazepin for epilepsia. 2006;273:255–6. https://doi.org/10.1007/s00404-005-0064-4 PMID:16208481
  177. Shane-McWhorter L, Cerveny JD, MacFarlane LL, Osborn C. Enhanced metabolism of levonorgestrel during phenobarbital treatment and resultant pregnancy. 1998;18:1360–4. https://doi.org/10.1002/j.1875-9114.1998.tb03161.x PMID:9855340
  178. Gaffield ME, Culwell KR, Lee CR. The use of hormonal contraception among women taking anticonvulsant therapy. 2011;83:16–29. https://doi.org/10.1016/j.contraception.2010.06.013 PMID:21134499
  179. Reimers A, Helde G, Brodtkorb E. Ethinyl estradiol, not progestogens, reduces lamotrigine serum concentrations. 2005;46:1414–7. https://doi.org/10.1111/j.1528-1167.2005.10105.x PMID:16146436
  180. Berry-Bibee ENKM, Kim MJ, Simmons KB, et al. Drug interactions between hormonal contraceptives and psychotropic drugs: a systematic review. 2016;94:650–67. https://doi.org/10.1016/j.contraception.2016.07.011 PMID:27444984
  181. Berry-Bibee ENKM, Kim MJ, Tepper NK, Riley HE, Curtis KM. Co-administration of St. John’s wort and hormonal contraceptives: a systematic review. 2016;94:668–77. https://doi.org/10.1016/j.contraception.2016.07.010 PMID:27444983

Appendix D: Classifications for Combined Hormonal Contraceptives

Combined hormonal contraceptives (CHCs) include combined oral contraceptives (COCs) (containing a progestin plus ethinyl estradiol [EE] ≤35 μg, estradiol valerate, or estetrol); combined transdermal patches (levonorgestrel/EE or norelgestromin/EE); and combined vaginal rings (etonogestrel/EE or segesterone acetate/EE) (Box D1) (Table D1). Limited information is available about the safety of COCs with estradiol valerate or estetrol, combined transdermal patches, and combined vaginal rings among users with specific medical conditions. Evidence indicates that estradiol valerate and estetrol COCs, combined transdermal patches, and combined vaginal rings provide comparable safety and pharmacokinetic profiles to EE-containing COCs with similar hormone formulations (133). Pending further studies, the evidence available for recommendations about EE-containing COCs applies to the recommendations for estradiol valerate and estetrol COCs, the combined transdermal patch, and vaginal rings. Therefore, the estradiol valerate and estetrol COCs, the patches, and the rings should have the same categories as EE-containing COCs, except where noted. The assigned categories should be considered a preliminary best judgment, which will be reevaluated as new data become available.

COCs, patches, and rings do not protect against sexually transmitted infections (STIs), including HIV infection, and patients using CHCs should be counseled that consistent and correct use of external (male) latex condoms reduces the risk for STIs, including HIV infection (34). Use of internal (female) condoms can provide protection from transmission of STIs, although data are limited (34). Patients also should be counseled that pre-exposure prophylaxis, when taken as prescribed, is highly effective for preventing HIV infection (35).

Return to your place in the textBOX D1. Categories for classifying combined hormonal contraceptives

U.S. MEC 1 = A condition for which there is no restriction for the use of the contraceptive method

U.S. MEC 2 = A condition for which the advantages of using the method generally outweigh the theoretical or proven risks

U.S. MEC 3 = A condition for which the theoretical or proven risks usually outweigh the advantages of using the method

U.S. MEC 4 = A condition that represents an unacceptable health risk if the contraceptive method is used

Abbreviation: U.S. MEC = U.S. Medical Eligibility Criteria for Contraceptive Use.



TABLE D1. Classifications for combined hormonal contraceptives, including pill, patch, and ring
Condition CHC Clarification/Evidence/Comment
Personal Characteristics and Reproductive History
Pregnancy NA Clarification: Use of CHCs is not required. No known harm to the patient, the course of pregnancy, or the fetus occurs if CHCs are inadvertently used during pregnancy.
Age Evidence: Evidence is inconsistent about whether CHC use affects fracture risk (3647), although three recent studies demonstrate no effect (36,37,47). CHC use might decrease BMD in adolescents, especially in those choosing very low-dose formulations (COCs containing <30 µg ethinyl estradiol) (4861). CHC use has little to no effect on BMD in premenopausal women (6276) and might preserve bone mass in those who are perimenopausal (7785). BMD is a surrogate marker for fracture risk that might not be valid for premenopausal women and therefore might not accurately predict current or future (postmenopausal) fracture risk (8688).
Comment: The risk for cardiovascular disease increases with age and might increase with CHC use. In the absence of other adverse clinical conditions, CHCs can be used until menopause.
a. Menarche to <40 years 1
b. ≥40 years 2
Parity
a. Nulliparous 1
b. Parous 1
Breastfeeding
a. <21 days postpartum 4 Clarification (breastfeeding): Breastfeeding provides important health benefits for breastfeeding parent and infant. The U.S. Dietary Guidelines for Americans and American Academy of Pediatrics recommend that infants be exclusively breastfed for about the first 6 months with continued breastfeeding while introducing appropriate complementary foods for 1 year or longer (89) or up to age 2 years or longer (90).
Evidence (breastfeeding): Clinical studies demonstrate conflicting results regarding effects on breastfeeding continuation or exclusivity in women exposed to COCs during lactation. No consistent effects on infant growth or illness have been reported. Adverse health outcomes or manifestations of exogenous estrogen in infants exposed to CHCs through breast milk have not been demonstrated; however, studies have been inadequately designed to determine whether a risk for either serious or subtle long-term effects exists (91).
Evidence: One study examined use of CHCs during the postpartum period and found that VTE rates were higher for CHC users compared with nonusers at all time points postpartum (92). Rates were significantly different only after 13 weeks postpartum; however, the numbers needed to harm were lowest in the first 6 weeks postpartum. VTE risk is increased during pregnancy and the postpartum period; this risk is most pronounced in the first 3 weeks after delivery, decreasing to near baseline levels by 42 days postpartum (9397).
Comment: Risk factors for breastfeeding difficulties include previous breastfeeding difficulties, certain medical conditions, certain perinatal complications, and preterm birth. For all breastfeeding persons, with or without risk factors for breastfeeding difficulties, discussions about contraception should include information about risks, benefits, and alternatives.
b. 21 to <30 days postpartum
   i. With other risk factors for VTE (e.g., age ≥35 years, previous VTE, thrombophilia, immobility, transfusion at delivery, peripartum cardiomyopathy, BMI ≥30 kg/m2, postpartum hemorrhage, postcesarean delivery, preeclampsia, or smoking) 3 Clarification: For persons with other risk factors for VTE, these risk factors might increase the classification to a category 4.
Clarification (breastfeeding): Breastfeeding provides important health benefits for breastfeeding parent and infant. The U.S. Dietary Guidelines for Americans and American Academy of Pediatrics recommend that infants be exclusively breastfed for about the first 6 months with continued breastfeeding while introducing appropriate complementary foods for 1 year or longer (89) or up to age 2 years or longer (90).
Evidence (breastfeeding): Clinical studies demonstrate conflicting results regarding effects on breastfeeding continuation or exclusivity in women exposed to COCs during lactation. No consistent effects on infant growth or illness have been reported. Adverse health outcomes or manifestations of exogenous estrogen in infants exposed to CHCs through breast milk have not been demonstrated; however, studies have been inadequately designed to determine whether a risk for either serious or subtle long-term effects exists (91).
Evidence: One study examined use of CHCs during the postpartum period and found that VTE rates were higher for CHC users compared with nonusers at all time points postpartum (92). Rates were significantly different only after 13 weeks postpartum; however, the numbers needed to harm were lowest in the first 6 weeks postpartum. VTE risk is increased during pregnancy and the postpartum period; this risk is most pronounced in the first 3 weeks after delivery, decreasing to near baseline levels by 42 days postpartum (9397).
Comment: Risk factors for breastfeeding difficulties include previous breastfeeding difficulties, certain medical conditions, certain perinatal complications, and preterm birth. For all breastfeeding persons, with or without risk factors for breastfeeding difficulties, discussions about contraception should include information about risks, benefits, and alternatives.
   ii. Without other risk factors for VTE 3 Clarification (breastfeeding): Breastfeeding provides important health benefits for breastfeeding parent and infant. The U.S. Dietary Guidelines for Americans and American Academy of Pediatrics recommend that infants be exclusively breastfed for about the first 6 months with continued breastfeeding while introducing appropriate complementary foods for 1 year or longer (89) or up to age 2 years or longer (90).
Evidence (breastfeeding): Clinical studies demonstrate conflicting results regarding effects on breastfeeding continuation or exclusivity in women exposed to COCs during lactation. No consistent effects on infant growth or illness have been reported. Adverse health outcomes or manifestations of exogenous estrogen in infants exposed to CHCs through breast milk have not been demonstrated; however, studies have been inadequately designed to determine whether a risk for either serious or subtle long-term effects exists (91).
Evidence: One study examined use of CHCs during the postpartum period and found that VTE rates were higher for CHC users compared with nonusers at all time points postpartum (92). Rates were significantly different only after 13 weeks postpartum; however, the numbers needed to harm were lowest in the first 6 weeks postpartum. VTE risk is increased during pregnancy and the postpartum period; this risk is most pronounced in the first 3 weeks after delivery, decreasing to near baseline levels by 42 days postpartum (9397).
Comment: Risk factors for breastfeeding difficulties include previous breastfeeding difficulties, certain medical conditions, certain perinatal complications, and preterm birth. For all breastfeeding persons, with or without breastfeeding difficulties, discussions about contraception should include information about risks, benefits, and alternatives.
c. 30–42 days postpartum
   i. With other risk factors for VTE (e.g., age ≥35 years, previous VTE, thrombophilia, immobility, transfusion at delivery, peripartum cardiomyopathy, BMI ≥30 kg/m2, postpartum hemorrhage, postcesarean delivery, preeclampsia, or smoking) 3 Clarification: For persons with other risk factors for VTE, these risk factors might increase the classification to a category 4.
Clarification (breastfeeding): Breastfeeding provides important health benefits for breastfeeding parent and infant. The U.S. Dietary Guidelines for Americans and American Academy of Pediatrics recommend that infants be exclusively breastfed for about the first 6 months with continued breastfeeding while introducing appropriate complementary foods for 1 year or longer (89) or up to age 2 years or longer (90).
Evidence (breastfeeding): Clinical studies demonstrate conflicting results regarding effects on breastfeeding continuation or exclusivity in women exposed to COCs during lactation. No consistent effects on infant growth or illness have been reported. Adverse health outcomes or manifestations of exogenous estrogen in infants exposed to CHCs through breast milk have not been demonstrated; however, studies have been inadequately designed to determine whether a risk for either serious or subtle long-term effects exists (91).
Evidence: One study examined use of CHCs during the postpartum period and found that VTE rates were higher for CHC users compared with nonusers at all time points postpartum (92). Rates were significantly different only after 13 weeks postpartum; however, the numbers needed to harm were lowest in the first 6 weeks postpartum. VTE risk is increased during pregnancy and the postpartum period; this risk is most pronounced in the first 3 weeks after delivery, decreasing to near baseline levels by 42 days postpartum (9397).
Comment: Risk factors for breastfeeding difficulties include previous breastfeeding difficulties, certain medical conditions, certain perinatal complications, and preterm birth. For all breastfeeding persons, with or without breastfeeding difficulties, discussions about contraception should include information about risks, benefits, and alternatives.
   ii. Without other risk factors for VTE 2 Clarification (breastfeeding): Breastfeeding provides important health benefits for breastfeeding parent and infant. The U.S. Dietary Guidelines for Americans and American Academy of Pediatrics recommend that infants be exclusively breastfed for about the first 6 months with continued breastfeeding while introducing appropriate complementary foods for 1 year or longer (89) or up to age 2 years or longer (90).
Evidence (breastfeeding): Clinical studies demonstrate conflicting results regarding effects on breastfeeding continuation or exclusivity in women exposed to COCs during lactation. No consistent effects on infant growth or illness have been reported. Adverse health outcomes or manifestations of exogenous estrogen in infants exposed to CHCs through breast milk have not been demonstrated; however, studies have been inadequately designed to determine whether a risk for either serious or subtle long-term effects exists (91).
Evidence: One study examined use of CHCs during the postpartum period and found that VTE rates were higher for CHC users compared with nonusers at all time points postpartum (92). Rates were significantly different only after 13 weeks postpartum; however, the numbers needed to harm were lowest in the first 6 weeks postpartum. VTE risk is increased during pregnancy and the postpartum period; this risk is most pronounced in the first 3 weeks after delivery, decreasing to near baseline levels by 42 days postpartum (9397).
Comment: Risk factors for breastfeeding difficulties include previous breastfeeding difficulties, certain medical conditions, certain perinatal complications, and preterm birth. For all breastfeeding persons, with or without breastfeeding difficulties, discussions about contraception should include information about risks, benefits, and alternatives.
d. >42 days postpartum 2 Clarification (breastfeeding): Breastfeeding provides important health benefits for breastfeeding parent and infant. The U.S. Dietary Guidelines for Americans and American Academy of Pediatrics recommend that infants be exclusively breastfed for about the first 6 months with continued breastfeeding while introducing appropriate complementary foods for 1 year or longer (89) or up to age 2 years or longer (90).
Evidence: Clinical studies demonstrate conflicting results regarding effects on breastfeeding continuation or exclusivity in women exposed to COCs during lactation. No consistent effects on infant growth or illness have been reported. Adverse health outcomes or manifestations of exogenous estrogen in infants exposed to CHCs through breast milk have not been demonstrated; however, studies have been inadequately designed to determine whether a risk for either serious or subtle long-term effects exists (91).
Comment: Risk factors for breastfeeding difficulties include previous breastfeeding difficulties, certain medical conditions, certain perinatal complications, and preterm birth. For all breastfeeding persons, with or without breastfeeding difficulties, discussions about contraception should include information about risks, benefits, and alternatives.
Postpartum (nonbreastfeeding)
a. <21 days postpartum 4 Evidence: One study examined use of CHCs during the postpartum period and found that VTE rates were higher for CHC users compared with nonusers at all time points postpartum (92). Rates were significantly different only after 13 weeks postpartum; however, the numbers needed to harm were lowest in the first 6 weeks postpartum. VTE risk is increased during pregnancy and the postpartum period; this risk is most pronounced in the first 3 weeks after delivery, decreasing to near baseline levels by 42 days postpartum (9397). Risk for pregnancy during the first 21 days postpartum is very low but increases after that point; ovulation before first menses is common (98).
b. 21–42 days postpartum
   i. With other risk factors for VTE (e.g., age ≥35 years, previous VTE, thrombophilia, immobility, transfusion at delivery, peripartum cardiomyopathy, BMI ≥30 kg/m2 postpartum hemorrhage, postcesarean delivery, preeclampsia, or smoking) 3 Clarification: For persons with other risk factors for VTE, these risk factors might increase the classification to a category 4.
Evidence: One study examined use of CHCs during the postpartum period and found that VTE rates were higher for CHC users compared with nonusers at all time points postpartum (92). Rates were significantly different only after 13 weeks postpartum; however, the numbers needed to harm were lowest in the first 6 weeks postpartum. VTE risk is increased during pregnancy and the postpartum period; this risk is most pronounced in the first 3 weeks after delivery, decreasing to near baseline levels by 42 days postpartum (9397).
   ii. Without other risk factors for VTE 2 Evidence: One study examined use of CHCs during the postpartum period and found that VTE rates were higher for CHC users compared with nonusers at all time points postpartum (92). Rates were significantly different only after 13 weeks postpartum; however, the numbers needed to harm were lowest in the first 6 weeks postpartum. VTE risk is increased during pregnancy and the postpartum period; this risk is most pronounced in the first 3 weeks after delivery, decreasing to near baseline levels by 42 days postpartum (9397).
c. >42 days postpartum 1
Postabortion (spontaneous or induced)
a. First trimester abortion Clarification: CHCs may be started immediately after abortion completion or at time of medication abortion initiation.
Evidence: Evidence suggests that there is no increased risk for adverse events when CHCs are initiated after first trimester procedural or medication abortion (immediately or delayed) (99). Immediate initiation of COCs after first trimester procedural or medication abortion did not cause clinically significant changes in coagulation parameters compared with placebo, a hormonal IUD, a nonhormonal contraceptive method, or delayed COC initiation (100).
   i. Procedural (surgical) 1
   ii. Medication 1
   iii. Spontaneous abortion with no intervention 1
b. Second trimester abortion Clarification: CHCs may be started immediately after abortion completion or at time of medication abortion initiation.
Evidence: Limited evidence suggests that there is no increased risk for adverse events when CHCs are initiated after second trimester procedural abortion (immediately or delayed) (99).
   i. Procedural (surgical) 1
   ii. Medication 1
   iii. Spontaneous abortion with no intervention 1
c. Immediate postseptic abortion 1 Clarification: CHCs may be started immediately after abortion completion or at time of medication abortion initiation.
Past ectopic pregnancy 1 Comment: The risk for future ectopic pregnancy is increased among those who have had an ectopic pregnancy in the past. CHCs protect against pregnancy in general, including ectopic gestation.
History of pelvic surgery 1
Smoking Evidence: COC users who smoked were at increased risk for cardiovascular diseases, especially myocardial infarction, compared with those who did not smoke. Studies also demonstrated an increased risk for myocardial infarction with increasing number of cigarettes smoked per day (101113).
a. Age <35 years 2
b. Age ≥35 years
i. <15 cigarettes per day 3
ii. ≥15 cigarettes per day 4
Obesity Clarification: Risk for thrombosis increases with multiple risk factors, such as obesity, older age (e.g., ≥40 years), diabetes, smoking, family history of thrombosis, and dyslipidemia. When a person has multiple risk factors, any of which alone would increase risk for thrombosis, use of CHCs might increase thrombosis risk to an unacceptable level. However, a simple addition of categories for multiple risk factors is not intended; for example, a combination of two category 2 risk factors might not necessarily warrant a higher category.
Evidence: Although the absolute risk for VTE in healthy women of reproductive age is small, COC use and higher BMI independently increase risk for VTE, with the greatest relative risks among those with both risk factors. From a systematic review, COC users with obesity consistently had a relative risk for VTE of 5–8 times that of nonusers with obesity (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516). Research examining the interaction between COCs and BMI on VTE risk is limited, particularly for those in the highest BMI categories (BMI ≥35 kg/m2). Comparative studies on the risk for VTE among contraceptive patch or ring users by weight or BMI were not identified (114116).
Limited evidence suggests that COC users with obesity do not have a higher risk for acute myocardial infarction or stroke than do nonusers with obesity (114) (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
Limited evidence suggests that effectiveness of certain COC formulations might decrease with increasing BMI; however the observed reductions in effectiveness are minimal and evidence is conflicting (117124). Effectiveness of the patch might be reduced in women with BMI ≥30 kg/m2 or weight >90 kg (125).
a. BMI ≥30 kg/m2 2
b. Menarche to <18 years and BMI ≥30 kg/m2 2
History of bariatric surgery
This condition is associated with
increased risk for adverse health events
as a result of pregnancy (Box 3).
a. Restrictive procedures: decrease storage capacity of the stomach (vertical banded gastroplasty, laparoscopic adjustable gastric band, or laparoscopic sleeve gastrectomy) 1 Evidence: Limited evidence demonstrated no substantial decrease in effectiveness of oral contraceptives among women who underwent laparoscopic placement of an adjustable gastric band (126).
b. Malabsorptive procedures: decrease absorption of nutrients and calories by shortening the functional length of the small intestine (Roux-en-Y gastric bypass or biliopancreatic diversion) COCs: 3
Patch and ring: 1
Evidence: Limited evidence demonstrated no substantial decrease in effectiveness of oral contraceptives among women who underwent a biliopancreatic diversion; however, evidence from pharmacokinetic studies reported conflicting results of oral contraceptive effectiveness among women who underwent a jejunoileal bypass (126).
Comment: Bariatric surgical procedures involving a malabsorptive component have the potential to decrease oral contraceptive effectiveness, perhaps further decreased by postoperative complications, such as long-term diarrhea or vomiting.
Surgery
a. Minor surgery without immobilization 1
b. Major surgery
   ii. Without prolonged immobilization 2
   i. With prolonged immobilization 4
Cardiovascular Disease
Multiple risk factors for atherosclerotic cardiovascular disease (e.g., older age, smoking, diabetes, hypertension, low HDL, high LDL, or high triglyceride levels) 3/4 Clarification: When a person has multiple major risk factors, any of which alone would substantially increase risk for cardiovascular disease, use of CHCs might increase risk to an unacceptable level. However, a simple addition of categories for multiple risk factors is not intended; for example, a combination of two category 2 risk factors might not necessarily warrant a higher category.
Clarification: The recommendations apply to known pre-existing medical conditions or characteristics. Few if any screening tests are needed before initiation of contraception. See U.S. SPR (https://www.cdc.gov/contraception/hcp/usspr/HYPERLINK) (127).
Hypertension
Systolic blood pressure ≥160 mm Hg or
diastolic blood pressure ≥100 mm Hg are
associated with increased risk for adverse
health events as a result of pregnancy
(Box 3).
a. Adequately controlled hypertension 3 Clarification: For all categories of hypertension, classifications are based on the assumption that no other risk factors exist for cardiovascular disease. When multiple risk factors do exist, risk for cardiovascular disease might increase substantially. A single reading of blood pressure level is not sufficient to classify a person as hypertensive.
Clarification: Persons adequately treated for hypertension are at reduced risk for acute myocardial infarction and stroke compared with untreated persons. Although no data exist, CHC users with adequately controlled and monitored hypertension should be at reduced risk for acute myocardial infarction and stroke compared with untreated hypertensive CHC users.
Evidence: Among women with hypertension, COC users were at higher risk than nonusers for stroke, acute myocardial infarction, and peripheral arterial disease (101,103,110113,128142). Discontinuation of COCs in women with hypertension might improve blood pressure control (143).
b. Elevated blood pressure levels
(properly taken measurements)
Clarification: For all categories of hypertension, classifications are based on the assumption that no other risk factors exist for cardiovascular disease. When multiple risk factors do exist, risk for cardiovascular disease might increase substantially. A single reading of blood pressure level is not sufficient to classify a person as hypertensive.
Evidence: Among women with hypertension, COC users were at higher risk than nonusers for stroke, acute myocardial infarction, and peripheral arterial disease (101,103,110113,128142). Discontinuation of COCs in women with hypertension might improve blood pressure control (143).
   i. Systolic 140–159 mm Hg or diastolic 90–99 mm Hg 3
   ii. Systolic ≥160 mm Hg or diastolic ≥100 mm Hg 4
c. Vascular disease 4
History of high blood pressure during pregnancy (when current blood pressure is measurable and normal) 2 Evidence: Women with a history of high blood pressure in pregnancy who also used COCs had a higher risk for myocardial infarction and VTE than did COC users who did not have a history of high blood pressure during pregnancy. The absolute risks for acute myocardial infarction and VTE in this population remained small (112,129,141,142,144150).
Deep venous thrombosis/
Pulmonary embolism

This condition is associated with increased
risk for adverse health events as a result of
pregnancy (Box 3).
a. Current or history of DVT/PE, receiving anticoagulant therapy (therapeutic dose) (e.g., acute DVT/PE or long-term therapeutic dose) 3 Clarification: Persons using anticoagulant therapy are at risk for gynecologic complications of therapy, such as heavy or prolonged bleeding and hemorrhagic ovarian cysts. CHCs can be of benefit in preventing or treating these complications. When a contraceptive method is used as a therapy, rather than solely to prevent pregnancy, the risk/benefit ratio might differ and should be considered on a case-by-case basis.
Clarification: When a patient discontinues therapeutic dose of anticoagulant therapy, careful consideration should be given to transitioning from CHCs to a progestin-only or nonhormonal method, if acceptable to the patient.
Evidence: Limited evidence was identified on use of CHCs among women with DVT/PE receiving anticoagulant therapy (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516). In one study among women with a history of acute VTE currently receiving therapeutic anticoagulant therapy (i.e., rivaroxaban or enoxaparin/vitamin K antagonist [warfarin or acenocoumarol]), the incidence of recurrent VTE was similar among estrogen users (CHC or estrogen-only pills), POC users, and women not on hormonal therapy (151).
b. History of DVT/PE, receiving
anticoagulant therapy (prophylactic dose)
Clarification: Persons using anticoagulant therapy are at risk for gynecologic complications of therapy, such as heavy or prolonged bleeding and hemorrhagic ovarian cysts. CHCs can be of benefit in preventing or treating these complications. When a contraceptive method is used as a therapy, rather than solely to prevent pregnancy, the risk/benefit ratio might differ and should be considered on a case-by-case basis.
i. Higher risk for recurrent DVT/PE (one or more risk factors) 4
• Thrombophilia (e.g., factor V Leiden mutation; prothrombin gene mutation; protein S, protein C, and antithrombin deficiencies; or antiphospholipid syndrome)
• Active cancer (metastatic, receiving therapy, or within 6 months after clinical remission), excluding nonmelanoma skin cancer
• History of recurrent DVT/PE
   ii. Lower risk for recurrent DVT/PE (no risk factors) 3
c. History of DVT/PE, not receiving
anticoagulant therapy
   i. Higher risk for recurrent DVT/PE (one or more risk factors) 4
• History of estrogen-associated DVT/PE
• Pregnancy-associated DVT/PE
• Idiopathic DVT/PE
• Thrombophilia (e.g., factor V Leiden mutation; prothrombin gene mutation; protein S, protein C, and antithrombin deficiencies; or antiphospholipid syndrome)
• Active cancer (metastatic,
receiving therapy, or within 6 months after clinical remission), excluding nonmelanoma skin
cancer
• History of recurrent DVT/PE
   ii. Lower risk for recurrent DVT/PE (no risk factors) 3
d. Family history (first-degree relatives) 2 Comment: Certain conditions that increase the risk for DVT/PE are heritable.
Thrombophilia (e.g., factor V Leiden mutation; prothrombin gene mutation; protein S, protein C, and antithrombin deficiencies; or antiphospholipid syndrome)
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
4 Clarification: Routine screening in the general population before contraceptive initiation is not recommended.
Clarification: If a person has current or history of DVT/PE, see recommendations for DVT/PE.
Clarification: Classification of antiphospholipid syndrome includes presence of a clinical feature (e.g., thrombosis or obstetric morbidity) and persistently abnormal antiphospholipid antibody test on two or more occasions at least 12 weeks apart (152).
Evidence: Among women with factor V Leiden mutation, prothrombin gene mutation, antithrombin deficiency, and protein C deficiency, COC users had an increased risk for venous and arterial thrombosis compared with nonusers. Evidence was inconsistent on risk for thrombosis among women with protein S deficiency using COCs. No evidence was identified on COC use among persons with antiphospholipid syndrome (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
Superficial venous disorders
a. Varicose veins 1 Evidence: One study suggested that among women with varicose veins, the rate of VTE and superficial venous thrombosis was higher in oral contraceptive users compared with nonusers; however, statistical significance was not reported and the number of events was small (153).
b. Superficial venous thrombosis (acute or history) 3 Clarification: Superficial venous thrombosis might be associated with an increased risk for VTE. If a person has risk factors for concurrent DVT (e.g., thrombophilia or cancer) or has current or history of DVT, see recommendations for DVT/PE. Superficial venous thrombosis associated with a peripheral intravenous catheter is less likely to be associated with additional thrombosis and use of CHCs may be considered.
Evidence: One study demonstrated that among women with superficial venous thrombosis, the risk for VTE was higher in oral contraceptive users compared with nonusers (153).
Current and history of ischemic heart disease
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
4
Stroke (history of cerebrovascular accident)
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
4
Valvular heart disease
Complicated valvular heart disease is
associated with increased risk for adverse
health events as a result of pregnancy
(Box 3).
a. Uncomplicated 2
b. Complicated (pulmonary hypertension, risk for atrial fibrillation, or history of subacute bacterial endocarditis) 4 Comment: Among persons with valvular heart disease, CHC use might further increase the risk for arterial thrombosis; persons with complicated valvular heart disease are at greatest risk.
Peripartum cardiomyopathy
This condition is associated with increased
risk for adverse health events as a result of
pregnancy (Box 3).
Evidence: No direct evidence exists about the safety of CHCs among women with peripartum cardiomyopathy. Limited indirect evidence from noncomparative studies of women with cardiac disease demonstrated few cases of hypertension and transient ischemic attack in women with cardiac disease using COCs. No cases of heart failure were reported (154).
Comment: COCs might increase fluid retention in healthy persons; fluid retention might worsen heart failure in persons with peripartum cardiomyopathy. COCs might induce cardiac arrhythmias in healthy persons; persons with peripartum cardiomyopathy have a high incidence of cardiac arrhythmias.
a. Normal or mildly impaired cardiac
function (New York Heart Association
Functional Class I or II: no limitation of
activities or slight, mild limitation of
activity) (155)
   i. <6 months 4
   ii. ≥6 months 3
b. Moderately or severely impaired cardiac function (New York Heart Association Functional Class III or IV: marked limitation of activity or should be at complete rest) (155) 4
Renal Disease
Chronic kidney disease
This condition is associated with increased
risk for adverse health events as a result of
pregnancy (Box 3).
a. Current nephrotic syndrome 4 Evidence: No direct evidence was identified on CHC use among persons with CKD with current nephrotic syndrome (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516). Persons with severe CKD or nephrotic syndrome are at higher risk for thrombosis than the general population (156158). Use of CHCs might further elevate risk for thrombosis among those with CKD with current nephrotic syndrome.
Comment: A person might have CKD without current nephrotic syndrome but might have other conditions often associated with CKD (e.g., diabetes, hypertension, and SLE). See recommendations for other conditions if they apply.
b. Hemodialysis 4 Evidence: No direct evidence was identified on CHC use among persons with CKD on hemodialysis (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516). Persons with CKD on dialysis are at higher risk for thrombosis than the general population (156158). Use of CHCs might further elevate risk for thrombosis among those with CKD on dialysis.
Comment: A person might have CKD without hemodialysis, but might have other conditions often associated with CKD (e.g., diabetes, hypertension, and SLE). See recommendations for other conditions if they apply.
c. Peritoneal dialysis 4 Evidence: No direct evidence was identified on CHC use among persons with CKD on peritoneal dialysis (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516). Persons with CKD on dialysis are at higher risk for thrombosis than the general population (156158). Use of CHCs might further elevate risk for thrombosis among those with CKD.
Comment: A person might have CKD without peritoneal dialysis, but might have other conditions often associated with CKD (e.g., diabetes, hypertension, and SLE). See recommendations for other conditions if they apply.
Rheumatic Diseases
Systemic lupus erythematosus
This condition is associated with increased
risk for adverse health events as a result of
pregnancy (Box 3).
a. Positive (or unknown) antiphospholipid antibodies 4 Clarification: Persons with SLE are at increased risk for ischemic heart disease, stroke, and VTE. Categories assigned to such conditions in U.S. MEC should be the same for persons with SLE who have these conditions. For all subconditions of SLE, classifications are based on the assumption that no other risk factors for cardiovascular disease are present; these classifications must be modified in the presence of such risk factors (159177).
Evidence: Antiphospholipid antibodies are associated with a higher risk for both arterial and venous thrombosis (178,179).
b. Severe thrombocytopenia 2 Clarification: Persons with SLE are at increased risk for ischemic heart disease, stroke, and VTE. Categories assigned to such conditions in U.S. MEC should be the same for persons with SLE who have these conditions. For all subconditions of SLE, classifications are based on the assumption that no other risk factors for cardiovascular disease are present; these classifications must be modified in the presence of such risk factors (159177).
c. Immunosuppressive therapy 2 Clarification: Persons with SLE are at increased risk for ischemic heart disease, stroke, and VTE. Categories assigned to such conditions in U.S. MEC should be the same for persons with SLE who have these conditions. For all subconditions of SLE, classifications are based on the assumption that no other risk factors for cardiovascular disease are present; these classifications must be modified in the presence of such risk factors (159177).
d. None of the above 2 Clarification: Persons with SLE are at increased risk for ischemic heart disease, stroke, and VTE. Categories assigned to such conditions in U.S. MEC should be the same for persons with SLE who have these conditions. For all subconditions of SLE, classifications are based on the assumption that no other risk factors for cardiovascular disease are present; these classifications must be modified in the presence of such risk factors (159177).
Rheumatoid arthritis Evidence: Limited evidence demonstrates no consistent pattern of improvement or worsening of rheumatoid arthritis with use of oral contraceptives, progesterone, or estrogen (180).
a. Not receiving immunosuppressive therapy 2
b. Receiving immunosuppressive therapy 2
Neurologic Conditions
Headaches
a. Nonmigraine (mild or severe) 1 Clarification: Classification depends on accurate diagnosis of those severe headaches that are migraines and those headaches that are not, as well as diagnosis of ever experiencing aura. Aura is a specific focal neurologic symptom. For more information about headache classification see the International Headache Society’s International Classification of Headache Disorders, 3rd ed. (https://ichd-3.org) (181). Any new headaches or marked changes in headaches should be evaluated.
b. Migraine Clarification: Classification depends on accurate diagnosis of those severe headaches that are migraines and those headaches that are not, as well as diagnosis of ever experiencing aura. Aura is a specific focal neurologic symptom. For more information about headache classification see the International Headache Society’s International Classification of Headache Disorders, 3rd ed. (https://ichd-3.org) (181). Any new headaches or marked changes in headaches should be evaluated.
Clarification: Classification is for persons without any other risk factors for stroke (e.g., age, hypertension, and smoking).
Evidence: Among women with migraine, oral contraceptive use is associated with about a threefold increased risk for ischemic stroke compared with nonuse, although most studies did not specify migraine type or oral contraceptive formulation. The only study to examine migraine type found that the risk for ischemic stroke among women with migraine with aura was increased to a similar level among both oral contraceptive users and nonusers, compared with women without migraine (182). The risk for ischemic stroke is increased among women using COCs, compared with women not using COCs (101,183). The risk for ischemic stroke is also increased among women with migraine with aura, compared with women without migraine (184186). One older meta-analysis found that migraine without aura was associated with an increased risk for ischemic stroke, while two more recent meta-analyses did not find such an association (184186).
Comment: Menstrual migraine is a subtype of migraine without aura. For more information, see the International Headache Society’s International Classification of Headache Disorders, 3rd ed. (https://ichd-3.org) (181).
   i. Without aura (includes menstrual migraine) 2
   ii. With aura 4
Epilepsy
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 Clarification: If a person is taking anticonvulsants, see recommendations for Drug Interactions. Certain anticonvulsants lower COC effectiveness. The extent to which patch or ring use is similar to COC use in this regard remains unclear.
Multiple sclerosis Evidence: Limited evidence suggests that use of COCs or oral contraceptives (type not specified) among women with multiple sclerosis does not worsen the clinical course of disease (187).
Comment: No data exist that evaluate the increased risk for VTE among persons with multiple sclerosis using CHCs. However, persons with multiple sclerosis are at higher risk for VTE than those without multiple sclerosis.
a. Without prolonged immobility 1
b. With prolonged immobility 3
Depressive Disorders
Depressive disorders 1 Clarification: If a person is receiving psychotropic medications or St. John’s wort, see recommendations for Drug Interactions.
Evidence: COC use was not associated with increased depressive symptoms in women with depression or scoring above threshold levels on a validated depression screening instrument compared with baseline or with nonusers with depression. One small study of women with bipolar disorder found that oral contraceptives did not significantly change mood across the menstrual cycle (188).
Reproductive Tract Infections and Disorders
Vaginal bleeding patterns
a. Irregular pattern without heavy bleeding 1 Comment: Irregular menstrual bleeding patterns are common among healthy persons.
b. Heavy or prolonged bleeding (includes regular and irregular patterns) 1 Clarification: Unusually heavy bleeding should raise the suspicion of a serious underlying condition.
Evidence: A Cochrane Collaboration Review identified one RCT evaluating the effectiveness of COC use compared with naproxen and danazol in treating menorrhagia. Women with menorrhagia did not report worsening of the condition or any adverse events related to COC use (189).
Unexplained vaginal bleeding
(suspicious for serious condition) before evaluation
2 Clarification: If pregnancy or an underlying pathological condition (e.g., pelvic malignancy) is suspected, it must be evaluated and the category adjusted after evaluation.
Comment: No conditions that cause vaginal bleeding will be worsened in the short-term by use of CHCs.
Endometriosis 1 Evidence: A Cochrane Collaboration Review identified one RCT evaluating the effectiveness of COC use compared with a gonadotropin-releasing hormone analog in treating the symptoms of endometriosis. Women with endometriosis did not report worsening of the condition or any adverse events related to COC use (190).
Benign ovarian tumors (including cysts) 1
Severe dysmenorrhea 1 Evidence: Risk for side effects with COC use was not higher among women with dysmenorrhea than among women not using COCs. Certain COC users had a reduction in pain and bleeding (191,192).
Gestational trophoblastic disease
This condition is associated with increased
risk for adverse health events as a result of
pregnancy (Box 3).
Clarification: For all subconditions of gestational trophoblastic disease, classifications are based on the assumption that persons are under close medical supervision because of the need for monitoring of β-hCG levels for appropriate disease surveillance.
Evidence: After molar pregnancy evacuation, the balance of evidence found COC use did not increase the risk for postmolar trophoblastic disease, and β–hCG levels regressed more rapidly in certain COC users than in nonusers (193). Limited evidence suggests that use of COCs during chemotherapy does not significantly affect the regression or treatment of postmolar trophoblastic disease compared with women who used a nonhormonal contraceptive method or DMPA during chemotherapy (193).
a. Suspected gestational trophoblastic
disease (immediate postevacuation)
   i. Uterine size first trimester 1
   ii. Uterine size second trimester 1
b. Confirmed gestational trophoblastic
disease (after initial evacuation and during
monitoring)
   i. Undetectable or nonpregnant β-hCG levels 1
   ii. Decreasing β-hCG levels 1
   iii. Persistently elevated β-hCG levels or malignant disease, with no evidence or suspicion of intrauterine disease 1
   iv. Persistently elevated β-hCG levels or malignant disease, with evidence or suspicion of intrauterine disease 1
Cervical ectropion 1 Comment: Cervical ectropion is not a risk factor for cervical cancer, and restriction of CHC use is unnecessary.
Cervical intraepithelial neoplasia 2 Evidence: Among women with persistent human papillomavirus infection, long-term COC use (≥5 years) might increase the risk for carcinoma in situ and invasive carcinoma (194). Limited evidence on women with low-grade squamous intraepithelial lesions found use of the vaginal ring did not worsen the condition (9).
Cervical cancer (awaiting treatment) 2 Comment: Theoretical concern exists that CHC use might affect prognosis of the existing disease. While awaiting treatment, persons may use CHCs. In general, treatment of this condition can render a person infertile.
Breast disease
Breast cancer is associated with increased
risk for adverse health events as a result of
pregnancy (Box 3).
a. Undiagnosed mass 2 Clarification: Evaluation of mass should be pursued as early as possible.
b. Benign breast disease 1
c. Family history of cancer 1 Evidence: Women with breast cancer susceptibility genes (e.g., BRCA1 and BRCA2) have a higher baseline risk for breast cancer than women without these genes. The baseline risk for breast cancer also is higher among women with a family history of breast cancer than among those who do not have such a history. However, evidence does not suggest that the increased risk for breast cancer among women with either a family history of breast cancer or breast cancer susceptibility genes is modified by the use of COCs (195212).
d. Breast cancer Comment: Breast cancer is a hormonally sensitive tumor, and the prognosis for persons with current or recent breast cancer might worsen with CHC use.
   i. Current 4
   ii. Past and no evidence of current disease for 5 years 3
Endometrial hyperplasia 1
Endometrial cancer
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 Comment: COC use reduces the risk for endometrial cancer; whether patch or ring use reduces the risk for endometrial cancer is not known. While awaiting treatment, patients may use CHCs. In general, treatment of this condition can render a person infertile.
Ovarian cancer
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 Comment: COC use reduces the risk for ovarian cancer; whether patch or ring use reduces the risk for ovarian cancer is not known. While awaiting treatment, patients may use CHCs. In general, treatment of this condition can render a person infertile.
Uterine fibroids 1 Comment: COCs do not appear to cause growth of uterine fibroids, and patch and ring also are not expected to cause growth.
Pelvic inflammatory disease Comment: COCs might reduce the risk for PID among persons with STIs but do not protect against HIV infection or lower genital tract STIs. Whether use of patch or ring reduces the risk for PID among persons with STIs is unknown; however, they do not protect against HIV infection or lower genital tract STIs.
a. Current PID 1
b. Past PID
   i. With subsequent pregnancy 1
   ii. Without subsequent pregnancy 1
Sexually transmitted infections
a. Current purulent cervicitis or chlamydial infection or gonococcal infection 1
b. Vaginitis (including Trichomonas vaginalis and bacterial vaginosis) 1
c. Other factors related to STIs 1
HIV
High risk for HIV infection 1 Evidence: Low-to-moderate-quality evidence from 11 observational studies suggested no association between COC use (it was assumed that studies that did not specify oral contraceptive type examined mostly, if not exclusively, COC use) and HIV acquisition. No studies of patch or ring were identified (213,214).
HIV infection
For persons with HIV infection who are not clinically well or not receiving ARV therapy, this condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 Clarification: Drug interactions might exist between hormonal contraceptives and ARV drugs; see recommendations for Drug Interactions.
Evidence: Overall, evidence does not support an association between COC use and progression of HIV. Limited direct evidence does not support an association between COC use and transmission of HIV to noninfected partners; studies measuring genital viral shedding as a proxy for infectivity have had mixed results. Studies measuring whether hormonal contraceptive methods affect plasma HIV viral load generally have found no effect (215217).
Other Infections
Schistosomiasis
Schistosomiasis with fibrosis of the liver is
associated with increased risk for adverse
health events as a result of pregnancy
(Box 3).
a. Uncomplicated 1 Evidence: Among women with uncomplicated schistosomiasis, COC use had no adverse effects on liver function (218224).
b. Fibrosis of the liver (if severe, see recommendations for Cirrhosis) 1
Tuberculosis
This condition is associated with
increased risk for adverse health events
as a result of pregnancy (Box 3).
Clarification: If a person is taking rifampin, see recommendations for Drug Interactions. Rifampin is likely to decrease COC effectiveness. The extent to which patch or ring use is similar to COC use in this regard remains unclear.
a. Nonpelvic 1
b. Pelvic 1
Malaria 1
Endocrine Conditions
Diabetes
Insulin-dependent diabetes; diabetes
with nephropathy, retinopathy, or
neuropathy; diabetes with other vascular
disease; or diabetes of >20 years’
duration are associated with increased
risk for adverse health events as a result
of pregnancy (Box 3).
a. History of gestational disease 1 Evidence: The development of non–insulin-dependent diabetes in women with a history of gestational diabetes is not increased by use of COCs (225232). Likewise, lipid levels appear to be unaffected by COC use (233235).
b. Nonvascular disease Evidence: Among women with insulin-dependent or non–insulin-dependent diabetes, COC use had limited effect on daily insulin requirements and no effect on long-term diabetes control (e.g., glycosylated hemoglobin levels) or progression to retinopathy. Changes in lipid profile and hemostatic markers were limited, and most changes remained within normal values (236245).
   i. Non-insulin dependent 2
   ii. Insulin dependent 2
c. Nephropathy, retinopathy, or neuropathy 3/4 Clarification: The category should be assessed according to the severity of the condition.
d. Other vascular disease or diabetes of >20 years’ duration 3/4 Clarification: The category should be assessed according to the severity of the condition.
Thyroid disorders
a. Simple goiter 1
b. Hyperthyroid 1
c. Hypothyroid 1
Gastrointestinal Conditions
Inflammatory bowel disease (ulcerative colitis or Crohn’s disease) 2/3 Clarification: For persons with mild IBD and with no other risk factor for VTE, the benefits of CHC use generally outweigh the risks (category 2). However, for persons with IBD who are at increased risk for VTE (e.g., those with active or extensive disease, surgery, immobilization, corticosteroid use, vitamin deficiencies, or fluid depletion), the risks of CHC use generally outweigh the benefits (category 3).
Evidence: Risk for disease relapse was not significantly higher among women with IBD using oral contraceptives (most studies did not specify type) than among nonusers (246). Absorption of COCs among women with mild ulcerative colitis and no or small ileal resections was similar to the absorption among healthy women (246). Findings might not apply to women with Crohn’s disease or more extensive bowel resections. No data exist that evaluate the increased risk for VTE among women with IBD using CHCs. However, women with IBD are at higher risk than unaffected women for VTE (246).
Gallbladder disease Comment: CHCs might cause a small increased risk for gallbladder disease. CHCs might worsen existing gallbladder disease.
a. Asymptomatic 2
b. Symptomatic
   i. Current 3
   ii. Treated by cholecystectomy 2
   iii. Medically treated 3
History of cholestasis
a. Pregnancy related 2 Comment: History of pregnancy-related cholestasis might predict an increased risk for COC-related cholestasis.
b. Past COC related 3 Comment: History of COC-related cholestasis predicts an increased risk with subsequent COC use.
Viral hepatitis Initiation Continuation
a. Acute or flare 3/4 2 Clarification (initiation): The category should be assessed according to the severity of the condition.
Evidence: Limited evidence was identified on COC use among persons with acute viral hepatitis. Data suggest that in women with chronic viral hepatitis, COC use does not increase the risk or severity of fibrosis, nor does it increase the risk for hepatocellular carcinoma. For women with chronic viral hepatitis, COC use does not appear to trigger severe liver dysfunction (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
Comment: Hepatic metabolism of exogenous hormones might be impaired in persons with liver dysfunction, which could lead to increased estrogen levels in circulation and estrogen-related side effects and adverse events (e.g., thrombosis).
b. Chronic 1 1 Evidence: Data suggest that in women with chronic viral hepatitis, COC use does not increase the risk or severity of fibrosis, nor does it increase the risk for hepatocellular carcinoma. For women with chronic viral hepatitis, COC use does not appear to trigger severe liver dysfunction (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
Cirrhosis
Decompensated cirrhosis is associated
with increased risk for adverse health
events as a result of pregnancy (Box 3).
a. Compensated (normal liver function) 1 Evidence: No direct evidence was identified on CHC use among persons with compensated cirrhosis (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
b. Decompensated (impaired liver function) 4 Evidence: No direct evidence was identified on CHC use among persons with decompensated cirrhosis (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
Comment: Hepatic metabolism of exogenous hormones might be impaired in persons with liver dysfunction, which could lead to increased estrogen levels in circulation and estrogen-related side effects and adverse events (e.g., thrombosis). Any estrogen-related hepatotoxicity might be less tolerated in persons with existing liver dysfunction.
Liver tumors
Hepatocellular adenoma and malignant
liver tumors are associated with increased
risk for adverse health events
as a result of pregnancy (Box 3).
a. Benign
   i. Focal nodular hyperplasia 2 Evidence: Limited evidence suggests that COC use does not influence either progression or regression of focal nodular hyperplasia (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
   ii. Hepatocellular adenoma 4 Evidence: Evidence suggests that COC use is associated with progression of hepatocellular adenoma growth, while COC discontinuation is associated with stability or regression (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
b. Malignant (hepatocellular carcinoma) 4 Evidence: No direct evidence was identified on CHC use among persons with hepatocellular carcinoma (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
Respiratory Conditions
Cystic fibrosis
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 Clarification: Persons with cystic fibrosis are at increased risk for diabetes, liver disease, gallbladder disease, and VTE (particularly related to use of central venous catheters) and are frequently prescribed antibiotics. Categories assigned to such conditions in U.S. MEC should be the same for persons with cystic fibrosis who have these conditions. For cystic fibrosis, classifications are based on the assumption that no other conditions are present; these classifications must be modified in the presence of such conditions.
Clarification: Certain drugs to treat cystic fibrosis (e.g., lumacaftor) might reduce effectiveness of hormonal contraceptives, including oral, injectable, transdermal, and implantable contraceptives.
Evidence: Limited evidence suggests that use of COCs or oral contraceptives (type not specified) among women with cystic fibrosis is not associated with worsening of disease severity. Very limited evidence suggests that cystic fibrosis does not impair the effectiveness of hormonal contraception (247).
Hematologic Conditions
Thalassemia 1 Comment: Anecdotal evidence from countries where thalassemia is prevalent indicates that COC use does not worsen the condition.
Sickle cell disease
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
4 Evidence: Persons with sickle cell disease are at higher risk for stroke and venous thrombosis than the general population (248251). CHC use might further elevate risk for thrombosis among persons with sickle cell disease, but evidence is limited (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
Iron deficiency anemia 1 Comment: CHC use might decrease menstrual blood loss.
Solid Organ Transplantation
Solid organ transplantation
This condition is associated with
increased risk for adverse health events
as a result of pregnancy (Box 3).
a. No graft failure 2 Clarification: Persons with transplant due to Budd-Chiari syndrome should not use CHCs because of the increased risk for thrombosis.
Evidence: Limited evidence among CHC users indicated no adverse events and no overall changes in biochemical parameters (e.g., blood pressure, cholesterol) and no pregnancies (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516). However, one study reported discontinuations of COC use in two (8%) of 26 women as a result of serious medical complications, including acute graft rejection (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
b. Graft failure 4 Evidence: Limited evidence among CHC users indicated no adverse events and no overall changes in biochemical parameters (e.g., blood pressure, cholesterol) and no pregnancies (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516). However, one study reported discontinuations of COC use in two (8%) of 26 women as a result of serious medical complications, including acute graft rejection (Supplementary Appendix, https://stacks.cdc.gov/view/cdc/156516).
Drug Interactions
Antiretrovirals used for prevention
(PrEP) or treatment of HIV infection
Comment: These recommendations generally are for ARV agents used alone. However, most persons receiving ARV therapy are using multiple drugs in combination. In general, whether interactions between ARVs and hormonal contraceptives differ when ARVs are given alone or in combination is unknown.
See the following guidelines for the most up-to-date recommendations on drug-drug interactions between hormonal contraception and ARVs: 1) Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions to Reduce Perinatal HIV Transmission in the United States (https://clinicalinfo.hiv.gov/en/guidelines/perinatal/prepregnancy-counseling-childbearing-age-overview?view=full#table-3) (252) and 2) Guidelines for the Use of Antiretroviral Agents in Adults and Adolescents with HIV (https://clinicalinfo.hiv.gov/en/guidelines/hiv-clinical-guidelines-adult-and-adolescent-arv/drug-interactions-overview?view=full) (253).
a. Nucleoside reverse transcriptase
inhibitors (NRTIs)
   i. Abacavir (ABC) 1 Evidence: NRTIs do not appear to have significant risk for interactions with hormonal contraceptive methods (254259).
   ii. Tenofovir (TDF) 1
   iii. Zidovudine (AZT) 1
   iv. Lamivudine (3TC) 1
   v. Didanosine (DDI) 1
   vi. Emtricitabine (FTC) 1
   vii. Stavudine (D4T) 1
b. Nonnucleoside reverse transcriptase
inhibitors (NNRTIs)
   i. Efavirenz (EFV) 2 Clarification: Evidence suggests drug interactions between EFV and certain hormonal contraceptives. These interactions might reduce the effectiveness of the hormonal contraceptive.
Evidence: Two studies suggested that pregnancy rates might be higher among women using COCs and EFV compared with COCs alone, although one study found no difference in pregnancy rates (260262). Two studies found conflicting results on ovulations in women receiving COCs and EFV compared with EFV alone (263,264). Two pharmacokinetic studies demonstrated decreases in ethinyl estradiol and progestin concentrations in women receiving COCs and EFV compared with COCs alone (264,265). Pharmacokinetic studies demonstrated generally no changes in EFV concentrations with concomitant COC use (264266).
   ii. Etravirine (ETR) 1 Evidence: One study demonstrated no clinically relevant pharmacokinetic or pharmacodynamic changes in women using COCs and ETR compared with COCs alone (267).
   iii. Nevirapine (NVP) 1 Evidence: Five studies found no significant differences in pregnancy rates among women using COCs and NVP compared with women using COCs alone (260262,266,268). Three studies reported no ovulations among women receiving COCs and NVP (263,268,269). Two pharmacokinetic studies demonstrated decreased concentrations of ethinyl estradiol and progestin among women using COCs and NVP compared with COCs alone, and one study found no change in contraceptive hormone concentrations (263,269,270). Pharmacokinetic studies demonstrated generally no changes in NVP concentrations with concomitant COC use (263,270,271).
   iv. Rilpivirine (RPV) 1 Evidence: One study demonstrated no clinical significant pharmacokinetic changes or adverse events in women using COCs and RPV compared with COCs alone (272).
c. Ritonavir-boosted protease inhibitors
   i. Ritonavir-boosted atazanavir (ATV/r) 2 Clarification: Theoretically, drug interactions might occur between certain ritonavir-boosted protease inhibitors and certain hormonal contraceptives that might reduce the effectiveness of the hormonal contraceptive.
Evidence: One pharmacokinetic study demonstrated decreased estrogen but increased progestin concentrations in women using COCs and ATV/r compared with COCs alone (273).
   ii. Ritonavir-boosted darunavir (DRV/r) 2 Clarification: Theoretically, drug interactions might occur between certain ritonavir-boosted protease inhibitors and certain hormonal contraceptives that might reduce the effectiveness of the hormonal contraceptive.
Evidence: One pharmacokinetic study demonstrated no change in follicle-stimulating hormone or luteinizing hormone but decreases in ethinyl estradiol and norethindrone in women using COCs with DRV/r compared with COCs alone (274).
   iii. Ritonavir-boosted fosamprenavir (FPV/r) 2 Clarification: Theoretically, drug interactions might occur between certain ritonavir-boosted protease inhibitors and certain hormonal contraceptives that might reduce the effectiveness of the hormonal contraceptive.
Evidence: Information from the package label states that both ethinyl estradiol and norethindrone concentrations decreased with concurrent administration of COCs and FPV/r (275).
   iv. Ritonavir-boosted lopinavir (LPV/r) 1 Evidence: One study demonstrated a nonsignificant increase in pregnancy rates among women using COCs and LPV/r compared with COCs alone (260). One study demonstrated no ovulations in women using the combined hormonal patch and LPV/r compared with combined hormonal patch alone; ethinyl estradiol concentrations for COC and patch users decreased but norelgestromin concentrations increased with use of the patch (276).
   v. Ritonavir-boosted saquinavir (SQV/r) 2 Clarification: Theoretically, drug interactions might occur between certain ritonavir-boosted protease inhibitors and certain hormonal contraceptives that might reduce the effectiveness of the hormonal contraceptive.
Evidence: One pharmacokinetic study demonstrated no change in SQV concentrations in women using COC and SQV compared with COCs alone (277).
   iv. Ritonavir-boosted tipranavir (TPV/r) 2 Clarification: Theoretically, drug interactions might occur between certain ritonavir-boosted protease inhibitors and certain hormonal contraceptives that might reduce the effectiveness of the hormonal contraceptive.
Evidence: Information from the package label states that ethinyl estradiol concentrations decrease but norethindrone concentrations increased with concurrent administration of COCs and TPV/r (278).
d. Protease inhibitors without ritonavir
  i. Atazanavir (ATV) 2 Clarification: Theoretical concern exists that increased levels of ethinyl estradiol because of interactions with ATV might increase the risk for adverse events.
Evidence: Information from the package label states that there are inconsistent changes in ethinyl estradiol concentrations and increases in progestin concentrations with concurrent administration of two different COCs and ATV (279).
Comment: When ATV is administered with cobicistat, theoretical concern exists for a drug interaction with hormonal contraceptives. Cobicistat is an inhibitor of CYP3A and CYP2D6 and could theoretically increase contraceptive hormone levels. However, its effects on CYP enzymes and drug levels might vary when combined with other ARVs.
  ii. Fosamprenavir (FPV) 3 Clarification: Concern exists that interactions between FPV and hormonal contraceptives leading to decreased levels of FPV might diminish effectiveness of the ARV drug.
Evidence: Information from the package label states that amprenavir concentrations decreased with concurrent administration of COCs and amprenavir. Norethindrone concentrations increased and ethinyl estradiol concentrations did not change (275).
  iii. Indinavir (IDV) 1 Evidence: One small study found no pregnancies in women using COCs and IDV (262).
  iv. Nelfinavir (NFV) 2 Clarification: Evidence suggests drug interactions between certain protease inhibitors and certain hormonal contraceptives. These interactions might reduce the effectiveness of the hormonal contraceptive.
Evidence: One small study suggested that women using COCs and NFV might have had higher pregnancy rates than those using COCs alone (262).
e. CCR5 co-receptor antagonists
   i. Maraviroc (MVC) 1 Evidence: COC concentrations were not altered by co-administration with MVC (280).
f. HIV integrase strand transfer inhibitors
  i. Raltegravir (RAL) 1 Evidence: One pharmacokinetic study demonstrated increased concentrations of norgestimate and no change in ethinyl estradiol among women using COCs and RAL compared with COCs alone (281).
  ii. Dolutegravir (DTG) 1 Evidence: One study demonstrated no clinically relevant pharmacokinetic or pharmacodynamic changes in women using COCs and DTG compared with COCs alone (282).
  iii. Elvitegravir (EVG) 1 Evidence: Information from the package label states that ethinyl estradiol concentrations decreased and norgestimate concentrations increased with concurrent administration of COCs and EVG (283).
Comment: When EVG is administered with cobicistat, theoretical concern exists for a drug interaction with hormonal contraceptives. Cobicistat is an inhibitor of CYP3A and CYP2D6 and could theoretically increase contraceptive hormone levels. However, its effects on CYP enzymes and drug levels might vary when combined with other ARVs.
g. Fusion inhibitors
  i. Enfuvirtide 1
Anticonvulsant therapy
a. Certain anticonvulsants (phenytoin, carbamazepine, barbiturates, primidone, topiramate, oxcarbazepine) 3 Clarification: Although the interaction of certain anticonvulsants with CHCs is not harmful, it is likely to reduce the effectiveness of CHCs. Use of other contraceptives should be encouraged for persons who are long-term users of any of these drugs. When a COC is chosen, a preparation containing a minimum of 30 μg ethinyl estradiol should be used.
Evidence: Use of certain anticonvulsants might decrease the effectiveness of COCs (284288).
b. Lamotrigine 3 Clarification: The recommendation for lamotrigine applies only for situations where lamotrigine monotherapy is taken concurrently with COCs. Anticonvulsant treatment regimens that combine lamotrigine and non–enzyme-inducing antiepileptic drugs (e.g., sodium valproate) do not interact with COCs.
Evidence: Pharmacokinetic studies demonstrate levels of lamotrigine decrease significantly during COC use (288293). Certain women who used both COCs and lamotrigine experienced increased seizure activity in one trial (289).
Antimicrobial therapy
a. Broad-spectrum antibiotics 1 Evidence: Most broad-spectrum antibiotics do not affect the contraceptive effectiveness of COCs (294330), patch (331), or ring (332).
b. Antifungals 1 Evidence: Studies of antifungal agents have demonstrated no clinically significant pharmacokinetic interactions with COCs (333342) or ring (343).
c. Antiparasitics 1 Evidence: Studies of antiparasitic agents have demonstrated no clinically significant pharmacokinetic interactions with COCs (218,344348).
d. Rifampin or rifabutin therapy 3 Clarification: Although the interaction of rifampin or rifabutin therapy with CHCs is not harmful, it is likely to reduce the effectiveness of CHCs. Use of other contraceptives should be encouraged for persons who are long-term users of either of these drugs. When a COC is chosen, a preparation containing a minimum of 30 μg ethinyl estradiol should be used.
Evidence: The balance of the evidence suggests that rifampin reduces the effectiveness of COCs (349363). Data on rifabutin are limited, but effects on metabolism of COCs are less than with rifampin, and small studies have not demonstrated evidence of ovulation (351,357).
Psychotropic medications Comment: For many common psychotropic agents, limited or no theoretical concern exists for clinically significant drug interactions when co-administered with hormonal contraceptives. However, either no or very limited data exist examining potential interactions for these classes of medications. For psychotropic agents that are CYP1A2 substrates (e.g., duloxetine, mirtazapine, ziprasidone, olanzapine, clomipramine, imipramine, and amitriptyline), co-administration with CHCs could theoretically yield increased concentrations of the psychotropic drug. For agents with narrow therapeutic windows (e.g., tricyclic antidepressants), increased drug concentrations might pose safety concerns that could necessitate closer monitoring.
a. Selective serotonin reuptake inhibitors (SSRIs) 1 Evidence: Limited clinical and pharmacokinetic data do not demonstrate concern for SSRIs decreasing the effectiveness of oral contraceptives. Limited evidence suggests that for women taking SSRIs, the use of hormonal contraceptives was not associated with differences in effectiveness of the SSRI for treatment or in adverse events when compared with women not taking hormonal contraceptives (364).
Comment: Drugs that are inhibitors of CYP3A4 or CYP2C9 theoretically have the potential to increase levels of contraceptive steroids which might increase adverse events. Fluvoxamine is an SSRI known to be a moderate inhibitor of both CYP3A4 and CYP2C9; however, no clinical or pharmacokinetic studies were identified to explore potential drug-drug interactions.
St. John’s wort 2 Evidence: Although clinical data are limited, studies with pharmacokinetic and pharmacodynamics outcomes raise concern that St. John’s wort might decrease effectiveness of hormonal contraceptives, including increased risk for breakthrough bleeding and ovulation and increased metabolism of estrogen and progestins. Any interactions might be dependent on the dose of St. John’s wort, and the concentration of active ingredients across types of St. John’s wort preparations might vary (365).

Abbreviations: ARV = antiretroviral; BMD = bone mineral density; BMI = body mass index; CHC = combined hormonal contraceptive; CKD = chronic kidney disease; COC = combined oral contraceptive; CYP = cytochrome P450; DMPA = depot medroxyprogesterone acetate; DVT = deep venous thrombosis; hCG = human chorionic gonadotropin; HDL = high-density lipoprotein; IBD = inflammatory bowel disease; IUD = intrauterine device; LDL = low-density lipoprotein; NA = not applicable; PE = pulmonary embolism; PID = pelvic inflammatory disease; POC = progestin-only contraceptive; PrEP = pre-exposure prophylaxis; RCT = randomized clinical trial; SLE = systemic lupus erythematosus; SSRI = selective serotonin reuptake inhibitor; STI = sexually transmitted infection; U.S. MEC = U.S. Medical Eligibility Criteria for Contraceptive Use; U.S. SPR = U.S. Selected Practice Recommendations for Contraceptive Use; VTE = venous thromboembolism.

References

  1. Abrams LS, Skee DM, Natarajan J, Wong FA, Lasseter KC. Multiple-dose pharmacokinetics of a contraceptive patch in healthy women participants. 2001;64:287–94. https://doi.org/10.1016/S0010-7824(01)00273-6 PMID:11777488
  2. Ahrendt HJ, Nisand I, Bastianelli C, et al. Efficacy, acceptability and tolerability of the combined contraceptive ring, NuvaRing, compared with an oral contraceptive containing 30 microg of ethinyl estradiol and 3 mg of drospirenone. 2006;74:451–7. https://doi.org/10.1016/j.contraception.2006.07.004 PMID:17157101
  3. Audet MC, Moreau M, Koltun WD, et al. ; ORTHO EVRA/EVRA 004 Study Group. Evaluation of contraceptive efficacy and cycle control of a transdermal contraceptive patch vs an oral contraceptive: a randomized controlled trial. 2001;285:2347–54. https://doi.org/10.1001/jama.285.18.2347 PMID:11343482
  4. Bjarnadóttir RI, Tuppurainen M, Killick SR. Comparison of cycle control with a combined contraceptive vaginal ring and oral levonorgestrel/ethinyl estradiol. 2002;186:389–95. https://doi.org/10.1067/mob.2002.121103 PMID:11904596
  5. Boonyarangkul A, Taneepanichskul S. Comparison of cycle control and side effects between transdermal contraceptive patch and an oral contraceptive in women older than 35 years. 2007;90:1715–9. PMID:17957909
  6. Burkman RT. The transdermal contraceptive patch: a new approach to hormonal contraception. 2002;47:69–76. PMID:11991433
  7. Cole JA, Norman H, Doherty M, Walker AM. Venous thromboembolism, myocardial infarction, and stroke among transdermal contraceptive system users. 2007;109:339–46. https://doi.org/10.1097/01.AOG.0000250968.82370.04 PMID:17267834
  8. Devineni D, Skee D, Vaccaro N, et al. Pharmacokinetics and pharmacodynamics of a transdermal contraceptive patch and an oral contraceptive. 2007;47:497–509. https://doi.org/10.1177/0091270006297919 PMID:17389559
  9. Dieben TOM, Roumen FJME, Apter D. Efficacy, cycle control, and user acceptability of a novel combined contraceptive vaginal ring. 2002;100:585–93. PMID:12220783
  10. Dittrich R, Parker L, Rosen JB, Shangold G, Creasy GW, Fisher AC; Ortho Evra/Evra 001 Study Group. Transdermal contraception: evaluation of three transdermal norelgestromin/ethinyl estradiol doses in a randomized, multicenter, dose-response study. 2002;186:15–20. https://doi.org/10.1067/mob.2002.118844 PMID:11810078
  11. Duijkers I, Killick S, Bigrigg A, Dieben TOM. A comparative study on the effects of a contraceptive vaginal ring NuvaRing and an oral contraceptive on carbohydrate metabolism and adrenal and thyroid function. 2004;9:131–40. https://doi.org/10.1080/13625180400007199 PMID:15697102
  12. Duijkers IJ, Klipping C, Verhoeven CH, Dieben TOM. Ovarian function with the contraceptive vaginal ring or an oral contraceptive: a randomized study. 2004;19:2668–73. https://doi.org/10.1093/humrep/deh493 PMID:15333593
  13. Elkind-Hirsch KE, Darensbourg C, Ogden B, Ogden LF, Hindelang P. Contraceptive vaginal ring use for women has less adverse metabolic effects than an oral contraceptive. 2007;76:348–56. https://doi.org/10.1016/j.contraception.2007.08.001 PMID:17963858
  14. Hedon B, Helmerhorst FM, Cronje HS, et al. ; The EVRA 003 Study Group. Comparison of efficacy, cycle control, compliance and safety in users of a contraceptive patch versus an oral contraceptive. 2000;70:78. https://doi.org/10.1016/S0020-7292(00)85161-9
  15. Jick S, Kaye JA, Li L, Jick H. Further results on the risk of nonfatal venous thromboembolism in users of the contraceptive transdermal patch compared to users of oral contraceptives containing norgestimate and 35 microg of ethinyl estradiol. 2007;76:4–7. https://doi.org/10.1016/j.contraception.2007.03.003 PMID:17586129
  16. Jick SS, Jick H. Cerebral venous sinus thrombosis in users of four hormonal contraceptives: levonorgestrel-containing oral contraceptives, norgestimate-containing oral contraceptives, desogestrel-containing oral contraceptives and the contraceptive patch. 2006;74:290–2. https://doi.org/10.1016/j.contraception.2006.05.071 PMID:16982227
  17. Jick SS, Jick H. The contraceptive patch in relation to ischemic stroke and acute myocardial infarction. 2007;27:218–20. https://doi.org/10.1592/phco.27.2.218 PMID:17253912
  18. Jick SS, Kaye JA, Russmann S, Jick H. Risk of nonfatal venous thromboembolism in women using a contraceptive transdermal patch and oral contraceptives containing norgestimate and 35 microg of ethinyl estradiol. 2006;73:223–8. https://doi.org/10.1016/j.contraception.2006.01.001 PMID:16472560
  19. Magnusdóttir EM, Bjarnadóttir RI, Onundarson PT, et al. The contraceptive vaginal ring (NuvaRing) and hemostasis: a comparative study. 2004;69:461–7. https://doi.org/10.1016/j.contraception.2003.12.010 PMID:15157790
  20. Massai R, Mäkäräinen L, Kuukankorpi A, Klipping C, Duijkers I, Dieben T. The combined contraceptive vaginal ring and bone mineral density in healthy pre-menopausal women. 2005;20:2764–8. https://doi.org/10.1093/humrep/dei117 PMID:15980008
  21. Milsom I, Lete I, Bjertnaes A, et al. Effects on cycle control and bodyweight of the combined contraceptive ring, NuvaRing, versus an oral contraceptive containing 30 microg ethinyl estradiol and 3 mg drospirenone. 2006;21:2304–11. https://doi.org/10.1093/humrep/del162 PMID:16763008
  22. Oddsson K, Leifels-Fischer B, de Melo NR, et al. Efficacy and safety of a contraceptive vaginal ring (NuvaRing) compared with a combined oral contraceptive: a 1-year randomized trial. 2005;71:176–82. https://doi.org/10.1016/j.contraception.2004.09.001 PMID:15722066
  23. Pierson RA, Archer DF, Moreau M, Shangold GA, Fisher AC, Creasy GW. Ortho Evra/Evra versus oral contraceptives: follicular development and ovulation in normal cycles and after an intentional dosing error. 2003;80:34–42. https://doi.org/10.1016/S0015-0282(03)00556-9 PMID:12849799
  24. Radowicki S, Skórzewska K, Szlendak K. [Safety evaluation of a transdermal contraceptive system with an oral contraceptive]. 2005;76:884–9. PMID:16566363
  25. Sabatini R, Cagiano R. Comparison profiles of cycle control, side effects and sexual satisfaction of three hormonal contraceptives. 2006;74:220–3. https://doi.org/10.1016/j.contraception.2006.03.022 PMID:16904415
  26. Smallwood GH, Meador ML, Lenihan JP Jr, Shangold GA, Fisher AC, Creasy GW; ORTHO EVRA/EVRA 002 Study Group. Efficacy and safety of a transdermal contraceptive system. 2001;98:799–805. https://doi.org/10.1097/00006250-200111000-00016 PMID:11704172
  27. Timmer CJ, Mulders TM. Pharmacokinetics of etonogestrel and ethinylestradiol released from a combined contraceptive vaginal ring. 2000;39:233–42. https://doi.org/10.2165/00003088-200039030-00005 PMID:11020137
  28. Tuppurainen M, Klimscheffskij R, Venhola M, Dieben TOM. The combined contraceptive vaginal ring (NuvaRing) and lipid metabolism: a comparative study. 2004;69:389–94. https://doi.org/10.1016/j.contraception.2004.01.004 PMID:15105061
  29. Urdl W, Apter D, Alperstein A, et al. ; ORTHO EVRA/EVRA 003 Study Group. Contraceptive efficacy, compliance and beyond: factors related to satisfaction with once-weekly transdermal compared with oral contraception. 2005;121:202–10. https://doi.org/10.1016/j.ejogrb.2005.01.021 PMID:16054963
  30. van den Heuvel MW, van Bragt AJM, Alnabawy AKM, Kaptein MCJ. Comparison of ethinylestradiol pharmacokinetics in three hormonal contraceptive formulations: the vaginal ring, the transdermal patch and an oral contraceptive. 2005;72:168–74. https://doi.org/10.1016/j.contraception.2005.03.005 PMID:16102549
  31. Veres S, Miller L, Burington B. A comparison between the vaginal ring and oral contraceptives. 2004;104:555–63. https://doi.org/10.1097/01.AOG.0000136082.59644.13 PMID:15339769
  32. White T, Ozel B, Jain JK, Stanczyk FZ. Effects of transdermal and oral contraceptives on estrogen-sensitive hepatic proteins. 2006;74:293–6. https://doi.org/10.1016/j.contraception.2006.04.005 PMID:16982228
  33. Zieman M, Guillebaud J, Weisberg E, Shangold GA, Fisher AC, Creasy GW. Contraceptive efficacy and cycle control with the Ortho Evra/Evra transdermal system: the analysis of pooled data. 2002;77(Suppl 2):S13–8. https://doi.org/10.1016/S0015-0282(01)03275-7 PMID:11849631
  34. Workowski KA, Bachmann LH, Chan PA, et al. Sexually transmitted infections treatment guidelines, 2021. 2021;70(No. RR-4):1–187. https://doi.org/10.15585/mmwr.rr7004a1 PMID:34292926
  35. CDC. US Public Health Service preexposure prophylaxis for the prevention of HIV infection in the United States—2021 update: a clinical practice guideline. Atlanta, GA: US Department of Health and Human Services, CDC; 2021. https://www.cdc.gov/hiv/pdf/risk/prep/cdc-hiv-prep-guidelines-2021.pdf
  36. Memon S, Iversen L, Hannaford PC. Is the oral contraceptive pill associated with fracture in later life? New evidence from the Royal College of General Practitioners Oral Contraception Study. 2011;84:40–7. https://doi.org/10.1016/j.contraception.2010.11.019 PMID:21664509
  37. Vestergaard P, Rejnmark L, Mosekilde L. Fracture risk in very young women using combined oral contraceptives. 2008;78:358–64. https://doi.org/10.1016/j.contraception.2008.06.010 PMID:18929731
  38. Vestergaard P, Rejnmark L, Mosekilde L. Oral contraceptive use and risk of fractures. 2006;73:571–6. https://doi.org/10.1016/j.contraception.2006.01.006 PMID:16730486
  39. Barad D, Kooperberg C, Wactawski-Wende J, Liu J, Hendrix SL, Watts NB. Prior oral contraception and postmenopausal fracture: a Women’s Health Initiative observational cohort study. 2005;84:374–83. https://doi.org/10.1016/j.fertnstert.2005.01.132 PMID:16084878
  40. Michaëlsson K, Baron JA, Farahmand BY, Ljunghall S. Influence of parity and lactation on hip fracture risk. 2001;153:1166–72. https://doi.org/10.1093/aje/153.12.1166 PMID:11415951
  41. Michaëlsson K, Baron JA, Farahmand BY, Persson I, Ljunghall S. Oral-contraceptive use and risk of hip fracture: a case-control study. 1999;353:1481–4. https://doi.org/10.1016/S0140-6736(98)09044-8 PMID:10232314
  42. La Vecchia C, Tavani A, Gallus S. Oral contraceptives and risk of hip fractures. 1999;354:335–6. https://doi.org/10.1016/S0140-6736(05)75239-9 PMID:10440332
  43. Vessey M, Mant J, Painter R. Oral contraception and other factors in relation to hospital referral for fracture. Findings in a large cohort study. 1998;57:231–5. https://doi.org/10.1016/S0010-7824(98)00026-2 PMID:9649913
  44. O’Neill TW, Marsden D, Adams JE, Silman AJ. Risk factors, falls, and fracture of the distal forearm in Manchester, UK. 1996;50:288–92. https://doi.org/10.1136/jech.50.3.288 PMID:8935460
  45. Mallmin H, Ljunghall S, Persson I, Bergström R. Risk factors for fractures of the distal forearm: a population-based case-control study. 1994;4:298–304. https://doi.org/10.1007/BF01622186 PMID:7696821
  46. Cooper C, Hannaford P, Croft P, Kay CR. Oral contraceptive pill use and fractures in women: a prospective study. 1993;14:41–5. https://doi.org/10.1016/8756-3282(93)90254-8 PMID:8443001
  47. Meier C, Brauchli YB, Jick SS, Kraenzlin ME, Meier CR. Use of depot medroxyprogesterone acetate and fracture risk. 2010;95:4909–16. https://doi.org/10.1210/jc.2010-0032 PMID:20685865
  48. Cibula D, Skrenkova J, Hill M, Stepan JJ. Low-dose estrogen combined oral contraceptives may negatively influence physiological bone mineral density acquisition during adolescence. 2012;166:1003–11. https://doi.org/10.1530/EJE-11-1047 PMID:22436400
  49. Gai L, Jia Y, Zhang M, et al. Effect of two kinds of different combined oral contraceptives use on bone mineral density in adolescent women. 2012;86:332–6. https://doi.org/10.1016/j.contraception.2012.01.009 PMID:22364818
  50. Scholes D, Hubbard RA, Ichikawa LE, et al. Oral contraceptive use and bone density change in adolescent and young adult women: a prospective study of age, hormone dose, and discontinuation. 2011;96:E1380–7. https://doi.org/10.1210/jc.2010-3027 PMID:21752879
  51. Lattakova M, Borovsky M, Payer J, Killinger Z. Oral contraception usage in relation to bone mineral density and bone turnover in adolescent girls. 2009;14:207–14. https://doi.org/10.1080/13625180902838828 PMID:19565418
  52. Beksinska ME, Kleinschmidt I, Smit JA, Farley TMM. Bone mineral density in a cohort of adolescents during use of norethisterone enanthate, depot-medroxyprogesterone acetate or combined oral contraceptives and after discontinuation of norethisterone enanthate. 2009;79:345–9. https://doi.org/10.1016/j.contraception.2008.11.009 PMID:19341845
  53. Pikkarainen E, Lehtonen-Veromaa M, Möttönen T, Kautiainen H, Viikari J. Estrogen-progestin contraceptive use during adolescence prevents bone mass acquisition: a 4-year follow-up study. 2008;78:226–31. https://doi.org/10.1016/j.contraception.2008.05.002 PMID:18692613
  54. Cromer BA, Bonny AE, Stager M, et al. Bone mineral density in adolescent females using injectable or oral contraceptives: a 24-month prospective study. 2008;90:2060–7. https://doi.org/10.1016/j.fertnstert.2007.10.070 PMID:18222431
  55. Berenson AB, Rahman M, Breitkopf CR, Bi LX. Effects of depot medroxyprogesterone acetate and 20-microgram oral contraceptives on bone mineral density. 2008;112:788–99. https://doi.org/10.1097/AOG.0b013e3181875b78 PMID:18827121
  56. Harel Z, Riggs S, Vaz R, Flanagan P, Harel D, Machan JT. Bone accretion in adolescents using the combined estrogen and progestin transdermal contraceptive method Ortho Evra: a pilot study. 2010;23:23–31. https://doi.org/10.1016/j.jpag.2009.04.008 PMID:19647454
  57. Cobb KL, Bachrach LK, Sowers M, et al. The effect of oral contraceptives on bone mass and stress fractures in female runners. 2007;39:1464–73. https://doi.org/10.1249/mss.0b013e318074e532 PMID:17805075
  58. Beksinska ME, Kleinschmidt I, Smit JA, Farley TMM. Bone mineral density in adolescents using norethisterone enanthate, depot-medroxyprogesterone acetate or combined oral contraceptives for contraception. 2007;75:438–43. https://doi.org/10.1016/j.contraception.2007.02.001 PMID:17519149
  59. Lara-Torre E, Edwards CP, Perlman S, Hertweck SP. Bone mineral density in adolescent females using depot medroxyprogesterone acetate. 2004;17:17–21. https://doi.org/10.1016/j.jpag.2003.11.017 PMID:15010034
  60. Cromer BA, Blair JM, Mahan JD, Zibners L, Naumovski Z. A prospective comparison of bone density in adolescent girls receiving depot medroxyprogesterone acetate (Depo-Provera), levonorgestrel (Norplant), or oral contraceptives. 1996;129:671–6. https://doi.org/10.1016/S0022-3476(96)70148-8 PMID:8917232
  61. Polatti F, Perotti F, Filippa N, Gallina D, Nappi RE. Bone mass and long-term monophasic oral contraceptive treatment in young women. 1995;51:221–4. https://doi.org/10.1016/0010-7824(95)00036-A PMID:7796586
  62. Sørdal T, Grob P, Verhoeven C. Effects on bone mineral density of a monophasic combined oral contraceptive containing nomegestrol acetate/17β-estradiol in comparison to levonorgestrel/ethinylestradiol. 2012;91:1279–85. https://doi.org/10.1111/j.1600-0412.2012.01498.x PMID:22762147
  63. Gargano V, Massaro M, Morra I, Formisano C, Di Carlo C, Nappi C. Effects of two low-dose combined oral contraceptives containing drospirenone on bone turnover and bone mineral density in young fertile women: a prospective controlled randomized study. 2008;78:10–5. https://doi.org/10.1016/j.contraception.2008.01.016 PMID:18555812
  64. Nappi C, Di Spiezio Sardo A, Greco E, Tommaselli GA, Giordano E, Guida M. Effects of an oral contraceptive containing drospirenone on bone turnover and bone mineral density. 2005;105:53–60. https://doi.org/10.1097/01.AOG.0000148344.26475.fc PMID:15625142
  65. Berenson AB, Radecki CM, Grady JJ, Rickert VI, Thomas A. A prospective, controlled study of the effects of hormonal contraception on bone mineral density. 2001;98:576–82. https://doi.org/10.1016/S0029-7844(01)01495-8 PMID:11576570
  66. Berenson AB, Breitkopf CR, Grady JJ, Rickert VI, Thomas A. Effects of hormonal contraception on bone mineral density after 24 months of use. 2004;103:899–906. https://doi.org/10.1097/01.AOG.0000117082.49490.d5 PMID:15121563
  67. Elgán C, Samsioe G, Dykes AK. Influence of smoking and oral contraceptives on bone mineral density and bone remodeling in young women: a 2-year study. 2003;67:439–47. https://doi.org/10.1016/S0010-7824(03)00048-9 PMID:12814812
  68. Elgán C, Dykes AK, Samsioe G. Bone mineral density changes in young women: a two year study. 2004;19:169–77. https://doi.org/10.1080/09513590400012119 PMID:15724798
  69. Endrikat J, Mih E, Düsterberg B, et al. A 3-year double-blind, randomized, controlled study on the influence of two oral contraceptives containing either 20 microg or 30 microg ethinylestradiol in combination with levonorgestrel on bone mineral density. 2004;69:179–87. https://doi.org/10.1016/j.contraception.2003.10.002 PMID:14969664
  70. Paoletti AM, Orrù M, Lello S, et al. Short-term variations in bone remodeling markers of an oral contraception formulation containing 3 mg of drospirenone plus 30 microg of ethinyl estradiol: observational study in young postadolescent women. 2004;70:293–8. https://doi.org/10.1016/j.contraception.2004.04.004 PMID:15451333
  71. Nappi C, Di Spiezio Sardo A, Acunzo G, et al. Effects of a low-dose and ultra-low-dose combined oral contraceptive use on bone turnover and bone mineral density in young fertile women: a prospective controlled randomized study. 2003;67:355–9. https://doi.org/10.1016/S0010-7824(03)00025-8 PMID:12742557
  72. Reed SD, Scholes D, LaCroix AZ, Ichikawa LE, Barlow WE, Ott SM. Longitudinal changes in bone density in relation to oral contraceptive use. 2003;68:177–82. https://doi.org/10.1016/S0010-7824(03)00147-1 PMID:14561537
  73. Cobb KL, Kelsey JL, Sidney S, Ettinger B, Lewis CE. Oral contraceptives and bone mineral density in white and black women in CARDIA. Coronary Risk Development in Young Adults. 2002;13:893–900. https://doi.org/10.1007/s001980200123 PMID:12415437
  74. Burr DB, Yoshikawa T, Teegarden D, et al. Exercise and oral contraceptive use suppress the normal age-related increase in bone mass and strength of the femoral neck in women 18–31 years of age. 2000;27:855–63. https://doi.org/10.1016/S8756-3282(00)00403-8 PMID:11113398
  75. Recker RR, Davies KM, Hinders SM, Heaney RP, Stegman MR, Kimmel DB. Bone gain in young adult women. 1992;268:2403–8. https://doi.org/10.1001/jama.1992.03490170075028 PMID:1404797
  76. Mazess RB, Barden HS. Bone density in premenopausal women: effects of age, dietary intake, physical activity, smoking, and birth-control pills. 1991;53:132–42. https://doi.org/10.1093/ajcn/53.1.132 PMID:1984338
  77. Gambacciani M, Cappagli B, Lazzarini V, Ciaponi M, Fruzzetti F, Genazzani AR. Longitudinal evaluation of perimenopausal bone loss: effects of different low dose oral contraceptive preparations on bone mineral density. 2006;54:176–80. https://doi.org/10.1016/j.maturitas.2005.10.007 PMID:16332417
  78. Gambacciani M, Spinetti A, Taponeco F, Cappagli B, Piaggesi L, Fioretti P. Longitudinal evaluation of perimenopausal vertebral bone loss: effects of a low-dose oral contraceptive preparation on bone mineral density and metabolism. 1994;83:392–6. PMID:8127531
  79. Gambacciani M, Spinetti A, Cappagli B, et al. Hormone replacement therapy in perimenopausal women with a low dose oral contraceptive preparation: effects on bone mineral density and metabolism. 1994;19:125–31. https://doi.org/10.1016/0378-5122(94)90062-0 PMID:7968645
  80. Gambacciani M, Cappagli B, Ciaponi M, Benussi C, Genazzani AR. Hormone replacement therapy in perimenopause: effect of a low dose oral contraceptive preparation on bone quantitative ultrasound characteristics. 1999;6:43–8. https://doi.org/10.1097/00042192-199906010-00009 PMID:10100179
  81. Volpe A, Malmusi S, Zanni AL, Landi S, Cagnacci A. Oral contraceptives and bone metabolism. 1997;2:225–8. https://doi.org/10.3109/13625189709165298 PMID:9678077
  82. Hansen MA, Overgaard K, Riis BJ, Christiansen C. Potential risk factors for development of postmenopausal osteoporosis—examined over a 12-year period. 1991;1:95–102. https://doi.org/10.1007/BF01880450 PMID:1790399
  83. Shargil AA. Hormone replacement therapy in perimenopausal women with a triphasic contraceptive compound: a three-year prospective study. 1985;30:15–18–28, 18–28. PMID:2862116
  84. Taechakraichana N, Limpaphayom K, Ninlagarn T, Panyakhamlerd K, Chaikittisilpa S, Dusitsin N. A randomized trial of oral contraceptive and hormone replacement therapy on bone mineral density and coronary heart disease risk factors in postmenopausal women. 2000;95:87–94. PMID:10636509
  85. Taechakraichana N, Jaisamrarn U, Panyakhamlerd K, Chaikittisilpa S, Limpaphayom K. Difference in bone acquisition among hormonally treated postmenopausal women with normal and low bone mass. 2001;84(Suppl 2):S586–92. PMID:11853286
  86. Grimes DA, Schulz KF. Surrogate end points in clinical research: hazardous to your health. 2005;105:1114–8. https://doi.org/10.1097/01.AOG.0000157445.67309.19 PMID:15863552
  87. Schönau E. The peak bone mass concept: is it still relevant? 2004;19:825–31. https://doi.org/10.1007/s00467-004-1465-5 PMID:15197638
  88. Cohen A, Shane E. Treatment of premenopausal women with low bone mineral density. 2008;6:39–46. https://doi.org/10.1007/s11914-008-0007-7 PMID:18430399
  89. US Department of Agriculture; US Department of Health and Human Services. Dietary guidelines for Americans, 2020–2025. 9th ed. Washington, DC: US Department of Agriculture and US Department of Health and Human Services; 2020. https://www.dietaryguidelines.gov/sites/default/files/2021-03/Dietary_Guidelines_for_Americans-2020-2025.pdf
  90. Meek JY, Noble L; Section on Breastfeeding. Policy statement: breastfeeding and the use of human milk. 2022;150:e2022057988. https://doi.org/10.1542/peds.2022-057988 PMID:35921640
  91. Tepper NK, Phillips SJ, Kapp N, Gaffield ME, Curtis KM. Combined hormonal contraceptive use among breastfeeding women: an updated systematic review. 2016;94:262–74. https://doi.org/10.1016/j.contraception.2015.05.006 PMID:26002804
  92. Petersen JF, Bergholt T, Nielsen AK, Paidas MJ, Løkkegaard EC. Combined hormonal contraception and risk of venous thromboembolism within the first year following pregnancy. Danish nationwide historical cohort 1995–2009. 2014;112:73–8. https://doi.org/10.1160/TH13-09-0797 PMID:24499991
  93. Jackson E, Curtis KM, Gaffield ME. Risk of venous thromboembolism during the postpartum period: a systematic review. 2011;117:691–703. https://doi.org/10.1097/AOG.0b013e31820ce2db PMID:21343773
  94. Kamel H, Navi BB, Sriram N, Hovsepian DA, Devereux RB, Elkind MS. Risk of a thrombotic event after the 6-week postpartum period. 2014;370:1307–15. https://doi.org/10.1056/NEJMoa1311485 PMID:24524551
  95. Sultan AA, Tata LJ, West J, et al. Risk factors for first venous thromboembolism around pregnancy: a population-based cohort study from the United Kingdom. 2013;121:3953–61. https://doi.org/10.1182/blood-2012-11-469551 PMID:23550034
  96. Sultan AA, West J, Tata LJ, Fleming KM, Nelson-Piercy C, Grainge MJ. Risk of first venous thromboembolism in and around pregnancy: a population-based cohort study. 2012;156:366–73. https://doi.org/10.1111/j.1365-2141.2011.08956.x PMID:22145820
  97. Tepper NK, Boulet SL, Whiteman MK, et al. Postpartum venous thromboembolism: incidence and risk factors. 2014;123:987–96. https://doi.org/10.1097/AOG.0000000000000230 PMID:24785851
  98. Jackson E, Glasier A. Return of ovulation and menses in postpartum nonlactating women: a systematic review. 2011;117:657–62. https://doi.org/10.1097/AOG.0b013e31820ce18c PMID:21343770
  99. Kim C, Nguyen AT, Berry-Bibee E, Ermias Y, Gaffield ME, Kapp N. Systemic hormonal contraception initiation after abortion: a systematic review and meta-analysis. 2021;103:291–304. https://doi.org/10.1016/j.contraception.2021.01.017 PMID:33548267
  100. Gaffield ME, Kapp N, Ravi A. Use of combined oral contraceptives post abortion. 2009;80:355–62. https://doi.org/10.1016/j.contraception.2009.04.005 PMID:19751858
  101. Gillum LA, Mamidipudi SK, Johnston SC. Ischemic stroke risk with oral contraceptives: A meta-analysis. 2000;284:72–8. https://doi.org/10.1001/jama.284.1.72 PMID:10872016
  102. Jick SS, Walker AM, Stergachis A, Jick H. Oral contraceptives and breast cancer. 1989;59:618–21. https://doi.org/10.1038/bjc.1989.125 PMID:2713248
  103. Khader YS, Rice J, John L, Abueita O. Oral contraceptives use and the risk of myocardial infarction: a meta-analysis. 2003;68:11–7. https://doi.org/10.1016/S0010-7824(03)00073-8 PMID:12878281
  104. Lawson DH, Davidson JF, Jick H. Oral contraceptive use and venous thromboembolism: absence of an effect of smoking. 1977;2:729–30. https://doi.org/10.1136/bmj.2.6089.729 PMID:334332
  105. Lidegaard O, Edström B, Kreiner S. Oral contraceptives and venous thromboembolism. A case-control study. 1998;57:291–301. https://doi.org/10.1016/S0010-7824(98)00033-X PMID:9673836
  106. Nightingale AL, Lawrenson RA, Simpson EL, Williams TJ, MacRae KD, Farmer RD. The effects of age, body mass index, smoking and general health on the risk of venous thromboembolism in users of combined oral contraceptives. 2000;5:265–74. https://doi.org/10.1080/13625180008500402 PMID:11245554
  107. Petitti DB, Wingerd J, Pellegrin F, Ramcharan S. Risk of vascular disease in women. Smoking, oral contraceptives, noncontraceptive estrogens, and other factors. 1979;242:1150–4. https://doi.org/10.1001/jama.1979.03300110022020 PMID:470067
  108. Rosenberg L, Palmer JR, Rao RS, Shapiro S. Low-dose oral contraceptive use and the risk of myocardial infarction. 2001;161:1065–70. https://doi.org/10.1001/archinte.161.8.1065 PMID:11322840
  109. Straneva P, Hinderliter A, Wells E, Lenahan H, Girdler S. Smoking, oral contraceptives, and cardiovascular reactivity to stress. 2000;95:78–83. https://doi.org/10.1016/S0029-7844(99)00497-4 PMID:10636507
  110. Tanis BC, van den Bosch MA, Kemmeren JM, et al. Oral contraceptives and the risk of myocardial infarction. 2001;345:1787–93. https://doi.org/10.1056/NEJMoa003216 PMID:11752354
  111. Van den bosch MAAJ, Kemmeren JM, Tanis BC, et al. The RATIO study: oral contraceptives and the risk of peripheral arterial disease in young women. J Thromb Hemost 2003;1:439–44. https://doi.org/10.1046/j.1538-7836.2003.00079.x PMID:12871447
  112. World Health Organization Collaborative Study of Cardiovascular Disease and Steroid Hormone Contraception. Venous thromboembolic disease and combined oral contraceptives: results of international multicentre case-control study. 1995;346:1575–82. https://doi.org/10.1016/S0140-6736(95)91926-0 PMID:7500748
  113. Collaborative Group for the Study of Stroke in Young Women. Oral contraceptives and stroke in young women. Associated risk factors. 1975;231:718–22. https://doi.org/10.1001/jama.1975.03240190022010 PMID:1172861
  114. Horton LG, Simmons KB, Curtis KM. Combined hormonal contraceptive use among obese women and risk for cardiovascular events: a systematic review. 2016;94:590–604. https://doi.org/10.1016/j.contraception.2016.05.014 PMID:27263039
  115. Schink T, Princk C, Braitmaier M, Haug U. Use of combined oral contraceptives and risk of venous thromboembolism in young women: a nested case-control analysis using German claims data. 2022;129:2107–16. https://doi.org/10.1111/1471-0528.17268 PMID:35876787
  116. Traven SA, McGurk KM, Althoff AD, et al. Resident level involvement affects operative time and surgical complications in lower extremity fracture care. 2021;78:1755–61. https://doi.org/10.1016/j.jsurg.2021.03.004 PMID:33903063
  117. Brunner Huber LR, Hogue CJ, Stein AD, Drews C, Zieman M. Body mass index and risk for oral contraceptive failure: a case-cohort study in South Carolina. 2006;16:637–43. https://doi.org/10.1016/j.annepidem.2006.01.001 PMID:16516489
  118. Brunner Huber LR, Toth JL. Obesity and oral contraceptive failure: findings from the 2002 National Survey of Family Growth. 2007;166:1306–11. https://doi.org/10.1093/aje/kwm221 PMID:17785712
  119. Brunner LR, Hogue CJ. The role of body weight in oral contraceptive failure: results from the 1995 national survey of family growth. 2005;15:492–9. https://doi.org/10.1016/j.annepidem.2004.10.009 PMID:16029841
  120. Holt VL, Cushing-Haugen KL, Daling JR. Body weight and risk of oral contraceptive failure. 2002;99:820–7. https://doi.org/10.1016/S0029-7844(02)01939-7 PMID:11978293
  121. Holt VL, Scholes D, Wicklund KG, Cushing-Haugen KL, Daling JR. Body mass index, weight, and oral contraceptive failure risk. 2005;105:46–52. https://doi.org/10.1097/01.AOG.0000149155.11912.52 PMID:15625141
  122. Trussell J, Schwarz EB, Guthrie K. Obesity and oral contraceptive pill failure. 2009;79:334–8. https://doi.org/10.1016/j.contraception.2008.11.017 PMID:19341843
  123. Vessey M. Oral contraceptive failures and body weight: findings in a large cohort study. 2001;27:90–1. https://doi.org/10.1783/147118901101195092 PMID:12457519
  124. Yamazaki M, Dwyer K, Sobhan M, et al. Effect of obesity on the effectiveness of hormonal contraceptives: an individual participant data meta-analysis. 2015;92:445–52. https://doi.org/10.1016/j.contraception.2015.07.016 PMID:26247330
  125. Dragoman MV, Simmons KB, Paulen ME, Curtis KM. Combined hormonal contraceptive (CHC) use among obese women and contraceptive effectiveness: a systematic review. 2017;95:117–29. https://doi.org/10.1016/j.contraception.2016.10.010 PMID:27823942
  126. Paulen ME, Zapata LB, Cansino C, Curtis KM, Jamieson DJ. Contraceptive use among women with a history of bariatric surgery: a systematic review. 2010;82:86–94. https://doi.org/10.1016/j.contraception.2010.02.008 PMID:20682146
  127. Curtis KM, Nguyen AT, Tepper NK, et al. U.S. selected practice recommendations for contraceptive use, 2024. 2024;73(No. RR-3):1–77.
  128. Lidegaard Ø, Edström B, Kreiner S. Oral contraceptives and venous thromboembolism: a five-year national case-control study. 2002;65:187–96. https://doi.org/10.1016/S0010-7824(01)00307-9 PMID:11929640
  129. Croft P, Hannaford PC. Risk factors for acute myocardial infarction in women: evidence from the Royal College of General Practitioners’ oral contraception study. 1989;298:165–8. https://doi.org/10.1136/bmj.298.6667.165 PMID:2493841
  130. D’Avanzo B, La Vecchia C, Negri E, Parazzini F, Franceschi S. Oral contraceptive use and risk of myocardial infarction: an Italian case-control study. 1994;48:324–5. https://doi.org/10.1136/jech.48.3.324 PMID:8051537
  131. Dunn NR, Faragher B, Thorogood M, et al. Risk of myocardial infarction in young female smokers. 1999;82:581–3. https://doi.org/10.1136/hrt.82.5.581 PMID:10525513
  132. Hannaford PC, Croft PR, Kay CR. Oral contraception and stroke. Evidence from the Royal College of General Practitioners’ Oral Contraception Study. 1994;25:935–42. https://doi.org/10.1161/01.STR.25.5.935 PMID:8165687
  133. Heinemann LA, Lewis MA, Spitzer WO, Thorogood M, Guggenmoos-Holzmann I, Bruppacher R; Transnational Research Group on Oral Contraceptives and the Health of Young Women. Thromboembolic stroke in young women. A European case-control study on oral contraceptives. 1998;57:29–37. https://doi.org/10.1016/S0010-7824(97)00204-7 PMID:9554248
  134. Kemmeren JM, Tanis BC, van den Bosch MA, et al. Risk of Arterial Thrombosis in Relation to Oral Contraceptives (RATIO) study: oral contraceptives and the risk of ischemic stroke. 2002;33:1202–8. https://doi.org/10.1161/01.STR.0000015345.61324.3F PMID:11988591
  135. Lewis MA, Heinemann LA, Spitzer WO, MacRae KD, Bruppacher R. The use of oral contraceptives and the occurrence of acute myocardial infarction in young women. Results from the Transnational Study on Oral Contraceptives and the Health of Young Women. 1997;56:129–40. https://doi.org/10.1016/S0010-7824(97)00118-2 PMID:9347202
  136. Lidegaard O. Oral contraception and risk of a cerebral thromboembolic attack: results of a case-control study. 1993;306:956–63. https://doi.org/10.1136/bmj.306.6883.956 PMID:8490470
  137. Lidegaard O. Oral contraceptives, pregnancy and the risk of cerebral thromboembolism: the influence of diabetes, hypertension, migraine and previous thrombotic disease. 1995;102:153–9. https://doi.org/10.1111/j.1471-0528.1995.tb09070.x PMID:7756208
  138. Lubianca JN, Faccin CS, Fuchs FD. Oral contraceptives: a risk factor for uncontrolled blood pressure among hypertensive women. 2003;67:19–24. https://doi.org/10.1016/S0010-7824(02)00429-8 PMID:12521653
  139. Narkiewicz K, Graniero GR, D’Este D, Mattarei M, Zonzin P, Palatini P. Ambulatory blood pressure in mild hypertensive women taking oral contraceptives. A case-control study. 1995;8:249–53. https://doi.org/10.1016/0895-7061(95)96212-3 PMID:7794573
  140. Siritho S, Thrift AG, McNeil JJ, You RX, Davis SM, Donnan GA; Melbourne Risk Factor Study (MERFS) Group. Risk of ischemic stroke among users of the oral contraceptive pill: The Melbourne Risk Factor Study (MERFS) Group. 2003;34:1575–80. https://doi.org/10.1161/01.STR.0000077925.16041.6B PMID:12805499
  141. WHO Collaborative Study of Cardiovascular Disease and Steroid Hormone Contraception. Ischaemic stroke and combined oral contraceptives: results of an international, multicentre, case-control study. 1996;348:498–505. https://doi.org/10.1016/S0140-6736(95)12393-8 PMID:8757151
  142. WHO Collaborative Study of Cardiovascular Disease and Steroid Hormone Contraception. Acute myocardial infarction and combined oral contraceptives: results of an international multicentre case-control study. 1997;349:1202–9. https://doi.org/10.1016/S0140-6736(97)02358-1 PMID:9130941
  143. Lubianca JN, Moreira LB, Gus M, Fuchs FD. Stopping oral contraceptives: an effective blood pressure-lowering intervention in women with hypertension. 2005;19:451–5. https://doi.org/10.1038/sj.jhh.1001841 PMID:15759027
  144. Aberg H, Karlsson L, Melander S. Studies on toxaemia of pregnancy with special reference to blood pressure. II. Results after 6-11 years’ follow-up. 1978;83:97–102. https://doi.org/10.3109/03009737809179119 PMID:664120
  145. Carmichael SM, Taylor MM, Ayers CR. Oral contraceptives, hypertension, and toxemia. 1970;35:371–6. PMID:4190423
  146. Meinel H, Ihle R, Laschinski M. [Effect of hormonal contraceptives on blood pressure following pregnancy-induced hypertension]. 1987;109:527–31. PMID:3604495
  147. Pritchard JA, Pritchard SA. Blood pressure response to estrogen-progestin oral contraceptive after pregnancy-induced hypertension. 1977;129:733–9. https://doi.org/10.1016/0002-9378(77)90390-8 PMID:607805
  148. Sibai BM, Taslimi MM, el-Nazer A, Amon E, Mabie BC, Ryan GM. Maternal-perinatal outcome associated with the syndrome of hemolysis, elevated liver enzymes, and low platelets in severe preeclampsia-eclampsia. 1986;155:501–9. https://doi.org/10.1016/0002-9378(86)90266-8 PMID:3529964
  149. Sibai BM, Ramadan MK, Chari RS, Friedman SA. Pregnancies complicated by HELLP syndrome (hemolysis, elevated liver enzymes, and low platelets): subsequent pregnancy outcome and long-term prognosis. 1995;172:125–9. https://doi.org/10.1016/0002-9378(95)90099-3 PMID:7847520
  150. WHO Collaborative Study of Cardiovascular Disease and Steroid Hormone Contraception. Haemorrhagic stroke, overall stroke risk, and combined oral contraceptives: results of an international, multicentre, case-control study. 1996;348:505–10. https://doi.org/10.1016/S0140-6736(95)12394-6 PMID:8757152
  151. Martinelli I, Lensing AW, Middeldorp S, et al. Recurrent venous thromboembolism and abnormal uterine bleeding with anticoagulant and hormone therapy use. 2016;127:1417–25. https://doi.org/10.1182/blood-2015-08-665927 PMID:26696010
  152. Barbhaiya M, Zuily S, Naden R, et al. ; ACR/EULAR APS Classification Criteria Collaborators. The 2023 ACR/EULAR Antiphospholipid Syndrome Classification Criteria. 2023;75:1687–702. https://doi.org/10.1002/art.42624 PMID:37635643
  153. Tepper NK, Marchbanks PA, Curtis KM. Superficial venous disease and combined hormonal contraceptives: a systematic review. 2016;94:275–9. https://doi.org/10.1016/j.contraception.2015.03.010 PMID:25835269
  154. Tepper NK, Paulen ME, Marchbanks PA, Curtis KM. Safety of contraceptive use among women with peripartum cardiomyopathy: a systematic review. 2010;82:95–101. https://doi.org/10.1016/j.contraception.2010.02.004 PMID:20682147
  155. The Criteria Committee of the New York Heart Association. Nomenclature and criteria for diagnosis of diseases of the heart and great vessels. 9th ed. Boston, MA: Little, Brown and Co; 1994.
  156. Molnar AO, Bota SE, McArthur E, et al. Risk and complications of venous thromboembolism in dialysis patients. 2018;33:874–80. https://doi.org/10.1093/ndt/gfx212 PMID:28992258
  157. Tveit DP, Hypolite IO, Hshieh P, et al. Chronic dialysis patients have high risk for pulmonary embolism. 2002;39:1011–7. https://doi.org/10.1053/ajkd.2002.32774 PMID:11979344
  158. Wang IK, Shen TC, Muo CH, Yen TH, Sung FC. Risk of pulmonary embolism in patients with end-stage renal disease receiving long-term dialysis. 2017;32:1386–93. PMID:27448674
  159. Bernatsky S, Clarke A, Ramsey-Goldman R, et al. Hormonal exposures and breast cancer in a sample of women with systemic lupus erythematosus. 2004;43:1178–81. https://doi.org/10.1093/rheumatology/keh282 PMID:15226516
  160. Bernatsky S, Ramsey-Goldman R, Gordon C, et al. Factors associated with abnormal Pap results in systemic lupus erythematosus. 2004;43:1386–9. https://doi.org/10.1093/rheumatology/keh331 PMID:15280571
  161. Chopra N, Koren S, Greer WL, et al. Factor V Leiden, prothrombin gene mutation, and thrombosis risk in patients with antiphospholipid antibodies. 2002;29:1683–8. PMID:12180730
  162. Esdaile JM, Abrahamowicz M, Grodzicky T, et al. Traditional Framingham risk factors fail to fully account for accelerated atherosclerosis in systemic lupus erythematosus. 2001;44:2331–7. https://doi.org/10.1002/1529-0131(200110)44:10<2331::AID-ART395>3.0.CO;2-I PMID:11665973
  163. Julkunen HA. Oral contraceptives in systemic lupus erythematosus: side-effects and influence on the activity of SLE. 1991;20:427–33. https://doi.org/10.3109/03009749109096822 PMID:1771400
  164. Julkunen HA, Kaaja R, Friman C. Contraceptive practice in women with systemic lupus erythematosus. 1993;32:227–30. https://doi.org/10.1093/rheumatology/32.3.227 PMID:8448613
  165. Jungers P, Dougados M, Pélissier C, et al. Influence of oral contraceptive therapy on the activity of systemic lupus erythematosus. 1982;25:618–23. https://doi.org/10.1002/art.1780250603 PMID:7092961
  166. Manzi S, Meilahn EN, Rairie JE, et al. Age-specific incidence rates of myocardial infarction and angina in women with systemic lupus erythematosus: comparison with the Framingham Study. 1997;145:408–15. https://doi.org/10.1093/oxfordjournals.aje.a009122 PMID:9048514
  167. McAlindon T, Giannotta L, Taub N, D’Cruz D, Hughes G. Environmental factors predicting nephritis in systemic lupus erythematosus. 1993;52:720–4. https://doi.org/10.1136/ard.52.10.720 PMID:8257208
  168. McDonald J, Stewart J, Urowitz MB, Gladman DD. Peripheral vascular disease in patients with systemic lupus erythematosus. 1992;51:56–60. https://doi.org/10.1136/ard.51.1.56 PMID:1540039
  169. Mintz G, Gutiérrez G, Delezé M, Rodríguez E. Contraception with progestagens in systemic lupus erythematosus. 1984;30:29–38. https://doi.org/10.1016/0010-7824(84)90076-3 PMID:6434228
  170. Petri M. Musculoskeletal complications of systemic lupus erythematosus in the Hopkins Lupus Cohort: an update. 1995;8:137–45. https://doi.org/10.1002/art.1790080305 PMID:7654797
  171. Petri M. Lupus in Baltimore: evidence-based ‘clinical pearls’ from the Hopkins Lupus Cohort. 2005;14:970–3. https://doi.org/10.1191/0961203305lu2230xx PMID:16425579
  172. Petri M, Kim MY, Kalunian KC, et al. ; OC-SELENA Trial. Combined oral contraceptives in women with systemic lupus erythematosus. 2005;353:2550–8. https://doi.org/10.1056/NEJMoa051135 PMID:16354891
  173. Sánchez-Guerrero J, Uribe AG, Jiménez-Santana L, et al. A trial of contraceptive methods in women with systemic lupus erythematosus. 2005;353:2539–49. https://doi.org/10.1056/NEJMoa050817 PMID:16354890
  174. Sarabi ZS, Chang E, Bobba R, et al. Incidence rates of arterial and venous thrombosis after diagnosis of systemic lupus erythematosus. 2005;53:609–12. https://doi.org/10.1002/art.21314 PMID:16082635
  175. Schaedel ZE, Dolan G, Powell MC. The use of the levonorgestrel-releasing intrauterine system in the management of menorrhagia in women with hemostatic disorders. 2005;193:1361–3. https://doi.org/10.1016/j.ajog.2005.05.002 PMID:16202726
  176. Somers E, Magder LS, Petri M. Antiphospholipid antibodies and incidence of venous thrombosis in a cohort of patients with systemic lupus erythematosus. 2002;29:2531–6. PMID:12465147
  177. Urowitz MB, Bookman AA, Koehler BE, Gordon DA, Smythe HA, Ogryzlo MA. The bimodal mortality pattern of systemic lupus erythematosus. 1976;60:221–5. https://doi.org/10.1016/0002-9343(76)90431-9 PMID:1251849
  178. Choojitarom K, Verasertniyom O, Totemchokchyakarn K, Nantiruj K, Sumethkul V, Janwityanujit S. Lupus nephritis and Raynaud’s phenomenon are significant risk factors for vascular thrombosis in SLE patients with positive antiphospholipid antibodies. 2008;27:345–51. https://doi.org/10.1007/s10067-007-0721-z PMID:17805483
  179. Wahl DG, Guillemin F, de Maistre E, Perret C, Lecompte T, Thibaut G. Risk for venous thrombosis related to antiphospholipid antibodies in systemic lupus erythematosus—a meta-analysis. 1997;6:467–73. https://doi.org/10.1177/096120339700600510 PMID:9229367
  180. Farr SL, Folger SG, Paulen ME, Curtis KM. Safety of contraceptive methods for women with rheumatoid arthritis: a systematic review. 2010;82:64–71. https://doi.org/10.1016/j.contraception.2010.02.003 PMID:20682144
  181. Headache Classification Committee of the International Headache Society (IHS). The International Classification of Headache Disorders. 3rd edition. Cephalalgia 2018;38:1–211. https://www.ichd-3.org/wp-content/uploads/2018/01/The-International-Classification-of-Headache-Disorders-3rd-Edition-2018.pdf
  182. Tepper NK, Whiteman MK, Zapata LB, Marchbanks PA, Curtis KM. Safety of hormonal contraceptives among women with migraine: a systematic review. 2016;94:630–40. https://doi.org/10.1016/j.contraception.2016.04.016 PMID:27153744
  183. Xu Z, Li Y, Tang S, Huang X, Chen T. Current use of oral contraceptives and the risk of first-ever ischemic stroke: a meta-analysis of observational studies. 2015;136:52–60. https://doi.org/10.1016/j.thromres.2015.04.021 PMID:25936231
  184. Etminan M, Takkouche B, Isorna FC, Samii A. Risk of ischaemic stroke in people with migraine: systematic review and meta-analysis of observational studies. 2005;330:63. https://doi.org/10.1136/bmj.38302.504063.8F PMID:15596418
  185. Schürks M, Rist PM, Bigal ME, Buring JE, Lipton RB, Kurth T. Migraine and cardiovascular disease: systematic review and meta-analysis. 2009;339(oct27 1):b3914. https://doi.org/10.1136/bmj.b3914 PMID:19861375
  186. Spector JT, Kahn SR, Jones MR, Jayakumar M, Dalal D, Nazarian S. Migraine headache and ischemic stroke risk: an updated meta-analysis. 2010;123:612–24. https://doi.org/10.1016/j.amjmed.2009.12.021 PMID:20493462
  187. Zapata LB, Oduyebo T, Whiteman MK, Houtchens MK, Marchbanks PA, Curtis KM. Contraceptive use among women with multiple sclerosis: a systematic review. 2016;94:612–20. https://doi.org/10.1016/j.contraception.2016.07.013 PMID:27452316
  188. Pagano HP, Zapata LB, Berry-Bibee EN, Nanda K, Curtis KM. Safety of hormonal contraception and intrauterine devices among women with depressive and bipolar disorders: a systematic review. 2016;94:641–9. https://doi.org/10.1016/j.contraception.2016.06.012 PMID:27364100
  189. Iyer V, Farquhar C, Jepson R. Oral contraceptive pills for heavy menstrual bleeding. 2000;(2):CD000154. https://doi.org/10.1002/14651858.CD000154 PMID:10796696
  190. Davis L, Kennedy SS, Moore J, Prentice A. Oral contraceptives for pain associated with endometriosis. 2007;(3):CD001019. https://doi.org/10.1002/14651858.CD001019.pub2 PMID:17636650
  191. Hendrix SL, Alexander NJ. Primary dysmenorrhea treatment with a desogestrel-containing low-dose oral contraceptive. 2002;66:393–9. https://doi.org/10.1016/S0010-7824(02)00414-6 PMID:12499030
  192. Wong CL, Farquhar C, Roberts H. Oral contraceptive pills for primary dysmenorrhoea. 2001;(2). https://doi.org/10.1002/14651858.CD002120.pub3
  193. Gaffield ME, Kapp N, Curtis KM. Combined oral contraceptive and intrauterine device use among women with gestational trophoblastic disease. 2009;80:363–71. https://doi.org/10.1016/j.contraception.2009.03.022 PMID:19751859
  194. Smith JS, Green J, Berrington de Gonzalez A, et al. Cervical cancer and use of hormonal contraceptives: a systematic review. 2003;361:1159–67. https://doi.org/10.1016/S0140-6736(03)12949-2 PMID:12686037
  195. Black MM, Barclay THC, Polednak A, Kwon CS, Leis HP Jr, Pilnik S. Family history, oral contraceptive usage, and breast cancer. 1983;51:2147–51. https://doi.org/10.1002/1097-0142(19830601)51:11<2147::AID-CNCR2820511133>3.0.CO;2-X PMID:6839302
  196. Brinton LA, Hoover R, Szklo M, Fraumeni JF Jr. Oral contraceptives and breast cancer. 1982;11:316–22. https://doi.org/10.1093/ije/11.4.316 PMID:7152784
  197. Brohet RM, Goldgar DE, Easton DF, et al. Oral contraceptives and breast cancer risk in the international BRCA1/2 carrier cohort study: a report from EMBRACE, GENEPSO, GEO-HEBON, and the IBCCS Collaborating Group. 2007;25:3831–6. https://doi.org/10.1200/JCO.2007.11.1179 PMID:17635951
  198. Claus EB, Stowe M, Carter D. Oral contraceptives and the risk of ductal breast carcinoma in situ. 2003;81:129–36. https://doi.org/10.1023/A:1025728524310 PMID:14572155
  199. Collaborative Group on Hormonal Factors in Breast Cancer. Familial breast cancer: collaborative reanalysis of individual data from 52 epidemiological studies including 58,209 women with breast cancer and 101,986 women without the disease. 2001;358:1389–99. https://doi.org/10.1016/S0140-6736(01)06524-2 PMID:11705483
  200. Grabrick DM, Hartmann LC, Cerhan JR, et al. Risk of breast cancer with oral contraceptive use in women with a family history of breast cancer. 2000;284:1791–8. https://doi.org/10.1001/jama.284.14.1791 PMID:11025831
  201. Gronwald J, Byrski T, Huzarski T, et al. Influence of selected lifestyle factors on breast and ovarian cancer risk in BRCA1 mutation carriers from Poland. 2006;95:105–9. https://doi.org/10.1007/s10549-005-9051-5 PMID:16261399
  202. Haile RW, Thomas DC, McGuire V, et al. ; kConFab Investigators; Ontario Cancer Genetics Network Investigators. BRCA1 and BRCA2 mutation carriers, oral contraceptive use, and breast cancer before age 50. 2006;15:1863–70. https://doi.org/10.1158/1055-9965.EPI-06-0258 PMID:17021353
  203. Harris NV, Weiss NS, Francis AM, Polissar L. Breast cancer in relation to patterns of oral contraceptive use. 1982;116:643–51. https://doi.org/10.1093/oxfordjournals.aje.a113447 PMID:7137151
  204. Hennekens CH, Speizer FE, Lipnick RJ, et al. A case-control study of oral contraceptive use and breast cancer. 1984;72:39–42. https://doi.org/10.1093/jnci/72.1.39 PMID:6363789
  205. Jernström H, Loman N, Johannsson OT, Borg A, Olsson H. Impact of teenage oral contraceptive use in a population-based series of early-onset breast cancer cases who have undergone BRCA mutation testing. 2005;41:2312–20. https://doi.org/10.1016/j.ejca.2005.03.035 PMID:16118051
  206. Marchbanks PA, McDonald JA, Wilson HG, et al. Oral contraceptives and the risk of breast cancer. 2002;346:2025–32. https://doi.org/10.1056/NEJMoa013202 PMID:12087137
  207. Milne RL, Knight JA, John EM, et al. Oral contraceptive use and risk of early-onset breast cancer in carriers and noncarriers of BRCA1 and BRCA2 mutations. 2005;14:350–6. https://doi.org/10.1158/1055-9965.EPI-04-0376 PMID:15734957
  208. Narod SA, Dubé MP, Klijn J, et al. Oral contraceptives and the risk of breast cancer in BRCA1 and BRCA2 mutation carriers. 2002;94:1773–9. https://doi.org/10.1093/jnci/94.23.1773 PMID:12464649
  209. Rosenberg L, Palmer JR, Rao RS, et al. Case-control study of oral contraceptive use and risk of breast cancer. 1996;143:25–37. https://doi.org/10.1093/oxfordjournals.aje.a008654 PMID:8533744
  210. Silvera SAN, Miller AB, Rohan TE. Oral contraceptive use and risk of breast cancer among women with a family history of breast cancer: a prospective cohort study. 2005;16:1059–63. https://doi.org/10.1007/s10552-005-0343-1 PMID:16184471
  211. Ursin G, Henderson BE, Haile RW, et al. Does oral contraceptive use increase the risk of breast cancer in women with BRCA1/BRCA2 mutations more than in other women? 1997;57:3678–81. PMID:9288771
  212. Ursin G, Ross RK, Sullivan-Halley J, Hanisch R, Henderson B, Bernstein L. Use of oral contraceptives and risk of breast cancer in young women. 1998;50:175–84. https://doi.org/10.1023/A:1006037823178 PMID:9822222
  213. Curtis KM, Hannaford PC, Rodriguez MI, Chipato T, Steyn PS, Kiarie JN. Hormonal contraception and HIV acquisition among women: an updated systematic review. 2020;46:8–16. https://doi.org/10.1136/bmjsrh-2019-200509 PMID:31919239
  214. Tepper NK, Curtis KM, Cox S, Whiteman MK. Update to U.S. medical eligibility criteria for contraceptive use, 2016: updated recommendations for the use of contraception among women at high risk for HIV infection. 2020;69:405–10. https://doi.org/10.15585/mmwr.mm6914a3 PMID:32271729
  215. Polis CB, Phillips SJ, Curtis KM. Hormonal contraceptive use and female-to-male HIV transmission: a systematic review of the epidemiologic evidence. 2013;27:493–505. https://doi.org/10.1097/QAD.0b013e32835ad539 PMID:23079808
  216. Phillips SJ, Polis CB, Curtis KM. The safety of hormonal contraceptives for women living with HIV and their sexual partners. 2016;93:11–6. https://doi.org/10.1016/j.contraception.2015.10.002 PMID:26515194
  217. Phillips SJ, Curtis KM, Polis CB. Effect of hormonal contraceptive methods on HIV disease progression: a systematic review. 2013;27:787–94. https://doi.org/10.1097/QAD.0b013e32835bb672 PMID:23135169
  218. el-Raghy I, Back DJ, Osman F, Orme ML, Fathalla M. Contraceptive steroid concentrations in women with early active schistosomiasis: lack of effect of antischistosomal drugs. 1986;33:373–7. https://doi.org/10.1016/0010-7824(86)90099-5 PMID:3089682
  219. Gad-el-Mawla N, Abdallah A. Liver function in bilharzial females receiving contraceptive pills. 1969;16:308–10. PMID:5358869
  220. Gad-el-Mawla N, el-Roubi O, Sabet S, Abdallah A. Plasma lipids and lipoproteins in bilharzial females during oral contraceptive therapy. 1972;55:137–47. PMID:5051373
  221. Shaaban MM, Hammad WA, Falthalla MF, et al. Effects of oral contraception on liver function tests and serum proteins in women with active schistosomiasis. 1982;26:75–82. https://doi.org/10.1016/0010-7824(82)90174-3 PMID:7128137
  222. Shaaban MM, Ghaneimah SA, Mohamed MA, Abdel-Chani S, Mostafa SA. Effect of oral contraception on serum bile acid. 1984;22:111–5. https://doi.org/10.1016/0020-7292(84)90023-7 PMID:6145634
  223. Sy FS, Osteria TS, Opiniano V, Gler S. Effect of oral contraceptive on liver function tests of women with schistosomiasis in the Philippines. 1986;34:283–94. https://doi.org/10.1016/0010-7824(86)90009-0 PMID:3098499
  224. Tagy AH, Saker ME, Moussa AA, Kolgah A. The effect of low-dose combined oral contraceptive pills versus injectable contraceptive (Depot Provera) on liver function tests of women with compensated bilharzial liver fibrosis. 2001;64:173–6. https://doi.org/10.1016/S0010-7824(01)00248-7 PMID:11704097
  225. Beck P, Wells SA. Comparison of the mechanisms underlying carbohydrate intolerance in subclinical diabetic women during pregnancy and during post-partum oral contraceptive steroid treatment. 1969;29:807–18. https://doi.org/10.1210/jcem-29-6-807 PMID:4978229
  226. Kjos SL, Peters RK, Xiang A, Thomas D, Schaefer U, Buchanan TA. Contraception and the risk of type 2 diabetes mellitus in Latina women with prior gestational diabetes mellitus. 1998;280:533–8. https://doi.org/10.1001/jama.280.6.533 PMID:9707143
  227. Kung AW, Ma JT, Wong VC, et al. Glucose and lipid metabolism with triphasic oral contraceptives in women with history of gestational diabetes. 1987;35:257–69. https://doi.org/10.1016/0010-7824(87)90027-8 PMID:3111786
  228. Rådberg T, Gustafson A, Skryten A, Karlsson K. Metabolic studies in gestational diabetic women during contraceptive treatment: effects on glucose tolerance and fatty acid composition of serum lipids. 1982;13:17–29. https://doi.org/10.1159/000299480 PMID:7035304
  229. Skouby SO, Andersen O, Kühl C. Oral contraceptives and insulin receptor binding in normal women and those with previous gestational diabetes. 1986;155:802–7. https://doi.org/10.1016/S0002-9378(86)80024-2 PMID:3766633
  230. Skouby SO, Andersen O, Saurbrey N, Kühl C. Oral contraception and insulin sensitivity: in vivo assessment in normal women and women with previous gestational diabetes. 1987;64:519–23. https://doi.org/10.1210/jcem-64-3-519 PMID:3102539
  231. Skouby SO, Mølsted-Pedersen L, Kühl C. Low dosage oral contraception in women with previous gestational diabetes. 1982;59:325–8. PMID:6804901
  232. Xiang AH, Kawakubo M, Kjos SL, Buchanan TA. Long-acting injectable progestin contraception and risk of type 2 diabetes in Latino women with prior gestational diabetes mellitus. 2006;29:613–7. https://doi.org/10.2337/diacare.29.03.06.dc05-1940 PMID:16505515
  233. Kjos SL, Shoupe D, Douyan S, et al. Effect of low-dose oral contraceptives on carbohydrate and lipid metabolism in women with recent gestational diabetes: results of a controlled, randomized, prospective study. 1990;163:1822–7. https://doi.org/10.1016/0002-9378(90)90757-X PMID:2256489
  234. Rådberg T, Gustafson A, Skryten A, Karlsson K. Metabolic studies in women with previous gestational diabetes during contraceptive treatment: effects on serum lipids and high density lipoproteins. 1982;101:134–9. https://doi.org/10.1530/acta.0.1010134 PMID:7124287
  235. Skouby SO, Kühl C, Mølsted-Pedersen L, Petersen K, Christensen MS. Triphasic oral contraception: metabolic effects in normal women and those with previous gestational diabetes. 1985;153:495–500. https://doi.org/10.1016/0002-9378(85)90460-0 PMID:3933351
  236. Beck P, Arnett DM, Alsever RN, Eaton RP. Effect of contraceptive steroids on arginine-stimulated glucagon and insulin secretion in women. II. Carbohydrate and lipid physiology in insulin-dependent diabetics. 1976;25:23–31. https://doi.org/10.1016/0026-0495(76)90156-6 PMID:1246206
  237. Diab KM, Zaki MM. Contraception in diabetic women: comparative metabolic study of Norplant, depot medroxyprogesterone acetate, low dose oral contraceptive pill and CuT380A. 2000;26:17–26. https://doi.org/10.1111/j.1447-0756.2000.tb01195.x PMID:10761326
  238. Garg SK, Chase HP, Marshall G, Hoops SL, Holmes DL, Jackson WE. Oral contraceptives and renal and retinal complications in young women with insulin-dependent diabetes mellitus. 1994;271:1099–102. https://doi.org/10.1001/jama.1994.03510380055037 PMID:8151852
  239. Grigoryan OR, Grodnitskaya EE, Andreeva EN, Shestakova MV, Melnichenko GA, Dedov II. Contraception in perimenopausal women with diabetes mellitus. 2006;22:198–206. https://doi.org/10.1080/09513590600624317 PMID:16723306
  240. Margolis KL, Adami H-O, Luo J, Ye W, Weiderpass E. A prospective study of oral contraceptive use and risk of myocardial infarction among Swedish women. 2007;88:310–6. https://doi.org/10.1016/j.fertnstert.2006.11.206 PMID:17624338
  241. Petersen KR, Skouby SO, Jespersen J. Balance of coagulation activity with fibrinolysis during use of oral contraceptives in women with insulin-dependent diabetes mellitus. 1995;40(Suppl 2):105–11. PMID:8574252
  242. Petersen KR, Skouby SO, Sidelmann J, Jespersen J. Assessment of endothelial function during oral contraception in women with insulin-dependent diabetes mellitus. 1994;43:1379–83. https://doi.org/10.1016/0026-0495(94)90031-0 PMID:7968593
  243. Rådberg T, Gustafson A, Skryten A, Karlsson K. Oral contraception in diabetic women. A cross-over study on serum and high density lipoprotein (HDL) lipids and diabetes control during progestogen and combined estrogen/progestogen contraception. 1982;14:61–5. PMID:7040192
  244. Skouby SO, Jensen BM, Kühl C, Mølsted-Pedersen L, Svenstrup B, Nielsen J. Hormonal contraception in diabetic women: acceptability and influence on diabetes control and ovarian function of a nonalkylated estrogen/progestogen compound. 1985;32:23–31. https://doi.org/10.1016/0010-7824(85)90113-1 PMID:4053603
  245. Skouby SO, Mølsted-Pedersen L, Kühl C, Bennet P. Oral contraceptives in diabetic women: metabolic effects of four compounds with different estrogen/progestogen profiles. 1986;46:858–64. https://doi.org/10.1016/S0015-0282(16)49825-0 PMID:3781003
  246. Zapata LB, Paulen ME, Cansino C, Marchbanks PA, Curtis KM. Contraceptive use among women with inflammatory bowel disease: a systematic review. 2010;82:72–85. https://doi.org/10.1016/j.contraception.2010.02.012 PMID:20682145
  247. Whiteman MK, Oduyebo T, Zapata LB, Walker S, Curtis KM. Contraceptive safety among women with cystic fibrosis: a systematic review. 2016;94:621–9. https://doi.org/10.1016/j.contraception.2016.05.016 PMID:27287694
  248. Brunson A, Keegan T, Mahajan A, White R, Wun T. High incidence of venous thromboembolism recurrence in patients with sickle cell disease. 2019;94:862–70. https://doi.org/10.1002/ajh.25508 PMID:31074115
  249. Naik RP, Streiff MB, Haywood C Jr, Segal JB, Lanzkron S. Venous thromboembolism incidence in the Cooperative Study of Sickle Cell Disease. 2014;12:2010–6. https://doi.org/10.1111/jth.12744 PMID:25280124
  250. Noubiap JJ, Temgoua MN, Tankeu R, Tochie JN, Wonkam A, Bigna JJ. Sickle cell disease, sickle trait and the risk for venous thromboembolism: a systematic review and meta-analysis. 2018;16:27. https://doi.org/10.1186/s12959-018-0179-z PMID:30305805
  251. Ohene-Frempong K, Weiner SJ, Sleeper LA, et al. Cerebrovascular accidents in sickle cell disease: rates and risk factors. 1998;91:288–94. PMID:9414296
  252. Panel on Treatment of HIV During Pregnancy and Prevention of Perinatal Transmission. Recommendations for the use of antiretroviral drugs during pregnancy and interventions to reduce perinatal HIV transmission in the United States. Washington, DC: US Department of Health and Human Services; 2023. https://clinicalinfo.hiv.gov/en/guidelines/perinatal/recommendations-arv-drugs-pregnancy-overview
  253. Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of antiretroviral agents in adults and adolescents with HIV. Washington, DC: US Department of Health and Human Services; 2023. https://clinicalinfo.hiv.gov/sites/default/files/guidelines/documents/adult-adolescent-arv/guidelines-adult-adolescent-arv.pdf
  254. Aweeka FT, Rosenkranz SL, Segal Y, et al. ; NIAID AIDS Clinical Trials Group. The impact of sex and contraceptive therapy on the plasma and intracellular pharmacokinetics of zidovudine. 2006;20:1833–41. https://doi.org/10.1097/01.aids.0000244202.18629.36 PMID:16954724
  255. Kearney BP, Mathias A. Lack of effect of tenofovir disoproxil fumarate on pharmacokinetics of hormonal contraceptives. 2009;29:924–9. https://doi.org/10.1592/phco.29.8.924 PMID:19637945
  256. Todd CS, Deese J, Wang M, et al. ; FEM-PrEP Study Group. Sino-implant (II) continuation and effect of concomitant tenofovir disoproxil fumarate-emtricitabine use on plasma levonorgestrel concentrations among women in Bondo, Kenya. 2015;91:248–52. https://doi.org/10.1016/j.contraception.2014.10.008 PMID:25459097
  257. Murnane PM, Heffron R, Ronald A, et al. ; Partners PrEP Study Team. Pre-exposure prophylaxis for HIV-1 prevention does not diminish the pregnancy prevention effectiveness of hormonal contraception. 2014;28:1825–30. https://doi.org/10.1097/QAD.0000000000000290 PMID:24785951
  258. Kasonde M, Niska RW, Rose C, et al. Bone mineral density changes among HIV-uninfected young adults in a randomised trial of pre-exposure prophylaxis with tenofovir-emtricitabine or placebo in Botswana. 2014;9:e90111. https://doi.org/10.1371/journal.pone.0090111 PMID:24625530
  259. Callahan R, Nanda K, Kapiga S, et al. ; FEM-PrEP Study Group. Pregnancy and contraceptive use among women participating in the FEM-PrEP trial. 2015;68:196–203. https://doi.org/10.1097/QAI.0000000000000413 PMID:25590272
  260. Patel RC, Onono M, Gandhi M, et al. Pregnancy rates in HIV-positive women using contraceptives and efavirenz-based or nevirapine-based antiretroviral therapy in Kenya: a retrospective cohort study. 2015;2:e474–82. https://doi.org/10.1016/S2352-3018(15)00184-8 PMID:26520927
  261. Pyra M, Heffron R, Mugo NR, et al. ; Partners in Prevention HSVHIV Transmission Study and Partners PrEP Study Teams. Effectiveness of hormonal contraception in HIV-infected women using antiretroviral therapy. 2015;29:2353–9. https://doi.org/10.1097/QAD.0000000000000827 PMID:26544706
  262. Clark RA, Theall K. Population-based study evaluating association between selected antiretroviral therapies and potential oral contraceptive failure. 2004;37:1219–20. https://doi.org/10.1097/01.qai.0000136724.15758.ae PMID:15319685
  263. Landolt NK, Phanuphak N, Ubolyam S, et al. Efavirenz, in contrast to nevirapine, is associated with unfavorable progesterone and antiretroviral levels when coadministered with combined oral contraceptives. 2013;62:534–9. https://doi.org/10.1097/QAI.0b013e31827e8f98 PMID:23187949
  264. Sevinsky H, Eley T, Persson A, et al. The effect of efavirenz on the pharmacokinetics of an oral contraceptive containing ethinyl estradiol and norgestimate in healthy HIV-negative women. 2011;16:149–56. https://doi.org/10.3851/IMP1725 PMID:21447863
  265. Landolt NK, Phanuphak N, Ubolyam S, et al. Significant decrease of ethinylestradiol with nevirapine, and of etonogestrel with efavirenz in HIV-positive women. 2014;66:e50–2. https://doi.org/10.1097/QAI.0000000000000134 PMID:24608892
  266. Myer L, Carter RJ, Katyal M, Toro P, El-Sadr WM, Abrams EJ. Impact of antiretroviral therapy on incidence of pregnancy among HIV-infected women in Sub-Saharan Africa: a cohort study. 2010;7:e1000229. https://doi.org/10.1371/journal.pmed.1000229 PMID:20161723
  267. Schöller-Gyüre M, Kakuda TN, Woodfall B, et al. Effect of steady-state etravirine on the pharmacokinetics and pharmacodynamics of ethinylestradiol and norethindrone. 2009;80:44–52. https://doi.org/10.1016/j.contraception.2009.01.009 PMID:19501215
  268. Nanda K, Delany-Moretlwe S, Dubé K, et al. Nevirapine-based antiretroviral therapy does not reduce oral contraceptive effectiveness. 2013;27(Suppl 1):S17–25. https://doi.org/10.1097/QAD.0000000000000050 PMID:24088680
  269. Stuart GS, Moses A, Corbett A, et al. Combined oral contraceptives and antiretroviral PK/PD in Malawian women: pharmacokinetics and pharmacodynamics of a combined oral contraceptive and a generic combined formulation antiretroviral in Malawi. 2011;58:e40–3. https://doi.org/10.1097/QAI.0b013e31822b8bf8 PMID:21921726
  270. Mildvan D, Yarrish R, Marshak A, et al. Pharmacokinetic interaction between nevirapine and ethinyl estradiol/norethindrone when administered concurrently to HIV-infected women. 2002;29:471–7. https://doi.org/10.1097/00042560-200204150-00007 PMID:11981363
  271. Muro E, Droste JAH, Hofstede HT, Bosch M, Dolmans W, Burger DM. Nevirapine plasma concentrations are still detectable after more than 2 weeks in the majority of women receiving single-dose nevirapine: implications for intervention studies. 2005;39:419–21. https://doi.org/10.1097/01.qai.0000167154.37357.f9 PMID:16010163
  272. Crauwels HM, van Heeswijk RP, Buelens A, Stevens M, Hoetelmans RM. Lack of an effect of rilpivirine on the pharmacokinetics of ethinylestradiol and norethindrone in healthy volunteers. 2014;52:118–28. https://doi.org/10.5414/CP201943 PMID:24161160
  273. Zhang J, Chung E, Yones C, et al. The effect of atazanavir/ritonavir on the pharmacokinetics of an oral contraceptive containing ethinyl estradiol and norgestimate in healthy women. 2011;16:157–64. https://doi.org/10.3851/IMP1724 PMID:21447864
  274. Sekar VJ, Lefebvre E, Guzman SS, et al. Pharmacokinetic interaction between ethinyl estradiol, norethindrone and darunavir with low-dose ritonavir in healthy women. 2008;13:563–9. https://doi.org/10.1177/135965350801300415 PMID:18672535
  275. Glaxo Smith Kline. Lexiva (fosamprenavir calcium) [Package insert]. Research Triangle Park, NC:Glaxo Smith Kline; 2015.
  276. Vogler MA, Patterson K, Kamemoto L, et al. Contraceptive efficacy of oral and transdermal hormones when co-administered with protease inhibitors in HIV-1-infected women: pharmacokinetic results of ACTG trial A5188. 2010;55:473–82. https://doi.org/10.1097/QAI.0b013e3181eb5ff5 PMID:20842042
  277. Fröhlich M, Burhenne J, Martin-Facklam M, et al. Oral contraception does not alter single dose saquinavir pharmacokinetics in women. 2004;57:244–52. https://doi.org/10.1111/j.1365-2125.2003.01983.x PMID:14998420
  278. Boehringer Ingelheim Pharmaceuticals. Aptivus (tipranavir) [Package insert]. Ridgefield, CT: Boehringer Ingelheim Pharmaceuticals; 2005.
  279. Bristol-Myers Squibb. Reyataz (atazanavir sulfate) [Package insert]. Princeton, NJ: Bristol-Myers Squibb; 2003.
  280. Abel S, Russell D, Whitlock LA, Ridgway CE, Muirhead GJ. Effect of maraviroc on the pharmacokinetics of midazolam, lamivudine/zidovudine, and ethinyloestradiol/levonorgestrel in healthy volunteers. 2008;65(Suppl 1):19–26. https://doi.org/10.1111/j.1365-2125.2008.03132.x PMID:18333862
  281. Anderson MS, Hanley WD, Moreau AR, et al. Effect of raltegravir on estradiol and norgestimate plasma pharmacokinetics following oral contraceptive administration in healthy women. 2011;71:616–20. https://doi.org/10.1111/j.1365-2125.2010.03885.x PMID:21395656
  282. Song IH, Borland J, Chen S, Wajima T, Peppercorn AF, Piscitelli SC. Dolutegravir has no effect on the pharmacokinetics of oral contraceptives with norgestimate and ethinyl estradiol. 2015;49:784–9. https://doi.org/10.1177/1060028015580637 PMID:25862012
  283. Gilead Sciences. Vitekta (elvitegravir) [Package insert]. Foster City, CA: Gilead Sciences; 2012.
  284. Back DJ, Bates M, Bowden A, et al. The interaction of phenobarbital and other anticonvulsants with oral contraceptive steroid therapy. 1980;22:495–503. https://doi.org/10.1016/0010-7824(80)90102-X PMID:7471739
  285. Doose DR, Wang SS, Padmanabhan M, Schwabe S, Jacobs D, Bialer M. Effect of topiramate or carbamazepine on the pharmacokinetics of an oral contraceptive containing norethindrone and ethinyl estradiol in healthy obese and nonobese female subjects. 2003;44:540–9. https://doi.org/10.1046/j.1528-1157.2003.55602.x PMID:12681003
  286. Fattore C, Cipolla G, Gatti G, et al. Induction of ethinylestradiol and levonorgestrel metabolism by oxcarbazepine in healthy women. 1999;40:783–7. https://doi.org/10.1111/j.1528-1157.1999.tb00779.x PMID:10368079
  287. Rosenfeld WE, Doose DR, Walker SA, Nayak RK. Effect of topiramate on the pharmacokinetics of an oral contraceptive containing norethindrone and ethinyl estradiol in patients with epilepsy. 1997;38:317–23. https://doi.org/10.1111/j.1528-1157.1997.tb01123.x PMID:9070594
  288. Gaffield ME, Culwell KR, Lee CR. The use of hormonal contraception among women taking anticonvulsant therapy. 2011;83:16–29. https://doi.org/10.1016/j.contraception.2010.06.013 PMID:21134499
  289. Christensen J, Petrenaite V, Atterman J, et al. Oral contraceptives induce lamotrigine metabolism: evidence from a double-blind, placebo-controlled trial. 2007;48:484–9. https://doi.org/10.1111/j.1528-1167.2007.00997.x PMID:17346247
  290. Contin M, Albani F, Ambrosetto G, et al. Variation in lamotrigine plasma concentrations with hormonal contraceptive monthly cycles in patients with epilepsy. 2006;47:1573–5. https://doi.org/10.1111/j.1528-1167.2006.00558.x PMID:16981875
  291. Reimers A, Helde G, Brodtkorb E. Ethinyl estradiol, not progestogens, reduces lamotrigine serum concentrations. 2005;46:1414–7. https://doi.org/10.1111/j.1528-1167.2005.10105.x PMID:16146436
  292. Sabers A, Buchholt JM, Uldall P, Hansen EL. Lamotrigine plasma levels reduced by oral contraceptives. 2001;47:151–4. https://doi.org/10.1016/S0920-1211(01)00305-9 PMID:11673029
  293. Sabers A, Ohman I, Christensen J, Tomson T. Oral contraceptives reduce lamotrigine plasma levels. 2003;61:570–1. https://doi.org/10.1212/01.WNL.0000076485.09353.7A PMID:12939444
  294. Back DJ, Breckenridge AM, MacIver M, et al. The effects of ampicillin on oral contraceptive steroids in women. 1982;14:43–8. https://doi.org/10.1111/j.1365-2125.1982.tb04932.x PMID:6809025
  295. Back DJ, Grimmer SF, Orme ML, Proudlove C, Mann RD, Breckenridge AM. Evaluation of Committee on Safety of Medicines yellow card reports on oral contraceptive-drug interactions with anticonvulsants and antibiotics. 1988;25:527–32. https://doi.org/10.1111/j.1365-2125.1988.tb03341.x PMID:3408633
  296. Back DJ, Tjia J, Martin C, et al. The lack of interaction between temafloxacin and combined oral contraceptive steroids. 1991;43:317–23. https://doi.org/10.1016/0010-7824(91)90070-V PMID:1906791
  297. Bacon JF, Shenfield GM. Pregnancy attributable to interaction between tetracycline and oral contraceptives. 1980;280:293. https://doi.org/10.1136/bmj.280.6210.293 PMID:7357347
  298. Bainton R. Interaction between antibiotic therapy and contraceptive medication. 1986;61:453–5. https://doi.org/10.1016/0030-4220(86)90385-3 PMID:3459119
  299. Bollen M. Use of antibiotics when taking the oral contraceptive pill. [comment]. 1995;24:928–9. PMID:7794163
  300. Bromham DR, Cartmill RS. Knowledge and use of secondary contraception among patients requesting termination of pregnancy. 1993;306:556–7. https://doi.org/10.1136/bmj.306.6877.556 PMID:8461770
  301. Côté J. Interaction of griseofulvin and oral contraceptives. [Comment]. 1990;22:124–5. https://doi.org/10.1016/S0190-9622(08)80010-2 PMID:2298948
  302. Csemiczky G, Alvendal C, Landgren BM. Risk for ovulation in women taking a low-dose oral contraceptive (Microgynon) when receiving antibacterial treatment with a fluoroquinolone (ofloxacin). 1996;12:101–9. https://doi.org/10.1007/BF01849631 PMID:8863905
  303. de Groot AC, Eshuis H, Stricker BH. [Inefficacy of oral contraception during use of minocycline]. 1990;134:1227–9. PMID:2143563
  304. DeSano EA Jr, Hurley SC. Possible interactions of antihistamines and antibiotics with oral contraceptive effectiveness. 1982;37:853–4. https://doi.org/10.1016/S0015-0282(16)46350-8 PMID:6123451
  305. Donley TG, Smith RF, Roy B. Reduced oral contraceptive effectiveness with concurrent antibiotic use: a protocol for prescribing antibiotics to women of childbearing age. 1990;11:392–6. PMID:2083416
  306. Friedman CI, Huneke AL, Kim MH, Powell J. The effect of ampicillin on oral contraceptive effectiveness. 1980;55:33–7. PMID:7188714
  307. Grimmer SF, Allen WL, Back DJ, Breckenridge AM, Orme M, Tjia J. The effect of cotrimoxazole on oral contraceptive steroids in women. 1983;28:53–9. https://doi.org/10.1016/S0010-7824(83)80005-5 PMID:6414761
  308. Helms SE, Bredle DL, Zajic J, Jarjoura D, Brodell RT, Krishnarao I. Oral contraceptive failure rates and oral antibiotics. 1997;36:705–10. https://doi.org/10.1016/S0190-9622(97)80322-2 PMID:9146531
  309. Hempel E, Böhm W, Carol W, Klinger G. [Enzyme induction by drugs and hormonal contraception]. 1973;95:1451–7. PMID:4129505
  310. Hempel E, Zorn C, Graf K. [Effect of chemotherapy agents and antibiotics on hormonal contraception]. 1978;72:924–6. PMID:726527
  311. Hetényi G. Possible interactions between antibiotics and oral contraceptives. 1989;37:86–9. PMID:2686086
  312. Hughes BR, Cunliffe WJ. Interactions between the oral contraceptive pill and antibiotics. [Comment]. 1990;122:717–8. https://doi.org/10.1111/j.1365-2133.1990.tb07299.x PMID:2141276
  313. Joshi JV, Joshi UM, Sankholi GM, et al. A study of interaction of low-dose combination oral contraceptive with ampicillin and metronidazole. 1980;22:643–52. https://doi.org/10.1016/0010-7824(80)90089-X PMID:7214911
  314. Kakouris H, Kovacs GT. Pill failure and nonuse of secondary precautions. 1992;18:41–4.
  315. Kakouris H, Kovacs GT. How common are predisposing factors to pill failure among pill users? 1994;20:33–5.
  316. Kovacs GT, Riddoch G, Duncombe P, et al. Inadvertent pregnancies in oral contraceptive users. 1989;150:549–51. https://doi.org/10.5694/j.1326-5377.1989.tb136691.x PMID:2716563
  317. Lequeux A. [Pregnancy under oral contraceptives after treatment with tetracycline]. 1980;99:413–4. PMID:12336602
  318. London BM, Lookingbill DP. Frequency of pregnancy in acne patients taking oral antibiotics and oral contraceptives. 1994;130:392–3. https://doi.org/10.1001/archderm.1994.01690030128027 PMID:8129425
  319. Maggiolo F, Puricelli G, Dottorini M, Caprioli S, Bianchi W, Suter F. The effect of ciprofloxacin on oral contraceptive steroid treatments. 1991;17:451–4. PMID:1822438
  320. Murphy AA, Zacur HA, Charache P, Burkman RT. The effect of tetracycline on levels of oral contraceptives. 1991;164:28–33. https://doi.org/10.1016/0002-9378(91)90617-Z PMID:1986620
  321. Neely JL, Abate M, Swinker M, D’Angio R. The effect of doxycycline on serum levels of ethinyl estradiol, norethindrone, and endogenous progesterone. 1991;77:416–20. PMID:1992409
  322. Pillans PI, Sparrow MJ. Pregnancy associated with a combined oral contraceptive and itraconazole. [Comment]. 1993;106:436. PMID:8414287
  323. Scholten PC, Droppert RM, Zwinkels MG, Moesker HL, Nauta JJ, Hoepelman IM. No interaction between ciprofloxacin and an oral contraceptive. 1998;42:3266–8. https://doi.org/10.1128/AAC.42.12.3266 PMID:9835524
  324. Silber TJ. Apparent oral contraceptive failure associated with antibiotic administration. 1983;4:287–9. https://doi.org/10.1016/S0197-0070(83)80014-X PMID:6643209
  325. Sparrow MJ. Pill method failures. 1987;100:102–5. PMID:3470667
  326. Sparrow MJ. Pregnancies in reliable pill takers. 1989;102:575–7. PMID:2812591
  327. Sparrow MJ. Pill method failures in women seeking abortion: fourteen years experience. 1998;111:386–8. PMID:9830420
  328. van Dijke CP, Weber JC. Interaction between oral contraceptives and griseofulvin. 1984;288:1125–6. https://doi.org/10.1136/bmj.288.6424.1125-a PMID:6424759
  329. Wermeling DP, Chandler MH, Sides GD, Collins D, Muse KN. Dirithromycin increases ethinyl estradiol clearance without allowing ovulation. 1995;86:78–84. https://doi.org/10.1016/0029-7844(95)00075-3 PMID:7784027
  330. Young LK, Farquhar CM, McCowan LM, Roberts HE, Taylor J. The contraceptive practices of women seeking termination of pregnancy in an Auckland clinic. 1994;107:189–92. PMID:8196861
  331. Abrams LS, Skee D, Natarajan J, Wong FA. Pharmacokinetic overview of Ortho Evra/Evra. 2002;77(Suppl 2):S3–12. https://doi.org/10.1016/S0015-0282(01)03261-7 PMID:11849630
  332. Dogterom P, van den Heuvel MW, Thomsen T. Absence of pharmacokinetic interactions of the combined contraceptive vaginal ring NuvaRing with oral amoxicillin or doxycycline in two randomised trials. 2005;44:429–38. https://doi.org/10.2165/00003088-200544040-00007 PMID:15828855
  333. Devenport MH, Crook D, Wynn V, Lees LJ. Metabolic effects of low-dose fluconazole in healthy female users and non-users of oral contraceptives. 1989;27:851–9. https://doi.org/10.1111/j.1365-2125.1989.tb03449.x PMID:2547410
  334. Hilbert J, Messig M, Kuye O, Friedman H. Evaluation of interaction between fluconazole and an oral contraceptive in healthy women. 2001;98:218–23. PMID:11506836
  335. Kovács I, Somos P, Hámori M. Examination of the potential interaction between ketoconazole (Nizoral) and oral contraceptives with special regard to products of low hormone content (Rigevidon, Anteovin). 1986;34:167–70. PMID:3441880
  336. Lunell NO, Pschera H, Zador G, Carlström K. Evaluation of the possible interaction of the antifungal triazole SCH 39304 with oral contraceptives in normal healthy women. 1991;32:91–7. https://doi.org/10.1159/000293003 PMID:1748330
  337. McDaniel PA, Caldroney RD. Oral contraceptives and griseofulvin interactions. 1986;20:384. https://doi.org/10.1177/106002808602000511 PMID:3709350
  338. Meyboom RH, van Puijenbroek EP, Vinks MH, Lastdrager CJ. Disturbance of withdrawal bleeding during concomitant use of itraconazole and oral contraceptives. 1997;110:300. PMID:9293288
  339. Rieth H, Sauerbrey N. [Interaction studies with fluconazole, a new triazole antifungal drug]. 1989;139:370–4. PMID:2556861
  340. Sinofsky FE, Pasquale SA. The effect of fluconazole on circulating ethinyl estradiol levels in women taking oral contraceptives. 1998;178:300–4. https://doi.org/10.1016/S0002-9378(98)80016-1 PMID:9500490
  341. van Puijenbroek EP, Feenstra J, Meyboom RH. [Pill cycle disturbance in simultaneous use of itraconazole and oral contraceptives]. 1998;142:146–9. PMID:9557015
  342. Van Puijenbroek EP, Egberts AC, Meyboom RH, Leufkens HG. Signalling possible drug-drug interactions in a spontaneous reporting system: delay of withdrawal bleeding during concomitant use of oral contraceptives and itraconazole. 1999;47:689–93. https://doi.org/10.1046/j.1365-2125.1999.00957.x PMID:10383548
  343. Verhoeven CH, van den Heuvel MW, Mulders TM, Dieben TO. The contraceptive vaginal ring, NuvaRing, and antimycotic co-medication. 2004;69:129–32. https://doi.org/10.1016/j.contraception.2003.10.001 PMID:14759617
  344. Back DJ, Breckenridge AM, Grimmer SF, Orme ML, Purba HS. Pharmacokinetics of oral contraceptive steroids following the administration of the antimalarial drugs primaquine and chloroquine. 1984;30:289–95. https://doi.org/10.1016/0010-7824(84)90092-1 PMID:6439467
  345. Croft AM, Herxheimer A. Adverse effects of the antimalaria drug, mefloquine: due to primary liver damage with secondary thyroid involvement? 2002;2:6. https://doi.org/10.1186/1471-2458-2-6 PMID:11914150
  346. Karbwang J, Looareesuwan S, Back DJ, Migasana S, Bunnag D, Breckenridge AM. Effect of oral contraceptive steroids on the clinical course of malaria infection and on the pharmacokinetics of mefloquine in Thai women. 1988;66:763–7. PMID:3266115
  347. McGready R, Stepniewska K, Seaton E, et al. Pregnancy and use of oral contraceptives reduces the biotransformation of proguanil to cycloguanil. 2003;59:553–7. https://doi.org/10.1007/s00228-003-0651-x PMID:12955370
  348. Wanwimolruk S, Kaewvichit S, Tanthayaphinant O, Suwannarach C, Oranratnachai A. Lack of effect of oral contraceptive use on the pharmacokinetics of quinine. 1991;31:179–81. https://doi.org/10.1111/j.1365-2125.1991.tb05509.x PMID:2049234
  349. Back DJ, Breckenridge AM, Crawford F, et al. The effect of rifampicin on norethisterone pharmacokinetics. 1979;15:193–7. https://doi.org/10.1007/BF00563105 PMID:37091
  350. Back DJ, Breckenridge AM, Crawford FE, et al. The effect of rifampicin on the pharmacokinetics of ethynylestradiol in women. 1980;21:135–43. https://doi.org/10.1016/0010-7824(80)90125-0 PMID:7189454
  351. Barditch-Crovo P, Trapnell CB, Ette E, et al. The effects of rifampin and rifabutin on the pharmacokinetics and pharmacodynamics of a combination oral contraceptive. 1999;65:428–38. https://doi.org/10.1016/S0009-9236(99)70138-4 PMID:10223781
  352. Bolt HM, Bolt M, Kappus H. Interaction of rifampicin treatment with pharmacokinetics and metabolism of ethinyloestradiol in man. 1977;85:189–97. https://doi.org/10.1530/acta.0.0850189 PMID:577076
  353. Gupta KC, Ali MY. Failure of oral contraceptive with rifampicin. 1980;15:23. PMID:7269801
  354. Hirsch A, Tillement JP, Chretien J. Effets contrariants de la rifampicine sur les contraceptifs oraux: a propos de trois grossesses non desiree chez trois malades. 1975;2:174–82.
  355. Joshi JV, Joshi UM, Sankolli GM, et al. A study of interaction of a low-dose combination oral contraceptive with anti-tubercular drugs. 1980;21:617–29. https://doi.org/10.1016/0010-7824(80)90034-7 PMID:7428368
  356. Kropp R. [Rifampicin and oral contraceptives (author’s transl)]. 1974;28:270–2. PMID:4839951
  357. LeBel M, Masson E, Guilbert E, et al. Effects of rifabutin and rifampicin on the pharmacokinetics of ethinylestradiol and norethindrone. 1998;38:1042–50. https://doi.org/10.1177/009127009803801109 PMID:9824786
  358. Meyer B, Müller F, Wessels P, Maree J. A model to detect interactions between roxithromycin and oral contraceptives. 1990;47:671–4. https://doi.org/10.1038/clpt.1990.92 PMID:2113449
  359. Nocke-Finke L, Breuer H, Reimers D. [Effects of rifampicin on the menstrual cycle and on oestrogen excretion in patients taking oral contraceptives]. 1973;98:1521–3. https://doi.org/10.1055/s-0028-1107071 PMID:4580141
  360. Piguet B, Muglioni JF, Chaline G. [Letter: Oral contraception and rifampicin]. 1975;04:115–6. PMID:1138226
  361. Reimers D, Jezek A. [The simultaneous use of rifampicin and other antitubercular agents with oral contraceptives]. 1971;25:255–62. PMID:5556355
  362. Skolnick JL, Stoler BS, Katz DB, Anderson WH. Rifampin, oral contraceptives, and pregnancy. 1976;236:1382. https://doi.org/10.1001/jama.1976.03270130044027 PMID:989097
  363. Szoka PR, Edgren RA. Drug interactions with oral contraceptives: compilation and analysis of an adverse experience report database. 1988;49(Suppl 2):31S–8S. PMID:3282933
  364. Berry-Bibee EN, Kim MJ, Simmons KB, et al. Drug interactions between hormonal contraceptives and psychotropic drugs: a systematic review. 2016;94:650–67. https://doi.org/10.1016/j.contraception.2016.07.011 PMID:27444984
  365. Berry-Bibee EN, Kim MJ, Tepper NK, Riley HEM, Curtis KM. Co-administration of St. John’s wort and hormonal contraceptives: a systematic review. 2016;94:668–77. https://doi.org/10.1016/j.contraception.2016.07.010 PMID:27444983

Appendix E: Classifications for Barrier Methods

Classifications for barrier contraceptive methods include those for condoms, which include external (male) condoms (latex or synthetic) and internal (female) condoms, spermicide and vaginal pH modulator, and diaphragm with spermicide and cervical cap with spermicide (Box E1) (Table E1).

Patients should be counseled that consistent and correct use of external (male) latex condoms reduces the risk for sexually transmitted infections (STIs), including HIV infection (1). Use of internal (female) condoms can provide protection from transmission of STIs, although data are limited (1). Patients also should be counseled that pre-exposure prophylaxis, when taken as prescribed, is highly effective for preventing HIV infection (2).

Return to your place in the textBOX E1. Categories for classifying barrier methods

U.S. MEC 1 = A condition for which there is no restriction for the use of the contraceptive method

U.S. MEC 2 = A condition for which the advantages of using the method generally outweigh the theoretical or proven risks

U.S. MEC 3 = A condition for which the theoretical or proven risks usually outweigh the advantages of using the method

U.S. MEC 4 = A condition that represents an unacceptable health risk if the contraceptive method is used

Abbreviation: U.S. MEC = U.S. Medical Eligibility Criteria for Contraceptive Use.



TABLE E1. Classifications for barrier methods, including condoms, spermicide and vaginal pH modulator, and diaphragm with spermicide and cervical cap with spermicide
Condition Category Clarification/Evidence/Comment
Condom Spermicide/Vaginal pH modulator Diaphragm/Cap (with spermicide)
Personal Characteristics and Reproductive History
Pregnancy NA NA NA Clarification: None of these methods are relevant for contraception during known pregnancy. However, for persons who remain at risk for STIs or HIV infection during pregnancy, the correct and consistent use of condoms is recommended.
Age
a. Menarche to <40 years 1 1 1
b. ≥40 years 1 1 1
Parity
a. Nulliparous 1 1 1
b. Parous 1 1 2 Clarification: Risk for cervical cap failure is higher in parous persons than in nulliparous persons.
Postpartum (breastfeeding
and nonbreastfeeding)
a. <6 weeks postpartum 1 1 NA Clarification: Diaphragm and cap are unsuitable until uterine involution is complete.
b. ≥6 weeks postpartum 1 1 1
Postabortion (spontaneous
or induced)
a. First trimester abortion 1 1 1
b. Second trimester abortion 1 1 1 Clarification: Diaphragm and cap are unsuitable until 6 weeks after second trimester abortion.
c. Immediate postseptic abortion 1 1 1
Past ectopic pregnancy 1 1 1
History of pelvic surgery 1 1 1
Smoking
a. Age <35 years 1 1 1
b. Age ≥35 years
  i. <15 cigarettes per day 1 1 1
  ii. ≥15 cigarettes per day 1 1 1
Obesity
a. BMI ≥30 kg/m2 1 1 1
b. Menarche to <18 years and BMI ≥30 kg/m2 1 1 1
History of bariatric surgery
This condition is associated with
increased risk for adverse health
events as a result of pregnancy
(Box 3).
a. Restrictive procedures: decrease storage capacity of the stomach (vertical banded gastroplasty, laparoscopic adjustable gastric band, or laparoscopic sleeve gastrectomy) 1 1 1
b. Malabsorptive procedures: decrease absorption of nutrients and calories by shortening the functional length of the small intestine (Roux-en-Y gastric bypass or biliopancreatic diversion) 1 1 1
Surgery
a. Minor surgery without immobilization 1 1 1
b. Major surgery
   i. Without prolonged immobilization 1 1 1
   ii. With prolonged immobilization 1 1 1
Cardiovascular Disease
Multiple risk factors for atherosclerotic cardiovascular disease (e.g., older age, smoking, diabetes, hypertension, low HDL, high LDL, or high triglyceride levels) 1 1 1
Hypertension
Systolic blood pressure ≥160 mm
Hg or diastolic blood pressure
≥100 mm Hg are associated with
increased risk for adverse health
events as a result of pregnancy
(Box 3).
a. Adequately controlled hypertension 1 1 1
b. Elevated blood pressure levels
(properly taken measurements)
  i. Systolic 140–159 mm Hg or diastolic 90–99 mm Hg 1 1 1
  ii. Systolic ≥160 mm Hg or diastolic ≥100 mm Hg 1 1 1
c. Vascular disease 1 1 1
History of high blood pressure during pregnancy (when current blood pressure is measurable and normal) 1 1 1
Deep venous thrombosis/
Pulmonary embolism

This condition is associated with
increased risk for adverse health
events as a result of pregnancy
(Box 3).
a. Current or history of DVT/PE, receiving anticoagulant therapy (therapeutic dose) (e.g., acute DVT/PE or long-term therapeutic dose) 1 1 1
b. History of DVT/PE, receiving
anticoagulant therapy
(prophylactic dose)
  i. Higher risk for recurrent DVT/PE (one or more risk factors) 1 1 1
• Thrombophilia (e.g., factor V Leiden mutation; prothrombin gene mutation; protein S, protein C, and antithrombin deficiencies; or antiphospholipid syndrome)
• Active cancer (metastatic, receiving therapy, or within 6 months after clinical remission), excluding nonmelanoma skin cancer
• History of recurrent DVT/PE
  ii. Lower risk for recurrent DVT/PE (no risk factors) 1 1 1
c. History of DVT/PE, not receiving
anticoagulant therapy
  i. Higher risk for recurrent DVT/PE (one or more risk factors) 1 1 1
• History of estrogen-associated DVT/PE
• Pregnancy-associated DVT/PE
• Idiopathic DVT/PE
• Thrombophilia (e.g., factor V Leiden mutation; prothrombin gene mutation; protein S, protein C, and antithrombin deficiencies; or antiphospholipid syndrome)
• Active cancer (metastatic,
receiving therapy, or within 6 months after clinical remission), excluding nonmelanoma skin
cancer
• History of recurrent DVT/PE
  ii. Lower risk for recurrent DVT/PE (no risk factors) 1 1 1
d. Family history (first-degree relatives) 1 1 1
Thrombophilia (e.g., factor V Leiden mutation; prothrombin gene mutation; protein S, protein C, and antithrombin deficiencies; or antiphospholipid syndrome)
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 1 1 Clarification: Routine screening in the general population before contraceptive initiation is not recommended.
Superficial venous disorders
a. Varicose veins 1 1 1
b. Superficial venous thrombosis (acute or history) 1 1 1
Current and history of ischemic heart disease
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 1 1
Stroke (history of cerebrovascular accident)
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 1 1
Valvular heart disease
Complicated valvular heart
disease is associated with
increased risk for adverse health
events as a result of pregnancy
(Box 3).
a. Uncomplicated 1 1 1
b. Complicated (pulmonary hypertension, risk for atrial fibrillation, or history of subacute bacterial endocarditis) 1 1 2
Peripartum cardiomyopathy
This condition is associated with
increased risk for adverse health
events as a result of pregnancy
(Box 3).
a. Normal or mildly impaired
cardiac function (New York Heart
Association Functional Class I or
II: no limitation of activities or
slight, mild limitation of activity) (3)
  i. <6 months 1 1 1
  ii. ≥6 months 1 1 1
b. Moderately or severely impaired cardiac function (New York Heart Association Functional Class III or IV: marked limitation of activity or should be at complete rest) (3) 1 1 1
Renal Disease
Chronic kidney disease
This condition is associated with
increased risk for adverse health
events as a result of pregnancy
(Box 3).
a. Current nephrotic syndrome 1 1 1
b. Hemodialysis 1 1 1
c. Peritoneal dialysis 1 1 1
Rheumatic Diseases
Systemic lupus erythematosus
This condition is associated with
increased risk for adverse health
events as a result of pregnancy
(Box 3).
a. Positive (or unknown) antiphospholipid antibodies 1 1 1
b. Severe thrombocytopenia 1 1 1
c. Immunosuppressive therapy 1 1 1
d. None of the above 1 1 1
Rheumatoid arthritis
a. Not receiving immunosuppressive therapy 1 1 1
b. Receiving immunosuppressive therapy 1 1 1
Neurologic Conditions
Headaches
a. Nonmigraine (mild or severe) 1 1 1
b. Migraine
  i. Without aura (includes menstrual migraine) 1 1 1 Comment: Menstrual migraine is a subtype of migraine without aura. For more information see the International Headache Society’s International Classification of Headache Disorders, 3rd ed. (https://ichd-3.org) (4).
  ii. With aura 1 1 1
Epilepsy
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 1 1
Multiple sclerosis
a. Without prolonged immobility 1 1 1
b. With prolonged immobility 1 1 1
Depressive Disorders
Depressive disorders 1 1 1
Reproductive Tract Infections and Disorders
Unexplained vaginal bleeding
(suspicious for serious condition) before evaluation
1 1 1 Clarification: If pregnancy or an underlying pathological condition (e.g., pelvic malignancy) is suspected, it must be evaluated and the category adjusted after evaluation.
Endometriosis 1 1 1
Benign ovarian tumors (including cysts) 1 1 1
Severe dysmenorrhea 1 1 1
Gestational trophoblastic disease
This condition is associated with
increased risk for adverse health
events as a result of pregnancy
(Box 3).
a. Suspected gestational
trophoblastic disease (immediate
postevacuation)
  i. Uterine size first trimester 1 1 1
  ii. Uterine size second trimester 1 1 1
b. Confirmed gestational
trophoblastic disease (after initial
evacuation and during
monitoring)
  i. Undetectable or nonpregnant β–hCG levels 1 1 1
  ii. Decreasing β–hCG levels 1 1 1
  iii. Persistently elevated β-hCG levels or malignant disease, with no evidence or suspicion of intrauterine disease 1 1 1
  iv. Persistently elevated β-hCG levels or malignant disease, with evidence or suspicion of intrauterine disease 1 1 1
Cervical ectropion 1 1 1
Cervical intraepithelial neoplasia 1 1 1 Clarification: The cap should not be used. Diaphragm use has no restrictions.
Cervical cancer (awaiting treatment) 1 Vaginal pH modulator: 1
Spermicide: 2
1 Clarification: The cap should not be used. Diaphragm use has no restrictions.
Comment: Repeated and high-dose use of the spermicide nonoxynol-9 can cause vaginal and cervical irritation or abrasions.
Breast disease
Breast cancer is associated with
increased risk for adverse health
events as a result of pregnancy
(Box 3).
a. Undiagnosed mass 1 1 1
b. Benign breast disease 1 1 1
c. Family history of cancer 1 1 1
d. Breast cancer
  i. Current 1 1 1
  ii. Past and no evidence of current disease for 5 years 1 1 1
Endometrial hyperplasia 1 1 1
Endometrial cancer
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 1 1
Ovarian cancer
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 1 1
Uterine fibroids 1 1 1
Anatomical abnormalities 1 1 NA Clarification: The diaphragm cannot be used in certain cases of prolapse. Cap use is not appropriate for a person with markedly distorted cervical anatomy.
Pelvic inflammatory disease
a. Current PID 1 1 1
b. Past PID
  i. With subsequent pregnancy 1 1 1
  ii. Without subsequent pregnancy 1 1 1
Sexually transmitted infections
a. Current purulent cervicitis or chlamydial infection or gonococcal infection 1 1 1
b. Vaginitis (including Trichomonas vaginalis and bacterial vaginosis) 1 1 1
c. Other factors related to STIs 1 1 1
HIV
High risk for HIV infection 1 Vaginal pH modulator: 1
Spermicide: 4
4 Evidence: Repeated and high-dose use of the spermicide nonoxynol-9 was associated with increased risk for genital lesions, which might increase the risk for HIV infection (5).
Comment: Diaphragm and cap use is assigned category 4 because of concerns about the spermicide, not the diaphragm or cap.
HIV infection
For persons with HIV infection who are not clinically well or not receiving ARV therapy, this condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 Vaginal pH modulator: 1
Spermicide: 3
3 Comment: Use of spermicides, including with diaphragms and caps, can disrupt the cervical mucosa, which might increase viral shedding and HIV transmission to noninfected sex partners.
Other Infections
Schistosomiasis
Schistosomiasis with fibrosis of
the liver is associated with
increased risk for adverse health
events as a result of pregnancy
(Box 3).
a. Uncomplicated 1 1 1
b. Fibrosis of the liver 1 1 1
Tuberculosis
This condition is associated with
increased risk for adverse health
events as a result of pregnancy
(Box 3).
a. Nonpelvic 1 1 1
b. Pelvic 1 1 1
Malaria 1 1 1
History of toxic shock syndrome 1 1 3 Comment: Toxic shock syndrome has been reported in association with contraceptive sponge and diaphragm use.
Urinary tract infection 1 Vaginal pH modulator: 2
Spermicide: 1
2 Comment: Use of diaphragms and spermicides might increase risk for urinary tract infection.
Endocrine Conditions
Diabetes
Insulin-dependent diabetes;
diabetes with nephropathy,
retinopathy, or neuropathy;
diabetes with other vascular
disease; or diabetes of >20 years’
duration are associated with
increased risk for adverse health
events as a result of pregnancy
(Box 3).
a. History of gestational disease 1 1 1
b. Nonvascular disease
  i. Non-insulin dependent 1 1 1
  ii. Insulin dependent 1 1 1
c. Nephropathy, retinopathy, or neuropathy 1 1 1
d. Other vascular disease or diabetes of >20 years’ duration 1 1 1
Thyroid disorders
a. Simple goiter 1 1 1
b. Hyperthyroid 1 1 1
c. Hypothyroid 1 1 1
Gastrointestinal Conditions
Inflammatory bowel disease (ulcerative colitis or Crohn’s disease) 1 1 1
Gallbladder disease
a. Asymptomatic 1 1 1
b. Symptomatic
  i. Current 1 1 1
  ii. Treated by cholecystectomy 1 1 1
  iii. Medically treated 1 1 1
History of cholestasis
a. Pregnancy related 1 1 1
b. Past COC related 1 1 1
Viral hepatitis
a. Acute or flare 1 1 1
b. Chronic 1 1 1
Cirrhosis
Decompensated cirrhosis is
associated with increased risk for
adverse health events as a result
of pregnancy (Box 3).
a. Compensated (normal liver function) 1 1 1
b. Decompensated (impaired liver function) 1 1 1
Liver tumors
Hepatocellular adenoma and
malignant liver tumors are
associated with increased risk for
adverse health events as a result
of pregnancy (Box 3).
a. Benign
   i. Focal nodular hyperplasia 1 1 1
   ii. Hepatocellular adenoma 1 1 1
b. Malignant (hepatocellular carcinoma) 1 1 1
Respiratory Conditions
Cystic fibrosis
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 1 1
Hematologic Conditions
Thalassemia 1 1 1
Sickle cell disease
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 1 1
Iron deficiency anemia 1 1 1
Solid Organ Transplantation
Solid organ transplantation
This condition is associated with
increased risk for adverse health
events as a result of pregnancy
(Box 3).
a. No graft failure 1 1 1
b. Graft failure 1 1 1
Drug Interactions
Antiretrovirals used for
prevention (PrEP) or treatment
of HIV infection
Clarification: No drug interaction between ARV therapy and barrier method use is known. HIV infection is classified as category 1 for vaginal pH modulator and category 3 for spermicide and diaphragm and cap use (see recommendations for HIV infection). High risk for HIV infection is classified as category 1 for vaginal pH modulator and category 4 for spermicide and diaphragm or cap (see recommendations for High risk for HIV infection).
a. Nucleoside reverse
transcriptase inhibitors (NRTIs)
  i. Abacavir (ABC) 1 1/3/4 3/4
  ii. Tenofovir (TDF) 1 1/3/4 3/4
  iii. Zidovudine (AZT) 1 1/3/4 3/4
  iv. Lamivudine (3TC) 1 1/3/4 3/4
  v. Didanosine (DDI) 1 1/3/4 3/4
  vi. Emtricitabine (FTC) 1 1/3/4 3/4
  vii. Stavudine (D4T) 1 1/3/4 3/4
b. Nonnucleoside reverse
transcriptase inhibitors (NNRTIs)
  i. Efavirenz (EFV) 1 1/3/4 3/4
  ii. Etravirine (ETR) 1 1/3/4 3/4
  iii. Nevirapine (NVP) 1 1/3/4 3/4
  iv. Rilpivirine (RPV) 1 1/3/4 3/4
c. Ritonavir-boosted protease
inhibitors
  i. Ritonavir-boosted atazanavir (ATV/r) 1 1/3/4 3/4
  ii. Ritonavir-boosted darunavir (DRV/r) 1 1/3/4 3/4
  iii. Ritonavir-boosted fosamprenavir (FPV/r) 1 1/3/4 3/4
  iv. Ritonavir-boosted lopinavir (LPV/r) 1 1/3/4 3/4
  v. Ritonavir-boosted saquinavir (SQV/r) 1 1/3/4 3/4
  vi. Ritonavir-boosted tipranavir (TPV/r) 1 1/3/4 3/4
d. Protease inhibitors without ritonavir
  i. Atazanavir (ATV) 1 1/3/4 3/4
  ii. Fosamprenavir (FPV) 1 1/3/4 3/4
  iii. Indinavir (IDV) 1 1/3/4 3/4
  iv. Nelfinavir (NFV) 1 1/3/4 3/4
e. CCR5 co-receptor antagonists
  i. Maraviroc (MVC) 1 1/3/4 3/4
f. HIV integrase strand transfer
inhibitors
  i. Raltegravir (RAL) 1 1/3/4 3/4
  ii. Dolutegravir (DTG) 1 1/3/4 3/4
  iii. Elvitegravir (EVG) 1 1/3/4 3/4
g. Fusion inhibitors
  i. Enfuvirtide 1 1/3/4 3/4
Anticonvulsant therapy
a. Certain anticonvulsants (phenytoin, carbamazepine, barbiturates, primidone, topiramate, or oxcarbazepine) 1 1 1
b. Lamotrigine 1 1 1
Antimicrobial therapy
a. Broad-spectrum antibiotics 1 1 1
b. Antifungals 1 1 1
c. Antiparasitics 1 1 1
d. Rifampin or rifabutin therapy 1 1 1
Psychotropic medications
a. Selective serotonin reuptake inhibitors (SSRIs) 1 1 1
St. John’s wort 1 1 1
Allergy to latex 3 1 3 Clarification: The condition of allergy to latex does not apply to plastic condoms or diaphragms.

Abbreviations: ARV = antiretroviral; BMI = body mass index; COC = combined oral contraceptive; DVT = deep venous thrombosis; hCG = human chorionic gonadotropin; HDL = high-density lipoprotein; LDL = low-density lipoprotein; NA = not applicable; PE = pulmonary embolism; PID = pelvic inflammatory disease; PrEP = pre-exposure prophylaxis; STI = sexually transmitted infection.

References

  1. Workowski KA, Bachmann LH, Chan PA, et al. Sexually transmitted infections treatment guidelines, 2021. 2021;70(No. RR-4):1–187. https://doi.org/10.15585/mmwr.rr7004a1 PMID:34292926
  2. CDC. US Public Health Service preexposure prophylaxis for the prevention of HIV infection in the United States—2021 update: a clinical practice guideline. Atlanta, GA: US Department of Health and Human Services, CDC; 2021. https://www.cdc.gov/hiv/pdf/risk/prep/cdc-hiv-prep-guidelines-2021.pdf
  3. The Criteria Committee of the New York Heart Association. Nomenclature and criteria for diagnosis of diseases of the heart and great vessels. 9th ed. Boston, MA: Little, Brown and Co; 1994.
  4. Headache Classification Committee of the International Headache Society (IHS) The International Classification of Headache Disorders. 3rd ed. Cephalalgia 2018;38:1–211. https://www.ichd-3.org/wp-content/uploads/2018/01/The-International-Classification-of-Headache-Disorders-3rd-Edition-2018.pdf
  5. Wilkinson D, Ramjee G, Tholandi M, Rutherford G. Nonoxynol-9 for preventing vaginal acquisition of HIV infection by women from men. 2002;2002:CD003936. https://doi.org/10.1002/14651858.CD003936 PMID:12519622

Appendix F: Classifications for Fertility Awareness–Based Methods

Fertility awareness–based (FAB) methods involve identifying the fertile days of the menstrual cycle, whether by observing fertility signs, such as cervical secretions and basal body temperature or by monitoring cycle days, and might include use of Food and Drug Administration–cleared contraceptive software applications (Box F1) (Table F1). FAB methods can be used in combination with abstinence or barrier methods during the fertile time. If barrier methods are used, see Classifications for Barrier Methods (Appendix E).

No medical conditions worsen because of FAB methods. In general, FAB methods can be used without concern for health effects in persons who choose them. However, multiple conditions make their use more complex. The existence of these conditions suggests that use of these methods should be delayed until the condition is corrected or resolved, or persons using FAB methods need special counseling; and a provider with particular training in use of these methods is generally necessary to ensure correct use.

FAB methods do not protect against sexually transmitted infections (STIs), including HIV infection, and patients using FAB methods should be counseled that consistent and correct use of external (male) latex condoms reduces the risk for STIs, including HIV infection (1). Use of internal (female) condoms can provide protection from transmission of STIs, although data are limited (1). Patients also should be counseled that pre-exposure prophylaxis, when taken as prescribed, is highly effective for preventing HIV infection (2).

Return to your place in the textBOX F1. Definitions for terms associated with fertility awareness–based methods

  • Symptoms-based methods: FAB methods based on observation of fertility signs (e.g., cervical secretions or basal body temperature) such as the cervical mucus method, the symptothermal method, and the TwoDay method.
  • Calendar-based methods: FAB methods based on calendar calculations such as the calendar rhythm method and the standard days method.
  • Accept: No medical reason exists to deny the particular FAB method to a patient in this circumstance.
  • Caution: The method normally is provided in a routine setting but with extra preparation and precautions. For FAB methods, this usually means that special counseling might be needed to ensure correct use of the method by a patient in this circumstance.
  • Delay: Use of this method should be delayed until the condition is evaluated or corrected. Alternative temporary methods of contraception should be offered.

Abbreviation: FAB = fertility awareness–based.



TABLE F1. Fertility awareness–based methods, including symptoms-based and calendar-based methods
Condition Category Clarification/Evidence/Comment
Symptoms-based method Calendar-based method
Personal Characteristics and Reproductive History
Pregnancy NA NA Clarification: FAB methods are not relevant during pregnancy.
Life stage Comment: Menstrual irregularities are common in postmenarche and perimenopause and might complicate the use of FAB methods.
a. Postmenarche Caution Caution
b. Perimenopause Caution Caution
Breastfeeding Comment: Use of FAB methods when breastfeeding might be less effective than when not breastfeeding.
a. <6 weeks postpartum Delay Delay Comment: Persons who are primarily breastfeeding and are amenorrheic are unlikely to have sufficient ovarian function to produce detectable fertility signs and hormonal changes during the first 6 months postpartum. However, the likelihood of resumption of fertility increases with time postpartum and with substitution of breast milk by other foods.
b. ≥6 weeks postpartum Caution Delay
c. After menses begin Caution Caution Clarification: Once fertility signs are noted, particularly cervical secretions, then symptoms-based methods can be used. First postpartum menstrual cycles while breastfeeding vary significantly in length. Return to regularity takes several cycles. When there have been at least three postpartum menses and cycles are regular again, a calendar-based method can be used. When there have been at least four postpartum menses and the most recent cycle lasted 26–32 days, the standard days method can be used. Before that time, a barrier method should be offered if the patient plans to use a FAB method later.
Postpartum (nonbreastfeeding)
a. <4 weeks Delay Delay Clarification: Nonbreastfeeding persons are not likely to have detectable fertility signs or hormonal changes before 4 weeks postpartum. Although the risk for pregnancy is low, ovulation before first menses is common; therefore, a method appropriate for the postpartum period should be offered.
b. ≥4 weeks Accept Delay Clarification: Nonbreastfeeding persons are likely to have sufficient ovarian function to produce detectable fertility signs, hormonal changes, or both at this time; likelihood increases rapidly with time postpartum. Calendar-based methods can be used as soon as three postpartum menses have been completed. Methods appropriate for the postpartum period should be offered before that time.
Postabortion (spontaneous or induced) Caution Delay Clarification: After abortion, it is possible for ovarian function to produce detectable fertility signs, hormonal changes, or both; likelihood increases with time postabortion. Calendar-based methods can be used following at least one postabortion menses (e.g., persons who before this pregnancy primarily had cycles of 26–32 days can then use the standard days method). Methods appropriate for the postabortion period should be offered before that time.
Reproductive Tract Infections and Disorders
Irregular vaginal bleeding Delay Delay Clarification: Presence of this condition makes FAB methods unreliable. Therefore, barrier methods should be recommended until the bleeding pattern is compatible with proper method use. The condition should be evaluated and treated as necessary.
Vaginal discharge Delay Accept Clarification: Because vaginal discharge makes recognition of cervical secretions difficult, the condition should be evaluated and treated if needed before providing methods based on cervical secretions.
Other
Use of drugs that affect cycle regularity, hormones, or fertility signs Caution /Delay Caution/Delay Clarification: Use of certain mood-altering drugs (e.g., lithium, tricyclic antidepressants, and antianxiety therapies), as well as certain antibiotics and anti-inflammatory drugs, might alter cycle regularity or affect fertility signs. The condition should be carefully evaluated and a barrier method offered until the degree of effect has been determined or the drug is no longer being used.
Diseases that elevate
body temperature
a. Chronic diseases Caution Accept Clarification: Elevated temperatures might make basal body temperature difficult to interpret but have no effect on cervical secretions. Thus, use of a method that relies on temperature should be delayed until the acute febrile disease abates. Temperature-based methods are not appropriate for persons with chronically elevated temperatures. In addition, certain chronic diseases interfere with cycle regularity, making calendar-based methods difficult to interpret.
b. Acute diseases Delay Accept

Abbreviations: FAB = fertility awareness–based; NA = not applicable.

References

  1. Workowski KA, Bachmann LH, Chan PA, et al. Sexually transmitted infections treatment guidelines, 2021. 2021;70(No. RR-4):1–187. https://doi.org/10.15585/mmwr.rr7004a1 PMID:34292926
  2. CDC. US Public Health Service preexposure prophylaxis for the prevention of HIV infection in the United States—2021 update: a clinical practice guideline. Atlanta, GA: US Department of Health and Human Services, CDC; 2021. https://www.cdc.gov/hiv/pdf/risk/prep/cdc-hiv-prep-guidelines-2021.pdf

Appendix G: Lactational Amenorrhea Method

The Bellagio Consensus provided the scientific basis for defining the conditions under which breastfeeding can be used safely and effectively for birth-spacing purposes; programmatic guidelines were developed at a meeting of family planning experts for its use as a method of contraception, and the method was then named the lactational amenorrhea method (LAM) (13). These guidelines include the following three criteria, all of which must be met to ensure adequate protection from pregnancy: 1) amenorrhea, 2) fully or nearly fully breastfeeding (intervals between feedings not exceeding 4 hours during the day or 6 hours at night), and 3) <6 months postpartum (46).

The U.S. Dietary Guidelines for Americans recommend that infants be exclusively breastfed for about the first 6 months, with continued breastfeeding while introducing appropriate complementary foods for 1 year or longer (7). The American Academy of Pediatrics (AAP) recommends exclusive breastfeeding for about the first 6 months, with continued breastfeeding along with introducing appropriate complementary foods for up to age 2 years or longer (8).

No medical conditions exist for which use of LAM for contraception is restricted. However, breastfeeding might not be recommended for persons or infants with certain conditions.

LAM does not protect against sexually transmitted infections (STIs), including HIV infection, and patients using LAM should be counseled that consistent and correct use of external (male) latex condoms reduces the risk for STIs, including HIV infection (9). Use of internal (female) condoms can provide protection from transmission of STIs, although data are limited (9). Patients also should be counseled that pre-exposure prophylaxis, when taken as prescribed, is highly effective for preventing HIV infection (10).

HIV Infection. HIV transmission can occur during breastfeeding. For breastfeeding persons on antiretroviral therapy with a sustained undetectable HIV viral load during pregnancy, the risk for transmission through breastfeeding is <1%, but not zero. Patients with HIV infection should receive evidence-based, person-centered counseling to support shared decision-making about infant feeding. For comprehensive information, refer to Infant Feeding for Individuals with HIV in the United States (https://clinicalinfo.hiv.gov/en/guidelines/perinatal/counseling-and-managing-individuals-with-hiv-united-states-who-desire-breastfeed). These recommendations are included within the U.S. Department of Health and Human Services’s Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions to Reduce Perinatal HIV Transmission in the United States (https://clinicalinfo.hiv.gov/en/guidelines/perinatal/whats-new-guidelines) (11).

Other Medical Conditions. CDC and AAP also recommend against both breastfeeding and feeding expressed milk for persons with untreated brucellosis, positivity for human T-cell lymphotropic virus types I or II, herpes simplex lesions on a breast, Ebola virus disease, or mpox. In addition, infants with classic galactosemia should not breastfeed (8,12,13) (https://www.cdc.gov/breastfeeding-special-circumstances/hcp/contraindications/index.html).

Medication Used During Breastfeeding. Although many medications do pass into breast milk, most have little or no effect on milk supply or on infant well-being. Few medications are contraindicated while breastfeeding. More information about specific medications and radioactive compounds is provided by AAP (14), LactMed (https://www.ncbi.nlm.nih.gov/books/NBK501922), Mother to Baby (http://www.mothertobaby.org), and InfantRisk Center (https://www.infantrisk.com/category/breastfeeding).

References

  1. Kennedy KI, Rivera R, McNeilly AS. Consensus statement on the use of breastfeeding as a family planning method. 1989;39:477–96. https://doi.org/10.1016/0010-7824(89)90103-0 PMID:2656086
  2. Labbok M, Cooney K, Coly S. Guidelines: breastfeeding, family planning, and the lactational amenorrhea method—LAM. Washington, DC: Institute for Reproductive Health, 1994.
  3. Labbok MH, Perez A, Valdes V, et al. The lactational amenorrhea method (LAM): a postpartum introductory family planning method with policy and program implications. 1994;10:93–109. https://doi.org/10.1007/BF01978103 PMID:7942265
  4. Hight-Laukaran V, Labbok MH, Peterson AE, Fletcher V, von Hertzen H, Van Look PF. Multicenter study of the lactational amenorrhea method (LAM): II. Acceptability, utility, and policy implications. 1997;55:337–46. https://doi.org/10.1016/S0010-7824(97)00041-3 PMID:9262928
  5. Labbok MH, Hight-Laukaran V, Peterson AE, Fletcher V, von Hertzen H, Van Look PF. Multicenter study of the lactational amenorrhea method (LAM): I. Efficacy, duration, and implications for clinical application. 1997;55:327–36. https://doi.org/10.1016/S0010-7824(97)00040-1 PMID:9262927
  6. Peterson AE, Peŕez-Escamilla R, Labbok MH, Hight V, von Hertzen H, Van Look P. Multicenter study of the lactational amenorrhea method (LAM) III: effectiveness, duration, and satisfaction with reduced client-provider contact. 2000;62:221–30. https://doi.org/10.1016/S0010-7824(00)00171-2 PMID:11172792
  7. US Department of Agriculture; US Department of Health and Human Services. Dietary guidelines for Americans, 2020–2025. 9th ed. Washington, DC: US Department of Agriculture and US Department of Health and Human Services; 2020. https://www.dietaryguidelines.gov/sites/default/files/2021-03/Dietary_Guidelines_for_Americans-2020-2025.pdf
  8. Meek JY, Noble L; Section on Breastfeeding. Policy statement: breastfeeding and the use of human milk. 2022;150:e2022057988. https://doi.org/10.1542/peds.2022-057988 PMID:35921640
  9. Workowski KA, Bachmann LH, Chan PA, et al. Sexually transmitted infections treatment guidelines, 2021. 2021;70(No. RR-4):1–187. https://doi.org/10.15585/mmwr.rr7004a1 PMID:34292926
  10. CDC. US Public Health Service preexposure prophylaxis for the prevention of HIV infection in the United States—2021 update: a clinical practice guideline. Atlanta, GA: US Department of Health and Human Services, CDC; 2021. https://www.cdc.gov/hiv/pdf/risk/prep/cdc-hiv-prep-guidelines-2021.pdf
  11. Panel on Treatment of HIV During Pregnancy and Prevention of Perinatal Transmission. Recommendations for the use of antiretroviral drugs during pregnancy and interventions to reduce perinatal HIV transmission in the United States. Washington, DC: US Department of Health and Human Services; 2023. https://clinicalinfo.hiv.gov/en/guidelines/perinatal/recommendations-arv-drugs-pregnancy-overview
  12. Meek JY, Noble L. Technical report: breastfeeding and the use of human milk. 2022;150:e2022057989. https://doi.org/10.1542/peds.2022-057989 PMID:35921641
  13. CDC. Contraindications to breastfeeding or feeding expressed breast milk to infants; 2023. https://www.cdc.gov/breastfeeding-special-circumstances/hcp/contraindications/index.html
  14. Sachs HC, Frattarelli DAC, Galinkin JL, et al. ; Committee on Drugs. The transfer of drugs and therapeutics into human breast milk: an update on selected topics. 2013;132:e796–809. https://doi.org/10.1542/peds.2013-1985 PMID:23979084

Appendix H: Coitus Interruptus (Withdrawal)

Coitus interruptus, also known as withdrawal, is a contraceptive method in which the penis is completely removed from the vagina and away from the external genitalia before ejaculation. Coitus interruptus prevents sperm from entering the vagina, thereby preventing contact between spermatozoa and the ovum.

Coitus interruptus has no directly associated health risks. Coitus interruptus does not protect against sexually transmitted infections (STIs), including HIV infection, and patients using coitus interruptus should be counseled that consistent and correct use of external (male) latex condoms reduces the risk for STIs, including HIV infection (1). Use of internal (female) condoms can provide protection from transmission of STIs, although data are limited (1). Patients also should be counseled that pre-exposure prophylaxis, when taken as prescribed, is highly effective for preventing HIV infection (2).

References

  1. Workowski KA, Bachmann LH, Chan PA, et al. Sexually transmitted infections treatment guidelines, 2021. 2021;70(No. RR-4):1–187. https://doi.org/10.15585/mmwr.rr7004a1 PMID:34292926
  2. CDC. US Public Health Service preexposure prophylaxis for the prevention of HIV infection in the United States—2021 update: a clinical practice guideline. Atlanta, GA: US Department of Health and Human Services, CDC; 2021. https://www.cdc.gov/hiv/pdf/risk/prep/cdc-hiv-prep-guidelines-2021.pdf

Appendix I: Permanent Contraception

Tubal surgery (including laparoscopic and abdominal approaches) and vasectomy are methods of permanent contraception available in the United States. In general, no medical conditions absolutely restrict a person’s eligibility for permanent contraception (with the exception of known allergy or hypersensitivity to any materials used to complete the permanent contraception procedure). However, certain conditions might increase a person’s surgical risk during tubal surgery; in these cases, careful consideration can be given to the risks and benefits of other acceptable long-acting or permanent alternatives, including intrauterine device, implant, and vasectomy.

Patients should be appropriately counseled that permanent contraception is intended to be irreversible and about the availability of highly effective, long-acting reversible methods of contraception. Most persons who choose permanent contraception remain satisfied with their decision. However, a small proportion of women regret this decision (1%–26% from different studies, with higher rates of regret reported by women who were younger at time of permanent contraception procedure) (1,2). Regret among men about vasectomy has been reported to be approximately 5% (3), similar to the proportion of women who report regretting their husbands’ vasectomy (6%) (4).

Permanent contraception does not protect against sexually transmitted infections (STIs), including HIV infection, and patients using permanent contraception should be counseled that consistent and correct use of external (male) latex condoms reduces the risk for STIs, including HIV infection (5). Use of internal (female) condoms can provide protection from transmission of STIs, although data are limited (5). Patients also should be counseled that pre-exposure prophylaxis, when taken as prescribed, is highly effective for preventing HIV infection (6).

References

  1. Hillis SD, Marchbanks PA, Tylor LR, Peterson HB. Poststerilization regret: findings from the United States Collaborative Review of Sterilization. 1999;93:889–95. https://doi.org/10.1097/00006250-199906000-00001 PMID:10362150
  2. Peterson HB. Sterilization. 2008;111:189–203. https://doi.org/10.1097/01.AOG.0000298621.98372.62 PMID:18165410
  3. Ehn BE, Liljestrand J. A long-term follow-up of 108 vasectomized men. Good counselling routines are important. 1995;29:477–81. https://doi.org/10.3109/00365599509180030 PMID:8719366
  4. Jamieson DJ, Kaufman SC, Costello C, Hillis SD, Marchbanks PA, Peterson HB; US Collaborative Review of Sterilization Working Group. A comparison of women’s regret after vasectomy versus tubal sterilization. 2002;99:1073–9. https://doi.org/10.1016/S0029-7844(02)01981-6 PMID:12052602
  5. Workowski KA, Bachmann LH, Chan PA, et al. Sexually transmitted infections treatment guidelines, 2021. 2021;70(No. RR-4):1–187. https://doi.org/10.15585/mmwr.rr7004a1 PMID:34292926
  6. CDC. US Public Health Service preexposure prophylaxis for the prevention of HIV infection in the United States—2021 update: a clinical practice guideline. Atlanta, GA: US Department of Health and Human Services, CDC; 2021. https://www.cdc.gov/hiv/pdf/risk/prep/cdc-hiv-prep-guidelines-2021.pdf

Appendix J: Classifications for Emergency Contraception

Classifications are given for the copper intrauterine device (Cu-IUD) as emergency contraception. The Cu-IUD can be placed within 5 days of the first act of unprotected intercourse as emergency contraception. In addition, when the day of ovulation can be estimated, the Cu-IUD can be placed beyond 5 days after sexual intercourse, as long as the placement does not occur >5 days after ovulation. The eligibility criteria for interval Cu-IUD placement also apply for the placement of Cu-IUDs as emergency contraception (Box J1) (Table J1) (1).

Classifications for emergency contraceptive pills (ECPs) are given for ulipristal acetate (UPA), levonorgestrel (LNG), and combined oral contraceptives (COCs). ECPs should be taken as soon as possible within 5 days of unprotected sexual intercourse (1).

Cu-IUDs, UPA, LNG, and COCs do not protect against sexually transmitted infections (STIs), including HIV infection, and patients using these methods should be counseled that consistent and correct use of external (male) latex condoms reduces the risk for STIs, including HIV infection (2). Use of internal (female) condoms can provide protection from transmission of STIs, although data are limited (2). Patients also should be counseled that pre-exposure prophylaxis, when taken as prescribed, is highly effective for preventing HIV infection (3).

Return to your place in the textBOX J1. Categories for classifying emergency contraception

U.S. MEC 1 = A condition for which there is no restriction for the use of the contraceptive method.

U.S. MEC 2 = A condition for which the advantages of using the method generally outweigh the theoretical or proven risks.

U.S. MEC 3 = A condition for which the theoretical or proven risks usually outweigh the advantages of using the method.

U.S. MEC 4 = A condition that represents an unacceptable health risk if the contraceptive method is used.

Abbreviation: U.S. MEC = U.S. Medical Eligibility Criteria for Contraceptive Use.



TABLE J1. Classifications for emergency contraception, including the copper intrauterine device, ulipristal acetate, levonorgestrel, and combined oral contraceptives
Condition Category Clarification/Evidence/Comment
Cu-IUD UPA LNG COC
Personal Characteristics and Reproductive History
Pregnancy 4 NA NA NA Clarification (IUD): The IUD is not indicated during pregnancy and should not be used because of the risk for serious pelvic infection and septic spontaneous abortion.
Clarification (ECPs): Although this method is not indicated for a patient with a known or suspected pregnancy, no harm to the patient, the course of pregnancy, or the fetus if ECPs are inadvertently used is known to exist.
Evidence: Evidence suggests that poor pregnancy outcomes are rare among pregnant women who used ECPs during conception cycle or early in pregnancy (4).
Breastfeeding 1 1 1 1 Evidence: Breastfeeding outcomes do not seem to differ between women exposed to LNG and those who are not exposed (4). One pharmacokinetic study demonstrated that LNG passes to breast milk but in minimal quantities (4). UPA and its active metabolite, monodemethyl-ulipristal acetate, are present in human milk in small amounts; no evidence is available on effects of UPA emergency contraception exposure on infants or children who are breastfed (5).
Past ectopic pregnancy 1 1 1 1
Obesity (BMI ≥30 kg/m2) 1 2 2 2 Clarification (ECPs): ECPs might be less effective among persons with BMI ≥30 kg/m2 than among persons with BMI <25 kg/m2. Despite this, no safety concerns exist.
Evidence: Limited evidence from secondary data analyses suggests that women with BMI ≥30 kg/m2 experience an increased risk for pregnancy after use of LNG compared with women with BMI <25 kg/m2. Two analyses suggest that women with obesity might also experience an increased risk for pregnancy after use of UPA compared with those without obesity, although this increase was not significant in one study (6).
History of bariatric surgery
This condition is associated with increased risk for adverse
health events as a result of pregnancy (Box 3).
a. Restrictive procedures: decrease storage capacity of the stomach (vertical banded gastroplasty, laparoscopic adjustable gastric band, or laparoscopic sleeve gastrectomy) 1 1 1 1
b. Malabsorptive procedures: decrease absorption of nutrients and calories by shortening the functional length of the small intestine (Roux-en-Y gastric bypass or biliopancreatic diversion) 1 1 1 1 Comment: Bariatric surgical procedures involving a malabsorptive component have the potential to decrease oral contraceptive effectiveness, perhaps further decreased by postoperative complications such as long-term diarrhea, vomiting, or both. Because of these malabsorptive concerns, an emergency IUD might be more appropriate than ECPs.
Cardiovascular Disease
History of severe cardiovascular disease (ischemic heart disease, cerebrovascular attack, or other thromboembolic conditions)
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 2 2 2 Comment: The duration of ECP use is less than that of regular use of COCs or POPs and thus would be expected to have less clinical impact.
Rheumatic Diseases
Rheumatoid arthritis
a. Not receiving immunosuppressive therapy 1 1 1 1
b. Receiving immunosuppressive therapy 2 1 1 1
Neurologic Conditions
Migraine 1 1 1 2 Comment: The duration of ECP use is less than that of regular use of COCs and thus would be expected to have less clinical impact.
Gastrointestinal Conditions
Inflammatory bowel disease (ulcerative colitis or Crohn’s disease) 1 1 1 1
Severe liver disease (including jaundice)
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 2 2 2 Comment: The duration of ECP use is less than that of regular use of COCs or POPs and thus would be expected to have less clinical impact.
Solid Organ Transplantation
Solid organ transplantation
This condition is associated with increased risk for adverse
health events as a result of pregnancy (Box 3).
a. No graft failure 1 1 1 1
b. Graft failure 2 1 1 1
Other
Repeated ECP use 1 1 1 Clarification (ECPs): Frequently repeated ECP use might be harmful for persons with conditions classified as category 2, 3, or 4 for CHC or POC use.
Evidence: In one case-control study, risk for ectopic pregnancy compared with intrauterine pregnancy did not increase after repeated use of LNG ECPs compared with nonuse (4).
Sexual assault 2 1 1 1 Clarification (IUD): Persons who have experienced sexual assault are at increased risk for STIs, including HIV infection. According to CDC STI treatment guidelines, routine presumptive treatment of chlamydia, gonorrhea, and trichomonas is recommended after sexual assault (2). Persons with current purulent cervicitis, chlamydial infection, or gonococcal infection should not undergo IUD placement (category 4).
CYP3A4 inducers (e.g., bosentan, carbamazepine, felbamate, griseofulvin, oxcarbazepine, phenytoin, rifampin, St. John’s wort, topiramate, efavirenz, and lumacaftor) 1 2 2 2 Clarification (ECPs): Strong CYP3A4 inducers might reduce the effectiveness of ECPs.
Evidence: According to labelling information, rifampin markedly decreases UPA levels by ≥90%, which might decrease its efficacy (5). Therefore, theoretical concerns extend to use of other CYP3A4 inducers as well as to COC and LNG ECPs, which have metabolic pathways similar to those of UPA. A small pharmacokinetic study found that concomitant efavirenz use decreased LNG levels in women taking LNG ECPs (1.5 mg) by 56% compared with LNG ECPs alone (7).

Abbreviations: BMI = body mass index; CHC = combined hormonal contraceptive; COC = combined hormonal contraceptive; Cu-IUD = copper intrauterine device; CYP = cytochrome P450; ECP = emergency contraceptive pill; IUD = intrauterine device; LNG = levonorgestrel; NA = not applicable; POC = progestin-only contraceptive; POP = progestin-only pill; STI = sexually transmitted infection; UPA = ulipristal acetate.

References

  1. Curtis KM, Nguyen AT, Tepper NK, et al. U.S. selected practice recommendations for contraceptive use, 2024. 2024;73(No. RR-3):1–77.
  2. Workowski KA, Bachmann LH, Chan PA, et al. Sexually transmitted infections treatment guidelines, 2021. 2021;70(No. RR-4):1–187. https://doi.org/10.15585/mmwr.rr7004a1 PMID:34292926
  3. CDC. US Public Health Service preexposure prophylaxis for the prevention of HIV infection in the United States—2021 update: a clinical practice guideline. Atlanta, GA: US Department of Health and Human Services, CDC; 2021. https://www.cdc.gov/hiv/pdf/risk/prep/cdc-hiv-prep-guidelines-2021.pdf
  4. Jatlaoui TC, Riley H, Curtis KM. Safety data for levonorgestrel, ulipristal acetate and Yuzpe regimens for emergency contraception. 2016;93:93–112. https://doi.org/10.1016/j.contraception.2015.11.001 PMID:26546020
  5. HRA Pharma America. Ella [Package insert]. Morristown, NJ: HRA Pharma America; 2021.
  6. Jatlaoui TC, Curtis KM. Safety and effectiveness data for emergency contraceptive pills among women with obesity: a systematic review. 2016;94:605–11. https://doi.org/10.1016/j.contraception.2016.05.002 PMID:27234874
  7. Carten ML, Kiser JJ, Kwara A, Mawhinney S, Cu-Uvin S. Pharmacokinetic interactions between the hormonal emergency contraception, levonorgestrel (Plan B), and efavirenz. 2012;2012:137192. https://doi.org/10.1155/2012/137192 PMID:22536010

Appendix K: Summary of Classifications for Hormonal Contraceptive Methods and Intrauterine Devices

Health care providers can use the summary table as a quick reference guide to the classifications for hormonal contraceptive methods and intrauterine contraception to compare classifications across these methods (Box K1) (Table K1). See the respective appendix for each method for clarifications to the numeric categories, as well as for summaries of the evidence and additional comments. Hormonal contraceptives and intrauterine devices do not protect against sexually transmitted infections (STIs), including HIV infection, and patients using these methods should be counseled that consistent and correct use of external (male) latex condoms reduces the risk for STIs, including HIV infection (1). Use of internal (female) condoms can provide protection from transmission of STIs, although data are limited (1). Patients also should be counseled that pre-exposure prophylaxis, when taken as prescribed, is highly effective for preventing HIV infection (2).

Return to your place in the textBOX K1. Categories for classifying hormonal contraceptives and intrauterine devices

U.S. MEC 1 = A condition for which there is no restriction for the use of the contraceptive method

U.S. MEC 2 = A condition for which the advantages of using the method generally outweigh the theoretical or proven risks

U.S. MEC 3 = A condition for which the theoretical or proven risks usually outweigh the advantages of using the method

U.S. MEC 4 = A condition that represents an unacceptable health risk if the contraceptive method is used

Abbreviation: U.S. MEC = U.S. Medical Eligibility Criteria for Contraceptive Use.



TABLE K1. Summary of classifications for hormonal contraceptive methods and intrauterine devices
Condition Cu-IUD LNG-IUD Implant DMPA POP CHC
Personal Characteristics and Reproductive History
Pregnancy 4* 4* NA* NA* NA* NA*
Age Menarche to
<20 years: 2
Menarche to
<20 years: 2
Menarche to
<18 years: 1
Menarche to
<18 years: 2
Menarche to
<18 years: 1
Menarche to
<40 years: 1
≥20 years: 1 ≥20 years: 1 18–45 years: 1 18–45 years: 1 18–45 years: 1 ≥40 years: 2
>45 years: 1 >45 years: 2 >45 years: 1
Parity
a. Nulliparous 2 2 1 1 1 1
b. Parous 1 1 1 1 1 1
Breastfeeding
a. <21 days postpartum 2* 2* 2* 4*
b. 21 to <30 days postpartum
  i. With other risk factors for VTE (e.g., age ≥35 years, previous VTE, thrombophilia, immobility, transfusion at delivery, peripartum cardiomyopathy, BMI ≥30 kg/m2, postpartum hemorrhage, postcesarean delivery, preeclampsia, or smoking) 2* 2* 2* 3*
  ii. Without other risk factors for VTE 2* 2* 2* 3*
c. 30–42 days postpartum
  i. With other risk factors for VTE (e.g., age ≥35 years, previous VTE, thrombophilia, immobility, transfusion at delivery, peripartum cardiomyopathy, BMI ≥30 kg/m2, postpartum hemorrhage, postcesarean delivery, preeclampsia, or smoking) 1* 2* 1* 3*
  ii. Without other risk factors for VTE 1* 1* 1* 2*
d. >42 days postpartum 1* 1* 1* 2*
Postpartum (nonbreastfeeding)
a. <21 days postpartum 1 2 1 4
b. 21–42 days postpartum
  i. With other risk factors for VTE (e.g., age ≥35 years, previous VTE, thrombophilia, immobility, transfusion at delivery, peripartum cardiomyopathy, BMI ≥30 kg/m2, postpartum hemorrhage, postcesarean delivery, preeclampsia, or smoking) 1 2 1 3*
  ii. Without other risk factors for VTE 1 1 1 2
c. >42 days postpartum 1 1 1 1
Postpartum (including cesarean delivery, breastfeeding,
or nonbreastfeeding)
a. <10 minutes after delivery of the placenta 2* 2*
b. 10 minutes after delivery of the placenta to <4 weeks 2* 2*
c. ≥4 weeks 1* 1*
d. Postpartum sepsis 4 4
Postabortion (spontaneous or induced)
a. First trimester abortion
  i. Procedural (surgical) 1* 1* 1* 1* 1* 1*
  ii. Medication 1* 1* 1* 1/2* 1* 1*
  iii. Spontaneous abortion with no intervention 1* 1* 1* 1* 1* 1*
b. Second trimester abortion
  i. Procedural (surgical) 2* 2* 1* 1* 1* 1*
  ii. Medication 2* 2* 1* 1* 1* 1*
  iii. Spontaneous abortion with no intervention 2* 2* 1* 1* 1* 1*
c. Immediate postseptic abortion 4 4 1* 1* 1* 1*
Past ectopic pregnancy 1 1 1 1 2 1
History of pelvic surgery (see recommendations for Postpartum [including cesarean delivery]) 1 1 1 1 1 1
Smoking
a. Age <35 years 1 1 1 1 1 2
b. Age ≥35 years
  i. <15 cigarettes per day 1 1 1 1 1 3
  ii. ≥15 cigarettes per day 1 1 1 1 1 4
Obesity
a. BMI ≥30 kg/m2 1 1 1 1 1 2*
b. Menarche to <18 years and BMI ≥30 kg/m2 1 1 1 2 1 2*
History of bariatric surgery
This condition is associated with increased risk for
adverse health events as a result of pregnancy (Box 3).
a. Restrictive procedures: decrease storage capacity of the stomach (vertical banded gastroplasty, laparoscopic adjustable gastric band, or laparoscopic sleeve gastrectomy) 1 1 1 1 1 1
b. Malabsorptive procedures: decrease absorption of nutrients and calories by shortening the functional length of the small intestine (Roux-en-Y gastric bypass or biliopancreatic diversion) 1 1 1 1 3 COCs: 3
Patch and ring: 1
Surgery
a. Minor surgery without immobilization 1 1 1 1 1 1
b. Major surgery
  i. Without prolonged immobilization 1 1 1 1 1 2
  ii. With prolonged immobilization 1 1 1 2 1 4
Cardiovascular Disease
Multiple risk factors for atherosclerotic cardiovascular disease (e.g., older age, smoking, diabetes, hypertension, low HDL, high LDL, or high triglyceride levels) 1 2 2* 3* 2* 3/4*
Hypertension
Systolic blood pressure ≥160 mm Hg or diastolic blood
pressure ≥100 mm Hg are associated with increased
risk for adverse health events as a result
of pregnancy (Box 3).
a. Adequately controlled hypertension 1* 1* 1* 2* 1* 3*
b. Elevated blood pressure levels (properly taken
measurements)
  i. Systolic 140–159 mm Hg or diastolic 90–99 mm Hg 1* 1* 1* 2* 1* 3*
  ii. Systolic ≥160 mm Hg or diastolic ≥100 mm Hg 1* 2* 2* 3* 2* 4*
c. Vascular disease 1* 2* 2* 3* 2* 4*
History of high blood pressure during pregnancy (when current blood pressure is measurable and normal) 1 1 1 1 1 2
Deep venous thrombosis/Pulmonary embolism
This condition is associated with increased risk
for adverse health events as a result of
pregnancy (Box 3).
a. Current or history of DVT/PE, receiving anticoagulant therapy (therapeutic dose) (e.g., acute DVT/PE or long-term therapeutic dose) 2* 2* 2* 2* 2* 3*
b. History of DVT/PE, receiving anticoagulant therapy
(prophylactic dose)
  i. Higher risk for recurrent DVT/PE (one or more risk factors) 2* 2* 2* 3* 2* 4*
• Thrombophilia (e.g., factor V Leiden mutation; prothrombin gene mutation; protein S, protein C, and antithrombin deficiencies; or antiphospholipid syndrome)
• Active cancer (metastatic, receiving therapy, or within 6 months after clinical remission), excluding nonmelanoma skin cancer
• History of recurrent DVT/PE
  ii. Lower risk for recurrent DVT/PE (no risk factors) 2* 2* 2* 2* 2* 3*
c. History of DVT/PE, not receiving anticoagulant therapy
  i. Higher risk for recurrent DVT/PE (one or more risk factors 1 2 2 3 2 4
• History of estrogen-associated DVT/PE
• Pregnancy-associated DVT/PE
• Idiopathic DVT/PE
• Thrombophilia (e.g., factor V Leiden mutation; prothrombin gene mutation; protein S, protein C, and antithrombin deficiencies; or antiphospholipid syndrome)
• Active cancer (metastatic,
receiving therapy, or within 6 months after clinical remission), excluding nonmelanoma skin
cancer
• History of recurrent DVT/PE
  ii. Lower risk for recurrent DVT/PE (no risk factors) 1 2 2 2 2 3
d. Family history (first-degree relatives) 1 1 1 1 1 2
Thrombophilia (e.g., factor V Leiden mutation; prothrombin gene mutation; protein S, protein C, and antithrombin deficiencies; or antiphospholipid syndrome)
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1* 2* 2* 3* 2* 4*
Superficial venous disorders
a. Varicose veins 1 1 1 1 1 1
b. Superficial venous thrombosis (acute or history) 1 1 1 2 1 3*
Current and history of ischemic heart disease
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 Initiation Continuation Initiation Continuation 3 Initiation Continuation 4
2 3 2 3 2 3
Stroke (history of cerebrovascular accident)
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 2 Initiation Continuation 3 Initiation Continuation 4
2 3 2 3
Valvular heart disease
Complicated valvular heart disease is associated with
increased risk for adverse health events as a result of
pregnancy (Box 3).
a. Uncomplicated 1 1 1 1 1 2
b. Complicated (pulmonary hypertension, risk for atrial fibrillation, or history of subacute bacterial endocarditis) 1 1 1 2 1 4
Peripartum cardiomyopathy
This condition is associated with increased risk for
adverse health events as a result of pregnancy (Box 3).
a. Normal or mildly impaired cardiac function (New
York Heart Association Functional Class I or II: no
limitation of activities or slight, mild limitation
of activity) (3)
  i. <6 months 2 2 1 2 1 4
  ii. ≥6 months 2 2 1 2 1 3
b. Moderately or severely impaired cardiac function (New York Heart Association Functional Class III or IV: marked limitation of activity or should be at complete rest) (3) 2 2 2 3 2 4
Renal Disease
Chronic kidney disease
This condition is associated with increased risk
for adverse health events as a result of pregnancy (Box 3).
Initiation Continuation Initiation Continuation
a. Current nephrotic syndrome 1 1 2 2 2 3 2*
DRSP POP with known hyperkalemia: 4*
4
b. Hemodialysis 1 1 2 2 2 3 2*
DRSP POP with known hyperkalemia: 4*
4
c. Peritoneal dialysis 2 1 2 2 2 3 2*
DRSP POP with known hyperkalemia: 4*
4
Rheumatic Diseases
Systemic lupus erythematosus
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
Initiation Continuation Initiation Continuation
a. Positive (or unknown) antiphospholipid antibodies 1* 1* 2* 2* 3* 3* 2* 4*
b. Severe thrombocytopenia 3* 2* 2* 2* 3* 2* 2* 2*
c. Immunosuppressive therapy 2* 1* 2* 2* 2* 2* 2* 2*
d. None of the above 1* 1* 2* 2* 2* 2* 2* 2*
Rheumatoid arthritis Initiation Continuation Initiation Continuation
a. Not receiving immunosuppressive therapy 1 1 1 1 1 2 1 2
b. Receiving immunosuppressive therapy 2 1 2 1 1 2/3* 1 2
Neurologic Conditions
Headaches
a. Nonmigraine (mild or severe) 1 1 1 1 1 1*
b. Migraine
  i. Without aura (includes menstrual migraine) 1 1 1 1 1 2*
  ii. With aura 1 1 1 1 1 4*
Epilepsy
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 1 1* 1* 1* 1*
Multiple sclerosis
a. Without prolonged immobility 1 1 1 2 1 1
b. With prolonged immobility 1 1 1 2 1 3
Depressive Disorders
Depressive disorders 1* 1* 1* 1* 1* 1*
Reproductive Tract Infections and Disorders
Vaginal bleeding patterns Initiation Continuation
a. Irregular pattern without heavy bleeding 1 1 1 2 2 2 1
b. Heavy or prolonged bleeding (includes regular and irregular patterns) 2* 1* 2* 2* 2* 2* 1*
Unexplained vaginal bleeding
(suspicious for serious condition) before evaluation
Initiation Continuation Initiation Continuation
4* 2* 4* 2* 3* 3* 2* 2*
Endometriosis 2 1 1 1 1 1
Benign ovarian tumors (including cysts) 1 1 1 1 1 1
Severe dysmenorrhea 2 1 1 1 1 1
Gestational trophoblastic disease
This condition is associated with increased risk for
adverse health events as a result of pregnancy (Box 3).
a. Suspected gestational trophoblastic disease
(immediate postevacuation)
  i. Uterine size first trimester 1* 1* 1* 1* 1* 1*
  ii. Uterine size second trimester 2* 2* 1* 1* 1* 1*
b. Confirmed gestational trophoblastic disease (after initial evacuation and during monitoring) Initiation Continuation Initiation Continuation
  i. Undetectable or nonpregnant β-hCG levels 1* 1* 1* 1* 1* 1* 1* 1*
  ii. Decreasing β-hCG levels 2* 1* 2* 1* 1* 1* 1* 1*
  iii. Persistently elevated β-hCG levels or malignant disease, with no evidence or suspicion of intrauterine disease 2* 1* 2* 1* 1* 1* 1* 1*
  iv. Persistently elevated β-hCG levels or malignant disease, with evidence or suspicion of intrauterine disease 4* 2* 4* 2* 1* 1* 1* 1*
Cervical ectropion 1 1 1 1 1 1
Cervical intraepithelial neoplasia 1 2 2 2 1 2
Cervical cancer (awaiting treatment) Initiation Continuation Initiation Continuation
4 2 4 2 2 2 1 2
Breast disease
Breast cancer is associated with increased risk for
adverse health events as a result of pregnancy (Box 3).
a. Undiagnosed mass 1 2* 2* 2* 2* 2*
b. Benign breast disease 1 1 1 1 1 1
c. Family history of cancer 1 1 1 1 1 1
d. Breast cancer
  i. Current 1 4 4 4 4 4
  ii. Past and no evidence of current disease for 5 years 1 3 3 3 3 3
Endometrial hyperplasia 1 1 1 1 1 1
Endometrial cancer
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
Initiation Continuation Initiation Continuation
4 2 4 2 1 1 1 1
Ovarian cancer
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1 1 1 1 1 1
Uterine fibroids 2 2 1 1 1 1
Anatomical abnormalities
a. Distorted uterine cavity (any congenital or acquired uterine abnormality distorting the uterine cavity in a manner that is incompatible with IUD placement) 4 4
b. Other abnormalities (including cervical stenosis or cervical lacerations) not distorting the uterine cavity or interfering with IUD placement 2 2
Pelvic inflammatory disease Initiation Continuation Initiation Continuation
a. Current PID 4 2* 4 2* 1 1 1 1
b. Past PID
  i. With subsequent pregnancy 1 1 1 1 1 1 1 1
  ii. Without subsequent pregnancy 2 2 2 2 1 1 1 1
Sexually transmitted infections Initiation Continuation Initiation Continuation
a. Current purulent cervicitis or chlamydial infection or gonococcal infection 4 2* 4 2* 1 1 1 1
b. Vaginitis (including Trichomonas vaginalis and bacterial vaginosis) 2 2 2 2 1 1 1 1
c. Other factors related to STIs 2* 2 2* 2 1 1 1 1
HIV
High risk for HIV infection Initiation Continuation Initiation Continuation
1* 1* 1* 1* 1 1 1 1
HIV infection
For persons with HIV infection who are not clinically well or not receiving ARV therapy, this condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1* 1* 1* 1*
a. Clinically well receiving ARV therapy 1 1 1 1
b. Not clinically well or not receiving ARV therapy 2 1 2 1
Other Infections
Schistosomiasis
Schistosomiasis with fibrosis of the liver is associated
with increased risk for adverse health events
as a result of pregnancy (Box 3).
a. Uncomplicated 1 1 1 1 1 1
b. Fibrosis of the liver (if severe, see recommendations for Cirrhosis) 1 1 1 1 1 1
Tuberculosis
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
Initiation Continuation Initiation Continuation
a. Nonpelvic 1 1 1 1 1* 1* 1* 1*
b. Pelvic 4 3 4 3 1* 1* 1* 1*
Malaria 1 1 1 1 1 1
Endocrine Conditions
Diabetes
Insulin-dependent diabetes; diabetes with nephropathy,
retinopathy, or neuropathy; diabetes with other vascular
disease; or diabetes of >20 years’ duration are associated
with increased risk for adverse health events
as a result of pregnancy (Box 3).
a. History of gestational disease 1 1 1 1 1 1
b. Nonvascular disease
  i. Non-insulin dependent 1 2 2 2 2 2
  ii. Insulin dependent 1 2 2 2 2 2
c. Nephropathy, retinopathy, or neuropathy 1 2 2 3 2 3/4*
d. Other vascular disease or diabetes of >20 years’ duration 1 2 2 3 2 3/4*
Thyroid disorders
a. Simple goiter 1 1 1 1 1 1
b. Hyperthyroid 1 1 1 1 1 1
c. Hypothyroid 1 1 1 1 1 1
Gastrointestinal Conditions
Inflammatory bowel disease (ulcerative colitis or Crohn’s disease) 1 1 1 2 2 2/3*
Gallbladder disease
a. Asymptomatic 1 2 2 2 2 2
b. Symptomatic
  i. Current 1 2 2 2 2 3
  ii. Treated by cholecystectomy 1 2 2 2 2 2
  iii. Medically treated 1 2 2 2 2 3
History of cholestasis
a. Pregnancy related 1 1 1 1 1 2
b. Past COC related 1 2 2 2 2 3
Viral hepatitis Initiation Continuation
a. Acute or flare 1 1 1 1 1 3/4* 2
b. Chronic 1 1 1 1 1 1 1
Cirrhosis
Decompensated cirrhosis is associated with increased
risk for adverse health events as a result of
pregnancy (Box 3).
a. Compensated (normal liver function) 1 1 1 1 1 1
b. Decompensated (impaired liver function) 1 2 2 3 2 4
Liver tumors
Hepatocellular adenoma and malignant liver tumors are
associated with increased risk for adverse health events
as a result of pregnancy (Box 3).
a. Benign
  i. Focal nodular hyperplasia 1 2 2 2 2 2
  ii. Hepatocellular adenoma 1 2 2 3 2 4
b. Malignant (hepatocellular carcinoma) 1 3 3 3 3 4
Respiratory Conditions
Cystic fibrosis
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
1* 1* 1* 2* 1* 1*
Hematologic Conditions
Thalassemia 2 1 1 1 1 1
Sickle cell disease
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
2 1 1 2/3* 1 4
Iron-deficiency anemia 2 1 1 1 1 1
Solid Organ Transplantation
Solid organ transplantation
This condition is associated with increased risk for adverse health events as a result of pregnancy (Box 3).
Initiation Continuation Initiation Continuation
a. No graft failure 1 1 1 1 2 2/3* 2 2*
b. Graft failure 2 1 2 1 2 2/3* 2 4
Drug Interactions
Antiretrovirals used for prevention (PrEP)
or treatment of HIV infection
See the following guidelines for the most up-to-date recommendations on drug-drug interactions between hormonal contraception and antiretrovirals: 1) Recommendations for the Use of Antiretroviral Drugs During Pregnancy and Interventions to Reduce Perinatal HIV Transmission in the United (https://clinicalinfo.hiv.gov/en/guidelines/perinatal/prepregnancy-counseling-childbearing-age-overview?view=full#table-3) (4) and 2) Guidelines for the Use of Antiretroviral Agents in Adults and Adolescents with HIV (https://clinicalinfo.hiv.gov/en/guidelines/hiv-clinical-guidelines-adult-and-adolescent-arv/drug-interactions-overview?view=full) (5).
a. Nucleoside reverse transcriptase inhibitors (NRTIs) Initiation Continuation Initiation Continuation
  i. Abacavir (ABC) 1/2* 1* 1/2* 1* 1 1 1 1
  ii. Tenofovir (TDF) 1/2* 1* 1/2* 1* 1 1 1 1
  iii. Zidovudine (AZT) 1/2* 1* 1/2* 1* 1 1 1 1
  iv. Lamivudine (3TC) 1/2* 1* 1/2* 1* 1 1 1 1
  v. Didanosine (DDI) 1/2* 1* 1/2* 1* 1 1 1 1
  vi. Emtricitabine (FTC) 1/2* 1* 1/2* 1* 1 1 1 1
  vii. Stavudine (D4T) 1/2* 1* 1/2* 1* 1 1 1 1
b. Nonnucleoside reverse transcriptase inhibitors (NNRTIs)
  i. Efavirenz (EFV) 1/2* 1* 1/2* 1* 2* 1* 2* 2*
  ii. Etravirine (ETR) 1/2* 1* 1/2* 1* 1 1 1 1
  iii. Nevirapine (NVP) 1/2* 1* 1/2* 1* 1 1 1 1
  iv. Rilpivirine (RPV) 1/2* 1* 1/2* 1* 1 1 1 1
c. Ritonavir-boosted protease inhibitors
  i. Ritonavir-boosted atazanavir (ATV/r) 1/2* 1* 1/2* 1* 2* 1* 2* 2*
  ii. Ritonavir-boosted darunavir (DRV/r) 1/2* 1* 1/2* 1* 2* 1* 2* 2*
  iii. Ritonavir-boosted fosamprenavir (FPV/r) 1/2* 1* 1/2* 1* 2* 1* 2* 2*
  iv. Ritonavir-boosted lopinavir (LPV/r) 1/2* 1* 1/2* 1* 1 1 1 1
  v. Ritonavir-boosted saquinavir (SQV/r) 1/2* 1* 1/2* 1* 2* 1* 2* 2*
  vi. Ritonavir-boosted tipranavir (TPV/r) 1/2* 1* 1/2* 1* 2* 1* 2* 2*
d. Protease inhibitors without ritonavir
  i. Atazanavir (ATV) 1/2* 1* 1/2* 1* 1 1 1 2*
  ii. Fosamprenavir (FPV) 1/2* 1* 1/2* 1* 2* 2* 2* 3*
  iii. Indinavir (IDV) 1/2* 1* 1/2* 1* 1 1 1 1
  iv. Nelfinavir (NFV) 1/2* 1* 1/2* 1* 2* 1* 2* 2*
e. CCR5 co-receptor antagonists
  i. Maraviroc (MVC) 1/2* 1* 1/2* 1* 1 1 1 1
f. HIV integrase strand transfer inhibitors
  i. Raltegravir (RAL) 1/2* 1* 1/2* 1* 1 1 1 1
  ii. Dolutegravir (DTG) 1/2* 1* 1/2* 1* 1 1 1 1
  iii. Elvitegravir (EVG) 1/2* 1* 1/2* 1* 1 1 1 1
g. Fusion inhibitors
  i. Enfuvirtide 1/2* 1* 1/2* 1* 1 1 1 1
Anticonvulsant therapy
a. Certain anticonvulsants (phenytoin, carbamazepine, barbiturates, primidone, topiramate, and oxcarbazepine) 1 1 2* 1* 3* 3*
b. Lamotrigine 1 1 1 1 1 3*
Antimicrobial therapy
a. Broad-spectrum antibiotics 1 1 1 1 1 1
b. Antifungals 1 1 1 1 1 1
c. Antiparasitics 1 1 1 1 1 1
d. Rifampin or rifabutin therapy 1 1 2* 1* 3* 3*
Psychotropic medications
a. Selective serotonin reuptake inhibitors (SSRIs) 1 1 1 1 1 1
St. John’s wort 1 1 2 1 2 2

Abbreviations: ARV = antiretroviral; BMI = body mass index; CHC = combined hormonal contraceptive; COC = combined oral contraceptive; Cu-IUD = copper intrauterine device; DMPA = depot medroxyprogesterone acetate; DRSP = drospirenone; DVT = deep venous thrombosis; hCG = human chorionic gonadotropin; HDL = high-density lipoprotein; IUD = intrauterine device; LDL = low-density lipoprotein; LNG-IUD = levonorgestrel intrauterine device; NA = not applicable; PE = pulmonary embolism; PID = pelvic inflammatory disease; POP = progestin-only pill; PrEP = pre-exposure prophylaxis; STI = sexually transmitted infection; VTE = venous thromboembolism.
* Consult the appendix for this contraceptive method for a clarification to this classification.

References

  1. Workowski KA, Bachmann LH, Chan PA, et al. Sexually transmitted infections treatment guidelines, 2021. 2021;70(No. RR-4):1–187. https://doi.org/10.15585/mmwr.rr7004a1 PMID:34292926
  2. CDC. US Public Health Service preexposure prophylaxis for the prevention of HIV infection in the United States—2021 update: a clinical practice guideline. Atlanta, GA: US Department of Health and Human Services, CDC; 2021. https://www.cdc.gov/hiv/pdf/risk/prep/cdc-hiv-prep-guidelines-2021.pdf
  3. The Criteria Committee of the New York Heart Association. Nomenclature and criteria for diagnosis of diseases of the heart and great vessels. 9th ed. Boston, MA: Little, Brown and Co; 1994.
  4. Panel on Treatment of HIV During Pregnancy and Prevention of Perinatal Transmission. Recommendations for the use of antiretroviral drugs during pregnancy and interventions to reduce perinatal HIV transmission in the United States. Washington, DC: US Department of Health and Human Services; 2023. https://clinicalinfo.hiv.gov/en/guidelines/perinatal/recommendations-arv-drugs-pregnancy-overview
  5. Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of antiretroviral agents in adults and adolescents with HIV. Washington, DC: US Department of Health and Human Services; 2023. https://clinicalinfo.hiv.gov/sites/default/files/guidelines/documents/adult-adolescent-arv/guidelines-adult-adolescent-arv.pdf

Top


MMWR and Morbidity and Mortality Weekly Report are service marks of the U.S. Department of Health and Human Services.
Use of trade names and commercial sources is for identification only and does not imply endorsement by the U.S. Department of Health and Human Services.
References to non-CDC sites on the Internet are provided as a service to MMWR readers and do not constitute or imply endorsement of these organizations or their programs by CDC or the U.S. Department of Health and Human Services. CDC is not responsible for the content of pages found at these sites. URL addresses listed in MMWR were current as of the date of publication.

All HTML versions of MMWR articles are generated from final proofs through an automated process. This conversion might result in character translation or format errors in the HTML version. Users are referred to the electronic PDF version (https://www.cdc.gov/mmwr) and/or the original MMWR paper copy for printable versions of official text, figures, and tables.

Questions or messages regarding errors in formatting should be addressed to mmwrq@cdc.gov.