ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Tacrolimus (systemic): Drug information

Tacrolimus (systemic): Drug information
(For additional information see "Tacrolimus (systemic): Patient drug information" and see "Tacrolimus (systemic): Pediatric drug information")

For abbreviations, symbols, and age group definitions used in Lexicomp (show table)
ALERT: US Boxed Warning
Malignancies and serious infection:

Increased risk for developing serious infections and malignancies with tacrolimus or other immunosuppressants that may lead to hospitalization or death.

Mortality in liver transplantation (Astagraf XL):

Increased mortality in female transplant patients with Astagraf XL. Astagraf XL is not approved for use in liver transplantation.

Brand Names: US
  • Astagraf XL;
  • Envarsus XR;
  • Prograf
Brand Names: Canada
  • ACH-Tacrolimus;
  • Advagraf;
  • Envarsus PA;
  • Prograf;
  • SANDOZ Tacrolimus
Pharmacologic Category
  • Calcineurin Inhibitor;
  • Immunosuppressant Agent
Dosing: Adult

Dosage guidance:

Dosage form information: ER products (Advagraf [Canadian product], Astagraf XL, Envarsus PA [Canadian product], and Envarsus XR) are not interchangeable or substitutable with IR tacrolimus. In addition, the once-daily formulations (Advagraf [Canadian product], Astagraf XL, Envarsus PA [Canadian product], and Envarsus XR) are not interchangeable with each other due to significantly different pharmacokinetic properties. Patients should be transitioned from IV to oral tacrolimus as soon as able to tolerate oral administration.

Graft-versus-host disease

Graft-versus-host disease (off-label use):

Prevention:

Oral: Convert from IV to immediate-release oral dose (1:4 ratio): Multiply total daily IV dose times 4 and administer in 2 divided oral doses per day, every 12 hours (Ref).

IV: Initial: 0.03 mg/kg/day (based on lean body weight) as continuous infusion. Treatment should begin at least 24 hours prior to stem cell infusion and continued only until oral medication can be tolerated (Ref).

Treatment:

Oral: Immediate release: 0.06 mg/kg twice daily (Ref).

IV: Initial: 0.03 mg/kg/day (based on lean body weight) as continuous infusion (Ref).

Myasthenia gravis, chronic immunosuppressive therapy

Myasthenia gravis, chronic immunosuppressive therapy (alternative agent) (off-label use):

Note: For use as monotherapy or in combination with glucocorticoids in patients with glucocorticoid-resistant or glucocorticoid-dependent disease (Ref).

Immediate release: Oral: Initial: 3 to 5 mg/day or 0.1 mg/kg/day, in 1 or 2 divided doses; titrate to achieve target trough concentrations. Maximum dose is not established. Onset of clinical response to tacrolimus may take up to 6 to 12 months (Ref).

Prevention of organ rejection in transplant recipients

Prevention of organ rejection in transplant recipients: Note: While recommendations for initial doses exist, drug interactions should be considered. After initial dose, titrate to achieve target trough concentrations based on donor/recipient factors and institutional protocols. Lower oral doses may be sufficient for maintenance therapy. Adjunctive therapy with other immunosuppressants, including but not limited to corticosteroids, is recommended early post-transplant. The IV route should only be used in patients not able to take oral medications and continued only until oral medication can be tolerated; anaphylaxis has been reported with IV administration. If switching from IV to oral, the oral dose should be started 8 to 12 hours after stopping the infusion. Sublingual administration (off-label route) may be considered in those unable to take oral.

Heart transplant: Use in combination with an antimetabolite agent (eg, azathioprine, mycophenolate mofetil). Instead of an antimetabolite, may also use in combination with an mTOR kinase inhibitor (eg, everolimus, sirolimus) (Ref).

Oral: Immediate release: Initial: 0.075 mg/kg/day in 2 divided doses, given every 12 hours; titrate to target trough concentrations.

IV: Initial: 0.01 mg/kg/day as a continuous infusion.

Conversion from oral to continuous IV infusion: According to the ISHLT guidelines for the care of heart transplant recipients, convert from oral to IV by administering 10% to 33% of the total oral daily dose as a continuous infusion over 24 hours or divided twice daily as an intermittent infusion over 4 hours (Ref).

Kidney transplant: Use in combination with azathioprine or mycophenolate mofetil. Note: In African American patients, larger doses may be required to attain target trough concentrations.

Oral:

Immediate release (Prograf): Initial: 0.1 mg/kg twice daily every 12 hours in combination with azathioprine or 0.05 mg/kg twice daily every 12 hours in combination with mycophenolate mofetil; titrate to target trough concentrations.

Conversion from immediate release oral to IV: Administer one-third (1/3rd) of the total daily oral dose as a continuous infusion over 24 hours.

Extended release (Advagraf [Canadian product], Astagraf XL):

With basiliximab induction (prior to reperfusion or within 48 hours of transplant completion): 0.15 to 0.2 mg/kg once daily (in combination with corticosteroids and mycophenolate); titrate to target trough concentrations.

Without basiliximab induction:

Preoperative dose (administer within 12 hours prior to reperfusion): 0.1 mg/kg (in combination with corticosteroids and mycophenolate).

Postoperative dose (administer at least 4 hours after preoperative dose and within 12 hours of reperfusion): 0.2 mg/kg once daily (in combination with corticosteroids and mycophenolate); titrate to target trough concentrations.

Conversion from IV to extended release (Astagraf XL): Administer the first oral ER dose 8 to 12 hours after discontinuation of IV tacrolimus.

Conversion from immediate release to extended release (Advagraf [Canadian product], Astagraf XL): Initiate ER treatment in a 1:1 ratio (mg:mg) using previously established total daily dose of immediate release (Ref). Administer once daily.

Extended release:

Envarsus XR: 0.14 mg/kg/day; titrate to target trough concentrations.

Conversion from IV to extended release (Envarsus XR): Administer the first oral ER dose 8 to 12 hours after discontinuation of IV tacrolimus.

Conversion from immediate release to extended release (Envarsus XR): Initiate ER treatment with a once-daily dose that is 70% to 80% of the total daily dose of the IR tacrolimus.

Envarsus PA [Canadian product]:0.17 mg/kg/day concomitantly used with corticosteroids and/or mycophenolic acids or azathioprine; initiate within 24 hours of transplantation; titrate to target trough concentrations.

Conversion from immediate release to extended release (Envarsus PA [Canadian product]): Patients stable on IR tacrolimus may be converted to extended release by initiating ER treatment in a 1:0.7 ratio (1 mg IR:0.7 mg ER) (or a 1:0.85 ratio for African American patients) using previously established total daily dose of IR product. Administer once daily.

IV: Initial: 0.03 to 0.05 mg/kg/day as a continuous infusion.

Liver transplant:

Oral:

Immediate release: Initial: 0.1 to 0.15 mg/kg/day in 2 divided doses, given every 12 hours in combination with corticosteroids (titrate to target trough concentrations); consider lower dose in liver transplant recipients with graft dysfunction.

Extended release:

Advagraf [Canadian product]: 0.1 to 0.2 mg/kg once daily in combination with corticosteroids; initiate within 12 to 18 hours of transplantation; titrate to target trough concentrations. Note: In the US, Astagraf XL is not approved for use in liver transplantation due to an increase in mortality in female liver transplant recipients receiving Astagraf XL.

Conversion from immediate release to extended release (Advagraf [Canadian product]): Patients stable on IR tacrolimus may be converted to extended release by initiating ER treatment in a 1:1 ratio (mg:mg) using previously established total daily dose of IR product. Administer once daily.

Envarsus PA [Canadian product]: Initial: 0.11 to 0.13 mg/kg once daily concomitantly used with corticosteroids and/or mycophenolic acids or azathioprine; initiate within 24 hours of transplantation; titrate to target trough concentrations.

Conversion from immediate release to extended release (Envarsus PA [Canadian product]): Patients stable on IR tacrolimus may be converted to extended release by initiating ER treatment in a 1:0.7 ratio (1 mg IR:0.7 mg ER) (or a 1:0.85 ratio for African American patients) using previously established total daily dose of IR product. Administer once daily.

IV: Initial: 0.03 to 0.05 mg/kg/day as a continuous infusion.

Lung transplant: Use in combination with an antimetabolite agent (eg, azathioprine, mycophenolate mofetil). Adjunct corticosteroid therapy is recommended in the early postoperative period.

Oral: Immediate release: Initial: 0.075 mg/kg/day in 2 divided doses, administered every 12 hours (titrate to target trough concentrations). Note: Patients with cystic fibrosis may require higher oral doses due to lower bioavailability (monitor trough concentrations and adjust dose accordingly).

IV: Initial: 0.01 to 0.03 mg/kg/day as a continuous infusion.

Off-label dosing:

Oral, nasogastric: Immediate release: 0.05 to 0.3 mg/kg/day in 2 divided doses, given every 12 hours (usual dose: 0.05 mg/kg every 12 hours); titrate to target trough concentrations (Ref). May also be administered sublingually at ~50% of the oral/NG dose (Ref).

Note: May convert from twice-daily dosing to once-daily dosing (on a mg per mg basis) using the ER formulation (Astagraf XL [US] or Advagraf [Canada]) in stable lung transplant recipients (Mendez 2014). May convert to ER Envarsus with a once-daily dose that is 70% to 80% of the total daily dose of IR tacrolimus although clinical trial data are lacking.

IV: 0.01 to 0.05 mg/kg over 24 hours as a continuous IV infusion; titrate to target trough concentrations (Ref). For patients receiving the initial dose of tacrolimus IV, may begin immediately after transplantation, or up to 2 days postoperatively depending on renal function and hemodynamic stability (Ref). When patient is able to take oral medication, may switch to an oral maintenance regimen (typically transitioned after extubation).

Pancreas transplant (off-label use): Oral, nasogastric: Immediate release: 0.1 mg/kg twice daily; titrate to target trough concentrations (Ref). May also be administered sublingually at ~50% of the oral/NG dose (Ref).

Immunosuppression after solid-organ transplant, sublingual administration: Immediate release: Sublingual (off-label route): Optimal dosing has not been determined. In dosing regimens using sublingual administration of the contents of IR tacrolimus capsules, the sublingual to oral dosing ratio has ranged from 1:3 to 1:1 (Ref). However, most studies suggest a dosing ratio of 1:2 (or 50% of the oral dose given sublingually), and most centers use this approach in practice (Ref). Adjust dose based on serum trough concentrations. Lower doses of sublingual tacrolimus may be required during coadministration of drugs that inhibit tacrolimus metabolism (Ref).

Dosage adjustment for concomitant therapy: Significant drug interactions exist, requiring dose/frequency adjustment or avoidance. Consult drug interactions database for more information.

Dosing: Kidney Impairment: Adult

The renal dosing recommendations are based upon the best available evidence and clinical expertise. Senior Editorial Team: Bruce Mueller, PharmD, FCCP, FASN, FNKF; Jason Roberts, PhD, BPharm (Hons), B App Sc, FSHP, FISAC; Michael Heung, MD, MS.

Note: Dose adjustment should be guided by therapeutic drug monitoring (Ref).

Kidney impairment prior to treatment initiation:

Note: Avoid concurrent nephrotoxins, consider more frequent therapeutic drug monitoring especially in patients receiving drugs that affect tacrolimus metabolism, and consider targeting the lower end of the therapeutic range to minimize the risk of acute and/or chronic nephrotoxicity with tacrolimus therapy (Ref).

Altered kidney function: IV, Oral: No initial dose adjustment necessary for any degree of kidney dysfunction (Ref).

Hemodialysis, intermittent (thrice weekly): IV, Oral: Unlikely to be significantly dialyzed (Ref): No initial dosage adjustment necessary (Ref).

Peritoneal dialysis: IV, Oral: Not removed by peritoneal dialysis (Ref): No initial dosage adjustment necessary (Ref).

CRRT: IV, Oral: No initial dosage adjustment necessary (Ref).

PIRRT (eg, sustained, low-efficiency diafiltration): IV, Oral: No initial dosage adjustment necessary (Ref).

Nephrotoxicity during treatment: Treatment decisions should be individualized and in consultation with nephrology and/or transplant specialists.

Dosing: Hepatic Impairment: Adult

Severe impairment (Child-Pugh class C ): Lower initial doses may be required (due to reduced clearance and increased half-life); closely monitor tacrolimus blood concentrations.

Liver transplant recipients: Use of tacrolimus in liver transplant recipients experiencing post-transplant hepatic impairment may be associated with increased risk of developing renal insufficiency related to high whole blood levels of tacrolimus. Monitor closely and consider dosage adjustment; some evidence suggests utilizing lower initial doses.

Dosing: Adjustment for Toxicity: Adult

Hypersensitivity: Stop infusion for signs/symptoms of anaphylaxis.

Myocardial hypertrophy: Consider dose reduction or discontinuation if myocardial hypertrophy is diagnosed.

Neurotoxicity: Consider dose reduction or discontinuation if neurotoxicity occurs.

Pure red cell aplasia: Consider discontinuation if pure red cell aplasia is diagnosed.

Dosing: Older Adult

Refer to adult dosing. Use with caution; begin at the low end of dosing range.

Dosing: Pediatric

(For additional information see "Tacrolimus (systemic): Pediatric drug information")

Dosage guidance:

Dosage form information: Tacrolimus granules may have higher AUC compared to capsule formulation; if changing between products, monitor tacrolimus concentrations. Extended-release products (Astagraf XL and Envarsus XR) are not interchangeable or substitutable with immediate-release tacrolimus (Prograf capsules and granules) or with each other due to significantly different pharmacokinetic properties.

Clinical considerations: Younger children generally require higher maintenance doses on a mg/kg basis than older children, adolescents, or adults (Ref). Adjust initial dose to achieve desired target tacrolimus concentration; consult institutional-specific protocols.

Graft-versus-host disease, prevention

Graft-versus-host disease (GVHD), prevention: Limited data available; dosing regimens variable:

Continuous infusion: Infants, Children, and Adolescents: IV: Initial: 0.03 mg/kg/day (based on lean body weight) as continuous IV infusion; begin prior to pretransplant (specific day varies with protocol); monitor serum drug concentrations and adjust dose accordingly; convert to oral formulation when tolerated (Ref).

Intermittent infusion: Children and Adolescents: IV: 0.015 mg/kg/dose every 12 hours as a 2-hour infusion. Treatment was initiated on day −2 or day −3 prior to transplant and continued until oral medication tolerated; dosing based on a descriptive study in 33 patients (median age: 10 years; range: 1.2 to 17 years); during the trial, no infusion related reactions were reported and no higher incidence of toxicity or adverse effects reported (Ref).

Nephrotic syndrome, steroid-resistant or steroid-dependent

Nephrotic syndrome, steroid-resistant or steroid-dependent: Limited data available: Children and Adolescents: Oral: Immediate release: Initial: 0.1 to 0.2 mg/kg/day in divided doses every 12 hours; maximum initial dose: 4 to 5 mg/dose; adjust dose to target serum concentration (Ref).

Transplant, heart; prevention of rejection

Transplant, heart; prevention of rejection: Infants, Children, and Adolescents: Dose should be individualized and titrated to target trough concentration:

Oral: Immediate release: Initial: 0.1 to 0.3 mg/kg/day divided every 12 hours (Ref); Note: Lower initial dose can be used in patients who receive cell-depleting induction treatment.

IV: Limited data available: 0.01 to 0.03 mg/kg/day as a continuous IV infusion (Ref). Note: Limit parenteral use to patients unable to take enteral formulations; patients should be switched to an enteral dosage form as soon as possible.

Transplant, kidney; prevention of rejection

Transplant, kidney; prevention of rejection: Note: Although manufacturer labeling suggests delaying tacrolimus initiation until renal function has recovered, current practice and expert recommendations are to initiate tacrolimus before or at the time of transplant (Ref).

Oral: Dose should be individualized and titrated to target trough concentrations:

Immediate release: Children and Adolescents: Initial: 0.2 to 0.3 mg/kg/day divided every 12 hours (Ref).

Extended release:

Astagraf XL: Used in combination with corticosteroids and mycophenolate:

With basiliximab induction:

Children ≥4 years and Adolescents <16 years: Initial: 0.3 mg/kg once daily; administer first dose within 24 hours of reperfusion. Titrate to target trough concentrations.

Adolescents ≥16 years: Initial: 0.15 to 0.2 mg/kg once daily; administer first dose prior to reperfusion or within 48 hours of transplant. Titrate to target trough concentrations.

Without basiliximab induction:

Adolescents ≥16 years:

Preoperative dose: 0.1 mg/kg once; administer within 12 hours prior to reperfusion.

Postoperative dose: 0.2 mg/kg once daily; administer first postoperative dose at least 4 hours after preoperative dose and within 12 hours of reperfusion. Titrate to target trough concentrations.

Conversion from immediate release to extended release (Astagraf XL): Children ≥5 years and Adolescents: Oral: Initiate extended-release treatment in a 1:1 ratio (mg:mg) using previously established total daily dose of immediate release (Ref). Administer once daily. Note: If converting from IV tacrolimus, administer the first extended-release dose 8 to 12 hours after discontinuation of IV (Ref).

Envarsus XR:

Conversion from immediate release to extended release (Envarsus XR): Very limited data: Adolescents: Oral: Administer 60% to 80% of the total daily dose of immediate-release tacrolimus. Titrate to target trough concentrations (Ref).

IV: Limited data available: Infants, Children, and Adolescents: 0.06 mg/kg/day as a continuous IV infusion (Ref). Note: Limit parenteral use to patients unable to take enteral formulations; patients should be switched to an enteral dosage form as soon as possible.

Transplant, liver; prevention of rejection

Transplant, liver; prevention of rejection: Infants, Children, and Adolescents:

Oral: Immediate release: Initial: 0.15 to 0.2 mg/kg/day divided every 12 hours

IV: 0.03 to 0.05 mg/kg/day as a continuous IV infusion. Note: Limit parenteral use to patients unable to take enteral formulations; patients should be switched to an enteral dosage form as soon as possible (initiate 8 to 12 hours after infusion is stopped).

Transplant, lung; prevention of rejection

Transplant, lung; prevention of rejection:

Infants, Children, and Adolescents:

Oral: Immediate release: Initial: 0.3 mg/kg/day divided every 12 hours; if antibody induction treatment administered, reduce dose to 0.1 mg/kg/day. Patients with lung transplant secondary to cystic fibrosis may require higher doses due to reduced bioavailability.

Dose conversion of different routes of administration:

Conversion of IV continuous infusion to immediate-release oral: GVHD prophylaxis: Limited data available: To convert from IV to oral dose (1:4 ratio): Multiply total daily IV dose by 4 and administer in 2 divided oral doses per day, every 12 hours; titrate to goal trough concentration (Ref).

Conversion of immediate-release oral to sublingual (using immediate-release capsule): Very limited data available: To convert from oral to sublingual dose (2:1 ratio): Divide oral dose by 2 for sublingual administration. Dosing based on experience in adult solid organ transplant recipients unable to tolerate enteral administration (Ref).

Dosing: Kidney Impairment: Pediatric

Note: Dose adjustment should be guided by therapeutic drug monitoring.

Kidney impairment prior to treatment initiation: IV, Oral: Infants, Children, and Adolescents:

Altered kidney function: Initiate with lower end of dosing range. Kidney impairment does not affect the elimination or serum concentrations of tacrolimus; however, tacrolimus may cause nephrotoxicity requiring dose reduction.

Hemodialysis: Unlikely to be significantly dialyzed; no initial dosage adjustment has been recommended.

Peritoneal dialysis: Based on adult data, tacrolimus is not removed by peritoneal dialysis (Ref).

CRRT: IV, Oral: Significant drug removal is unlikely based on adult studies (Ref).

Nephrotoxicity during treatment: Treatment decisions should be individualized and in consultation with nephrology and/or transplant specialists.

Dosing: Hepatic Impairment: Pediatric

Mild impairment:

Immediate-release tacrolimus: Infants, Children, and Adolescents: IV, Oral: There are no dosage recommendations in the manufacturer's labeling.

Astagraf XL: Children ≥5 years and Adolescents: Oral: No dose adjustment necessary.

Moderate impairment:

Immediate-release tacrolimus: Infants, Children, and Adolescents: IV, Oral: There are no dosage recommendations in the manufacturer's labeling.

Astagraf XL: Children ≥5 years and Adolescents: Oral: Monitor levels closely.

Severe impairment:

Immediate-release tacrolimus: Infants, Children, and Adolescents: IV, Oral: There are no dosage recommendations provided in the manufacturer's labeling; however, clearance is decreased in severe impairment; close monitoring is recommended.

Astagraf XL: Children ≥5 years and Adolescents: Oral: Lower initial dose recommended due to decreased clearance.

Liver transplant: Use of tacrolimus in liver transplant recipients experiencing post-transplant hepatic impairment may be associated with increased risk of developing kidney insufficiency related to high whole blood levels of tacrolimus; monitor closely and adjust dosing as needed.

Adverse Reactions (Significant): Considerations
Diabetes mellitus

In general, there is an increased risk for the development of diabetes mellitus following transplantation (also known as post-transplant diabetes mellitus [PTDM]) (Ref). The cause of PTDM is multifactorial, of which one modifiable risk factor is the use of calcineurin inhibitors (CNI) (eg, cyclosporine, tacrolimus) (Ref). When compared to cyclosporine, tacrolimus may have a higher incidence of new-onset diabetes and/or impaired fasting glucose (Ref); however dosing strategies employed in early trials comparing the two CNIs may not reflect contemporary practice (ie, CNI-minimization or avoidance strategies) (Ref). While the impact of CNIs on pancreatic beta cells may be reversible (Ref), the risk of graft rejection must be considered when adjusting immunosuppressants in transplant recipients (Ref). Clinicians should note that PTDM is different than the hyperglycemia that is often seen in first few weeks following transplant.

Mechanism: Time-related; exact mechanism has not been fully elucidated. Potential mechanisms include beta cell dysfunction and increased insulin resistance (Ref). Clinicians should note that the cause of PTDM is multifactorial; the use of a CNI is one factor.

Onset: Varied; PTDM may develop early (ie, ≤3 months) or years after transplantation (Ref).

Risk factors:

• Presence of other risk factors for type 2 diabetes

• High trough tacrolimus concentrations (eg, >15 ng/mL during the first month after transplantation) (Ref)

• Black or Hispanic kidney transplant recipients

• Hepatitis C virus infection (Ref)

Drug-induced thrombotic microangiopathy

Tacrolimus use for solid organ and hematopoietic stem cell transplantation may cause drug-induced thrombotic microangiopathy (DITMA) which can result in microangiopathic hemolytic anemia with thrombocytopenia (MAHA), microvascular thrombosis with vessel wall abnormalities (Al-Nouri 2015, Boyer 2013, Elfeky 2020, Jumani 2004, Kishida 2016, Nwaba 2013, Parissis 2010). Patients generally present with kidney impairment and hypertension (George 2014); however, other organ systems may also be impacted by tacrolimus-related DITMA (eg, pulmonary, dermatologic, musculoskeletal, hepatic, gastrointestinal) (Chiurchiu 2002). Graft failure may or may not occur (Karolin 2021). Resolution can be achieved with early detection of the syndrome, tacrolimus discontinuation or dosage reduction, and initiation of syndrome-specific supportive treatment; however, kidney impairment may persist (George 2014).

Mechanism: Dose- and time-dependent; secondary to endothelial dysfunction and increased platelet aggregation (George 2014).

Onset: Varied; characterized by a gradual onset of kidney failure that occurs over weeks or months (George 2014).

Risk factors:

• In general, risk factors for thrombotic microangiopathy include (Naesens 2009):

- Ischemia-reperfusion endothelial injury

- Severe infections

- Kidney infections

- Vascular rejection or graft-versus-host disease

- Human leukocyte antigen mismatch

- Anticardiolipin antibodies

- Malignancies

- Concomitant use of other drugs known to cause DITMA (eg, mTOR inhibitors, antiviral agents)

Hyperkalemia

Mild to severe hyperkalemia may occur in patients who receive a calcineurin inhibitor (CNI), including tacrolimus; one retrospective trial described more severe as well as a longer duration of hyperkalemia in patients treated with tacrolimus as compared to patients who received cyclosporin (Ref). CNI-induced hyperkalemia is commonly associated with hypertension, hyperchloremia, metabolic acidosis, and hypercalciuria (Ref).

Mechanism: Dose-related. CNIs may cause a hyporeninemic hypoaldosteronism (type 4 renal tubular acidosis) which is a result of tubular insensitivity to aldosterone and a subsequent decrease in the activity of the renin-angiotensin-aldosterone system (Ref). In addition, CNIs directly impact the ability to excrete potassium, in part via inhibition of the renal outer medullary potassium (ROMK) channel and Na+/K+-ATPase and activation of the chloride shunt mechanism (Ref). Consideration should also be made for the impact of kidney transplantation (eg, delayed graft function) and concomitant drugs (eg, trimethoprim) on potassium balance (Ref).

Onset: Varied; may occur soon after transplant or later during the course of therapy (Ref)

Risk factors:

• Concomitant use of other drugs that may cause hyperkalemia (eg, potassium-sparing diuretics, angiotensin-converting enzyme inhibitors, trimethoprim/sulfamethoxazole)

Hypersensitivity reactions (immediate and delayed)

Immediate hypersensitivity reactions, including anaphylaxis, have been reported (Ref). Delayed hypersensitivity reactions, including generalized pruritus, eczematous drug eruption, symmetric drug-related intertriginous and flexural exanthema, and contact urticaria following topical application have also been reported (Ref). In addition, use of systemic tacrolimus in post-transplant recipients has been associated with a greater occurrence of IgE-mediated sensitization and possible increased risk of allergic disease, including food allergies (Ref).

Mechanism:

Immediate hypersensitivity reactions: Non–dose-related; likely related to polyoxyl 60 hydrogenated castor oil, a solubilizer contained in IV tacrolimus (Ref).

Delayed hypersensitivity reactions: Non–dose-related; immunologic (ie, involving a T-cell mediated drug-specific immune response) (Ref).

Onset:

Immediate hypersensitivity reactions: Rapid; generally occurs within 1 hour of administration (Ref).

Delayed hypersensitivity reactions: Varied; contact urticaria has occurred after 2 days of topical tacrolimus application (Ref), and generalized rashes have been documented up to 30 months after initiation of treatment (Ref). New food allergies in post-transplant recipients who are treated with tacrolimus may occur after many months of therapy (Ref).

Risk factors:

• Greater IgE-mediated sensitization in patients treated with tacrolimus compared to cyclosporine (Ref)

• Macrolide cross-sensitivity: Although tacrolimus is a macrolide drug, cross-reactivity is unlikely to occur (Ref), albeit one case of possible cross-reactivity has been reported (Ref).

Hypertension

In general, there is an increased risk for the development of hypertension following kidney transplantation (Ref). The cause of post-transplant hypertension is multifactorial, of which one modifiable risk factor is the use of calcineurin inhibitors (CNI) (eg, cyclosporine, tacrolimus) (Ref). Tacrolimus-based regimens may be associated with a lower incidence of hypertension as compared to cyclosporine-based regimens (Ref); however, the risk of graft rejection must be considered when adjusting immunosuppressants in transplant recipients (Ref). Hypertension may lead to adverse short-term and long-term allograft outcomes as well as increased cardiovascular morbidity and mortality (Ref).

Mechanism: Exact mechanism is unknown. Multiple mechanisms may contribute including tubular salt reabsorption, peripheral vasoconstriction, and the sympathetic nervous system (Ref). Vasoconstriction is caused by an increase of vasoconstrictor mediators (eg, sympathetic tone, renin-angiotensin system, endothelin-1) and a decrease of vasodilator mediators (eg, prostaglandins, nitric oxide) (Ref). Tacrolimus may activate the sodium chloride cotransporter in the kidney; this effect appears to be mediated, in part, by the kinases WNK4 and SPAK (Ref). Clinicians should note that the cause of post-transplant hypertension is multifactorial; the use of a CNI is one factor.

Onset: Varied; may occur soon after transplant or later during the course of therapy (Ref).

Risk factors:

In general, demographic and transplant-specific risk factors for post-transplant hypertension include (Ref):

• Preexisting hypertension

• Elevated body mass index

• Males

• Black patients

• Older donor age

• Delayed graft function

• Glucocorticoid use

• Recurrent disease

• Acute rejection

• Post-transplant proteinuria

Infection

Tacrolimus is an immunosuppressant; therefore, use may result in bacterial, viral, fungal, and protozoal infections, including opportunistic infections. Infections may be severe and potentially fatal.

Viral infections reported with use of tacrolimus include polyomavirus infection (which may result in polyomavirus associated nephropathy [PVAN]), JC virus-associated progressive multifocal leukoencephalopathy, cytomegalovirus disease, and reactivation of hepatitis B or hepatitis C (Ref). PVAN, primarily from activation of BK virus, may lead to the deterioration of kidney function and/or kidney graft loss, especially in kidney transplant recipients (Ref). Risk of infectious complications in transplant recipients may be higher with calcineurin inhibitors (CNIs), including tacrolimus, as compared to mTOR inhibitors (eg, sirolimus, everolimus)(Ref); however, tacrolimus may be associated with a lower risk of infections as compared to glucocorticoids or other immunosuppressants (eg, cyclophosphamide) when used for the treatment of lupus nephritis or idiopathic membranous nephropathy (Ref).

Mechanism: Exact mechanism unknown; related to pharmacologic action. Tacrolimus inhibits T-lymphocyte activation by interacting with an intracellular protein, FKBP-12 and calcineurin dependent proteins to inhibit calcineurin phosphatase activity.

Onset: Varied; in general, the onset of infections following solid organ transplant varies greatly; the majority of clinically important infections occur within the first 180 days (Ref).

Risk factors:

• Concomitant use of other immunosuppressive agents or preexisting immune function impairment

• Increased tacrolimus exposure

CMV infection: Transplant recipients that are CMV seronegative at the time of transplant who receive a graft from a CMV seropositive donor

Malignancy

In general, use of immunosuppressive agents may result in an increased risk of lymphoproliferative disorders and/or neoplasms (including skin carcinoma) (Ref). The risk of malignant lymphoma may be higher in transplant recipients who receive tacrolimus as compared to cyclosporine (Ref); however, not all studies have observed this pattern (Ref).

Mechanism: Dose- and time-related; related to the pharmacologic action. Tacrolimus inhibits T-lymphocyte activation by interacting with an intracellular protein, FKBP-12 and calcineurin dependent proteins to inhibit calcineurin phosphatase activity.

Onset: Delayed. Post-transplant lymphoproliferative disorders (PTLD) and non-skin, non-lymphoma malignancies generally occur during the first year after transplant when immunosuppressive therapy is most aggressive; however, the incidence of skin malignancies increase more linearly with time (Ref).

Risk factors:

• Intensity of immunosuppression (Ref)

• Concomitant use of other immunosuppressive agents or preexisting immune function impairment

• Epstein-Barr virus (EBV) seronegative transplant recipients (Ref)

- In general, pediatric transplant recipients are at a higher risk of PTLD as these patients are more likely to be EBV seronegative at transplantation (Ref)

- Clinicians may also consider the role that EBV acute infection and/or reactivation may play in the risk of EBV-associated lymphoma in patients receiving immunosuppressive therapies; risk not well defined (Ref)

• Pretransplant malignancy (Ref)

• Fewer HLA matches (Ref)

• History of at least 1 prior rejection episode (Ref)

• Age <25 years or >60 years (Ref)

Skin carcinoma: Exposure to sunlight/UV light

Nephrotoxicity

Use of calcineurin inhibitors (CNIs), including tacrolimus, may result in acute or chronic nephrotoxicity. Tacrolimus is thought to have a lower nephrotoxicity potential as compared to cyclosporine; however, not all studies have observed this benefit (Ref).

Acute CNI nephrotoxicity: Considered to be acute, functional, dose-dependent, and generally reversible after dose reduction or discontinuation; characterized by increased serum creatinine concentrations, hyperkalemia, and/or hyperuricemia unexplained by other causes for acute kidney injury (AKI) with a concomitant increase in tacrolimus trough concentration to >20 ng/mL (Ref). Kidney biopsy may or may not reveal an acute arteriolopathy and/or tubular vacuolization (Ref). Acute microvascular toxicity may also occur and is differentiated by the presence of glomerular capillary thrombosis as well as thrombi in the arterioles; presentation may range from asymptomatic to hemolytic uremic syndrome (Ref).

Chronic tacrolimus nephrotoxicity (CTN): Considered to be chronic, structural, progressive, and irreversible; all three compartments of the kidney (vessels, tubulo-interstitium, glomeruli) may be involved (Ref). Nodular hyaline deposits in the media of afferent arterioles is considered a hallmark; kidney biopsy may reveal arteriolar hyalinosis, tubular atrophy and interstitial fibrosis (“striped fibrosis”), glomerular capsular fibrosis, global and focal segmental glomerulosclerosis, juxtaglomerular apparatus hyperplasia, and/or tubular microcalcifications (Ref). Additional extra-renal symptoms may be present due to tubular functional alterations and ion homeostasis (eg, hyperkalemia, hypomagnesemia, hyperchloremic metabolic acidosis, hyperuricemia); hypertension may also occur (Ref).

Mechanism: Exact mechanism has not been fully elucidated; however, the main mechanisms are changes in glomerular and tubular function with the main targets being tubular epithelial cells, vascular endothelial cells, arteriolar myocytes, and interstitial fibroblasts (Ref).

Acute CNI nephrotoxicity: Dose-related (Ref); secondary to vasoconstriction of the afferent and efferent glomerular arterioles and reductions in renal blood flow and GFR resulting in hypoperfusion and parenchymal ischemia. Vasoconstriction is caused by an increase of vasoconstrictor mediators (eg, sympathetic tone, renin-angiotensin system, endothelin-1) and a decrease of vasodilator mediators (eg, prostaglandins, nitric oxide) (Ref).

CTN: Dose- and time-related; exact mechanism is unknown; a combination of hemodynamic changes and direct toxic effects on tubular epithelial cells is thought to play a role (Ref). Nodular hyaline deposits may cause narrowing of the vascular lumen which contributes to the development of interstitial fibrosis, tubular atrophy, and glomerular sclerosis. In addition, renal vasoconstriction may result in local hypoxia or ischemia and subsequent cellular injury. Upregulation of transforming growth factor-beta (TGF-β) and activation of the renin-angiotensin system may also contribute to the tubular-interstitial effects seen in patients with CTN. Furthermore, CNIs may be directly toxic to tubular and interstitial cells as well cause a decrease in the excretion of other endogenous toxic substances via inhibition of P-glycoprotein (multidrug resistance protein 1 [MR1] or ATP-binding cassette subfamily B, member 1 [ABCB1]).

Onset: Varied; in one trial, the acute phase of CNI nephrotoxicity occurred with a median onset of 6 months post-transplantation, whereas the chronic phase occurred at a median onset of 3 years (Ref).

Risk factors:

• Systemic overexposure to tacrolimus (Ref)

• Increased local exposure to tacrolimus (eg, due to ABCB1 genotype or expression in renal tubular epithelial cells or drug interactions) (Ref)

• Increased exposure to metabolites of tacrolimus (eg, due to CYP3A4/5 genotype or expression in the renal tubular epithelial cells or drug interactions) (Ref)

• Older kidney age (Ref)

• Concomitant use of other nephrotoxic drugs, particularly nonsteroidal anti-inflammatory drugs (Ref)

• Genetic polymorphisms of other genes (eg, TGF-β, ACE), including patients with FOXP3 rs3761548 AA and AC genotypes (Ref)

• Patients with high tacrolimus clearance may be at an increased risk of interstitial fibrosis and tubular atrophy; intermittent underexposure may lead to alloimmune activation and subsequent fibrosis (Ref)

Neurotoxicity

Dose-related neurotoxicity may occur with use of calcineurin inhibitors (CNIs), including tacrolimus. Neurotoxicity may be more common with immediate-release tacrolimus as compared to cyclosporine (Ref).

Mild symptoms may include tremor, neuralgia, peripheral neuropathy, headache, mood changes (agitation, anxiety), and insomnia (Ref). Tremor is a common manifestation and generally involves the upper extremities, especially upon extension (Ref); tremor may be more common with immediate-release tacrolimus as compared to extended-release formulations (Ref). Headaches are often described as migrainous, occipital, and frequent (eg, weekly) (Ref).

More severe symptoms of neurotoxicity may occur less frequently and include seizures, visual disturbance (including cortical blindness), dysarthria and akinetic mutism, severe psychomotor disturbance, stupor, coma, delusions, psychosis, hallucinations, cerebellar ataxia, or asthenia (Ref).

Reversible posterior leukoencephalopathy syndrome (RPLS) has been rarely reported (Ref); symptoms (mental status changes, headache, hypertension, seizures, and visual disturbances) are reversible with dose reduction or discontinuation of therapy.

Mechanism: Dose-related; exact mechanism is unknown. Tacrolimus-induced neurotoxicity may be related to the ability of CNIs to increase the permeability of the blood brain barrier (BBB) via multiple mechanisms; once within the CNS, tacrolimus may cause neurotoxicity via calcineurin inhibition (Ref).

Onset: Varied; CNI-induced neurotoxicity may occur within weeks after transplantation (Ref). Onset of RPLS has been described days to years post-transplant; onset may differ depending on the transplanted organ (Ref).

Risk factors:

• Increased tacrolimus exposure; specifically dosing strategies that result in high peak concentrations (eg, immediate-release formulations, IV administration) or alterations in clearance or metabolism (eg, advanced liver failure, drug interactions) (Ref)

• Increased donor age (Ref)

• Prior history of hepatic encephalopathy in liver transplant recipients (Ref)

RPLS: Risk is increased in patients with significant fluid overload, elevated blood pressure, or impaired kidney function (Ref)

Pure red cell aplasia

Pure red cell aplasia (PRCA), a type of anemia which can range from subclinical to severe, and is a relatively rare disease that has been reported in patients receiving tacrolimus concomitantly with other immunosuppressive agents (eg, corticosteroids, mycophenolate mofetil) (Ref). Symptoms may include fatigue, lethargy, or pallor. Occurrence of PRCA may warrant treatment interruption, dosage adjustment, or discontinuation (Ref); the risk of reduced immunosuppression and graft rejection should be considered in transplant recipients.

Mechanism: Exact mechanism unknown (Ref); the impact of concurrent disease processes (eg, parvovirus B19 infection) should be considered (Ref).

Onset: Delayed (Ref)

Risk factors:

• Concomitant use of other immunosuppressive agents or preexisting immune function impairment

Adverse Reactions

The following adverse drug reactions and incidences are derived from product labeling unless otherwise specified. Adverse reactions reported with combination therapy.

>10%:

Cardiovascular: Acute cardiorespiratory failure, angina pectoris, atrial fibrillation, atrial flutter, bradycardia, cardiac arrhythmia, cardiac fibrillation, chest pain, deep vein thrombophlebitis, deep vein thrombosis, ECG abnormality (including abnormal QRS complex and ST segment changes on ECG), edema, flushing, heart failure, hypertension (table 1), hypotension, orthostatic hypotension, peripheral edema, phlebitis, syncope, tachycardia, thrombosis, vasodilation

Tacrolimus (Systemic): Adverse Reaction: Hypertension

Drug (Tacrolimus)

Comparator (various)

Population

Dosage Form (Tacrolimus)

Dosage Form (Comparator)

Indication

Number of Patients (Tacrolimus)

Number of Patients (Comparator)

39%

47%

Children & adolescents

Granules

N/A

Liver transplant

91

90

32%

35%

Adolescents & adults

N/A

N/A

Kidney transplant

212

212

28%

30%

Adolescents & adults

Extended release

Immediate release

Kidney transplant

214

212

47%

56%

Adolescents & adults

N/A

N/A

Liver transplant

250

250

62%

69%

Adults

N/A

N/A

Heart transplant

157

157

50%

52%

Adults

N/A

N/A

Kidney transplant

205

207

23%

23%

Adults

Extended release

Immediate release

Kidney transplant

268

275

Dermatologic: Acne vulgaris, alopecia, benign skin neoplasm, cellulitis, condyloma acuminatum, dermal ulcer, dermatitis, diaphoresis, ecchymoses, exfoliative dermatitis, fungal dermatitis, hyperhidrosis, hypotrichosis, pruritus, skin discoloration, skin photosensitivity, skin rash, tinea versicolor

Endocrine & metabolic: Acidosis, albuminuria, alkalosis, anasarca, Cushing syndrome, cushingoid appearance, decreased serum bicarbonate, decreased serum iron, dehydration, diabetes mellitus (including new-onset) (table 2), hirsutism, hypercalcemia, hypercholesterolemia, hyperglycemia, hyperkalemia (table 3), hyperphosphatemia, hypertriglyceridemia, hyperuricemia (table 4), hypervolemia, hypocalcemia, hypoglycemia, hypokalemia, hypomagnesemia (table 5), hyponatremia, hypophosphatemia, increased lactate dehydrogenase, metabolic acidosis (table 6), weight gain

Tacrolimus (Systemic): Adverse Reaction: Diabetes Mellitus

Drug (Tacrolimus)

Comparator (various)

Population

Dosage Form (Tacrolimus)

Dosage Form (Comparator)

Indication

Number of Patients (Tacrolimus)

Number of Patients (Comparator)

75%

61%

Adolescents & adults

N/A

N/A

Kidney transplant

212

212

36%

35%

Adolescents & adults

Extended release

N/A

Kidney transplant

162

151

26%

16%

Adults

N/A

N/A

Heart transplant

157

157

24%

9%

Adults

N/A

N/A

Kidney transplant

205

207

16%

14%

Adults

Extended release

Immediate release

Kidney transplant

268

275

13%

7%

N/A

N/A

N/A

Heart transplant

75

83

20%

4%

N/A

N/A

N/A

Kidney transplant

151

151

18%

13%

N/A

N/A

N/A

Liver transplant

239

236

Tacrolimus (Systemic): Adverse Reaction: Hyperkalemia

Drug (Tacrolimus)

Comparator (various)

Population

Dosage Form (Tacrolimus)

Dosage Form (Comparator)

Indication

Number of Patients (Tacrolimus)

Number of Patients (Comparator)

12%

10%

Children & adolescents

Granules

N/A

Liver transplant

91

90

26%

19%

Adolescents & adults

N/A

N/A

Kidney transplant

212

212

20%

23%

Adolescents & adults

Extended release

Immediate release

Kidney transplant

214

212

45%

26%

Adolescents & adults

N/A

N/A

Liver transplant

250

250

31%

32%

Adults

N/A

N/A

Kidney transplant

205

207

15%

11%

Adults

Extended release

Immediate release

Kidney transplant

268

275

Tacrolimus (Systemic): Adverse Reaction: Hyperuricemia

Drug (Tacrolimus)

Comparator (various)

Population

Dosage Form (Tacrolimus)

Dosage Form (Comparator)

Indication

Number of Patients (Tacrolimus)

Number of Patients (Comparator)

<15%

N/A

Adolescents & adults

Immediate release

N/A

Kidney transplant

N/A

N/A

Tacrolimus (Systemic): Adverse Reaction: Hypomagnesemia

Drug (Tacrolimus)

Comparator (various)

Population

Dosage Form (Tacrolimus)

Dosage Form (Comparator)

Indication

Number of Patients (Tacrolimus)

Number of Patients (Comparator)

40%

29%

Children & adolescents

Granules

N/A

Liver transplant

91

90

28%

22%

Adolescents & adults

N/A

N/A

Kidney transplant

212

212

24%

27%

Adolescents & adults

Extended release

Immediate release

Kidney transplant

214

212

48%

45%

Adolescents & adults

N/A

N/A

Liver transplant

250

250

34%

17%

Adults

N/A

N/A

Kidney transplant

205

207

12%

12%

Adults

Extended release

Immediate release

Kidney transplant

268

275

Tacrolimus (Systemic): Adverse Reaction: Metabolic Acidosis

Drug (Tacrolimus)

Comparator (various)

Population

Dosage Form (Tacrolimus)

Dosage Form (Comparator)

Indication

Number of Patients (Tacrolimus)

Number of Patients (Comparator)

26%

17%

Children & adolescents

Granules

N/A

Liver transplant

91

90

Gastrointestinal: Abdominal distention, abdominal pain, anorexia, aphthous stomatitis, biliary tract disease, cholangitis, cholestasis, constipation, diarrhea, duodenitis, dyspepsia, dysphagia, esophagitis, flatulence, gastritis, gastroenteritis, gastroesophageal reflux disease, gastrointestinal hemorrhage, gastrointestinal perforation, hernia of abdominal cavity, hiccups, increased appetite, intestinal obstruction, nausea, oral candidiasis, pancreatic pseudocyst, peritonitis, stomatitis, ulcerative esophagitis, vomiting

Genitourinary: Anuria, bladder spasm, cystitis, dysuria, hematuria, nocturia, oliguria, proteinuria, pyuria, toxic nephrosis, urinary frequency, urinary incontinence, urinary retention, urinary tract infection, urinary urgency, vaginitis

Hematologic & oncologic: Anemia, disorder of hemostatic components of blood, hemolytic anemia, hemorrhage, hypochromic anemia, hypoproteinemia, hypoprothrombinemia, increased hematocrit, Kaposi sarcoma, leukocytosis, leukopenia, neutropenia, polycythemia, thrombocytopenia, thrombotic microangiopathy

Hepatic: Abnormal hepatic function tests, ascites, cholestatic jaundice, granulomatous hepatitis, hepatitis (including acute and chronic), hepatotoxicity, hyperbilirubinemia, increased gamma-glutamyl transferase, increased liver enzymes, increased serum alanine aminotransferase, increased serum alkaline phosphatase, increased serum aspartate aminotransferase, jaundice

Hypersensitivity: Hypersensitivity reaction

Immunologic: Graft complications

Infection: Abscess, bacterial infection (may be serious) (table 7), BK virus (including nephropathy) (table 8), candidiasis, cytomegalovirus disease (including CMV viremia) (table 9), Epstein-Barr infection, herpes simplex infection, herpes zoster infection, infection (table 10) (including serious infection) (table 11), opportunistic infection (table 12), polyomavirus infection (table 13), sepsis (children & adolescents)

Tacrolimus (Systemic): Adverse Reaction: Bacterial Infection

Drug (Tacrolimus)

Comparator (various)

Population

Dosage Form (Tacrolimus)

Dosage Form (Comparator)

Indication

Number of Patients (Tacrolimus)

Number of Patients (Comparator)

8%

12%

Adolescents & adults

Extended release

Immediate release

Kidney transplant

214

212

30%

N/A

Adults

N/A

N/A

Heart transplant

331

N/A

13%

18%

Adults

Extended release

Immediate release

Kidney transplant

268

275

7%

5%

Adults

Extended release

Immediate release

Kidney transplant

162

162

Tacrolimus (Systemic): Adverse Reaction: BK Virus

Drug (Tacrolimus)

Comparator (various)

Population

Dosage Form (Tacrolimus)

Dosage Form (Comparator)

Indication

Number of Patients (Tacrolimus)

Number of Patients (Comparator)

6%

9%

Adults

Extended release

Immediate release

Kidney transplant

268

275

Tacrolimus (Systemic): Adverse Reaction: Cytomegalovirus Disease

Drug (Tacrolimus)

Comparator (various)

Population

Dosage Form (Tacrolimus)

Dosage Form (Comparator)

Indication

Number of Patients (Tacrolimus)

Number of Patients (Comparator)

15%

24%

Children & adolescents

Granules

N/A

Liver transplant

91

90

10%

12%

Adolescents & adults

Extended release

Immediate release

Kidney transplant

214

212

32%

30%

Adults

N/A

N/A

Heart transplant

157

157

11%

9%

Adults

Extended release

Immediate release

Kidney transplant

268

275

2%

1%

Adults

Extended release

Immediate release

Kidney transplant

162

162

Tacrolimus (Systemic): Adverse Reaction: Infection

Drug (Tacrolimus)

Comparator (various)

Population

Dosage Form (Tacrolimus)

Dosage Form (Comparator)

Indication

Number of Patients (Tacrolimus)

Number of Patients (Comparator)

25%

29%

Children & adolescents

Granules

N/A

Liver transplant

91

90

56%

N/A

Children & adolescents

N/A

N/A

N/A

76

N/A

69%

69%

Adolescents & adults

Extended release

Immediate release

Kidney transplant

214

212

24%

21%

Adults

N/A

N/A

Heart transplant

157

157

70%

65%

Adults

Extended release

Immediate release

Kidney transplant

268

275

46%

48%

Adults

Extended release

Immediate release

Kidney transplant

162

162

45%

49%

Adults

N/A

N/A

Kidney transplant

205

207

Tacrolimus (Systemic): Adverse Reaction: Serious Infection

Drug (Tacrolimus)

Comparator (various)

Population

Dosage Form (Tacrolimus)

Dosage Form (Comparator)

Indication

Number of Patients (Tacrolimus)

Number of Patients (Comparator)

22%

23%

Adolescents & adults

Extended release

Immediate release

Kidney transplant

214

212

26%

24%

Adults

Extended release

Immediate release

Kidney transplant

268

275

8%

9%

Adults

Extended release

Immediate release

Kidney transplant

162

162

Tacrolimus (Systemic): Adverse Reaction: Opportunistic Infection

Drug (Tacrolimus)

Comparator (various)

Population

Dosage Form (Tacrolimus)

Dosage Form (Comparator)

Indication

Number of Patients (Tacrolimus)

Number of Patients (Comparator)

15%

N/A

Adults

N/A

N/A

Heart transplant

331

N/A

Tacrolimus (Systemic): Adverse Reaction: Polyomavirus Infection

Drug (Tacrolimus)

Comparator (various)

Population

Dosage Form (Tacrolimus)

Dosage Form (Comparator)

Indication

Number of Patients (Tacrolimus)

Number of Patients (Comparator)

3%

5%

Adolescents & adults

Extended release

Immediate release

Kidney transplant

214

212

Nervous system: Abnormal dreams, abnormality in thinking, agitation, amnesia, anxiety, asthenia, ataxia, chills, confusion, depression, dizziness, drowsiness, emotional lability, encephalopathy, falling, fatigue, flaccid paralysis, hallucination, headache (table 14), hemorrhagic stroke, hypertonia, hypoesthesia, impaired mobility, insomnia (table 15), intolerance to temperature, mood elevation, myasthenia, myoclonus, nerve compression, nervousness, neuralgia, neuropathy, nightmares, pain, paralysis (monoparesis, quadriparesis, quadriplegia), paresthesia, peripheral neuropathy, psychomotor disturbance, psychosis, seizure, tremor (table 16), vertigo, voice disorder, writing difficulty

Tacrolimus (Systemic): Adverse Reaction: Headache

Drug (Tacrolimus)

Comparator (various)

Population

Dosage Form (Tacrolimus)

Dosage Form (Comparator)

Indication

Number of Patients (Tacrolimus)

Number of Patients (Comparator)

14%

N/A

Children & adolescents

N/A

N/A

N/A

76

N/A

24%

25%

Adolescents & adults

N/A

N/A

Kidney transplant

212

212

22%

24%

Adolescents & adults

Extended release

Immediate release

Kidney transplant

214

212

64%

60%

Adolescents & adults

N/A

N/A

Liver transplant

250

250

44%

38%

Adults

N/A

N/A

Kidney transplant

205

207

15%

10%

Adults

Extended release

Immediate release

Kidney transplant

268

275

Tacrolimus (Systemic): Adverse Reaction: Insomnia

Drug (Tacrolimus)

Comparator (various)

Population

Dosage Form (Tacrolimus)

Dosage Form (Comparator)

Indication

Number of Patients (Tacrolimus)

Number of Patients (Comparator)

30%

21%

Adolescents & adults

N/A

N/A

Kidney transplant

212

212

24%

28%

Adolescents & adults

Extended release

Immediate release

Kidney transplant

214

212

64%

68%

Adolescents & adults

N/A

N/A

Liver transplant

250

250

32%

30%

Adults

N/A

N/A

Kidney transplant

205

207

13%

11%

Adults

Extended release

Immediate release

Kidney transplant

268

275

Tacrolimus (Systemic): Adverse Reaction: Tremor

Drug (Tacrolimus)

Comparator (various)

Population

Dosage Form (Tacrolimus)

Dosage Form (Comparator)

Indication

Number of Patients (Tacrolimus)

Number of Patients (Comparator)

35%

34%

Adolescents & adults

Extended release

Immediate release

Kidney transplant

214

212

34%

20%

Adolescents & adults

N/A

N/A

Kidney transplant

212

212

56%

46%

Adolescents & adults

N/A

N/A

Liver transplant

250

250

15%

6%

Adults

N/A

N/A

Heart transplant

157

157

54%

34%

Adults

N/A

N/A

Kidney transplant

205

207

19%

17%

Adults

Extended release

Immediate release

Kidney transplant

268

275

Neuromuscular & skeletal: Arthralgia, back pain, gout, lower limb cramp, muscle cramps, muscle spasm, myalgia, osteoporosis

Ophthalmic: Amblyopia, blurred vision, conjunctivitis, visual disturbance

Otic: Otalgia, otitis media, tinnitus

Renal: Acute kidney injury, hydronephrosis, increased blood urea nitrogen, increased serum creatinine (table 17), nephrotoxicity (table 18), renal failure syndrome, renal insufficiency, renal tubular necrosis

Tacrolimus (Systemic): Adverse Reaction: Increased Serum Creatinine

Drug (Tacrolimus)

Comparator (various)

Population

Dosage Form (Tacrolimus)

Dosage Form (Comparator)

Indication

Number of Patients (Tacrolimus)

Number of Patients (Comparator)

46%

N/A

Children & adolescents

Extended release

Immediate release

Kidney transplant

13

N/A

23%

23%

Adolescents & adults

N/A

N/A

Kidney transplant

212

212

19%

23%

Adolescents & adults

Extended release

Immediate release

Kidney transplant

214

212

39%

25%

Adolescents & adults

N/A

N/A

Liver transplant

250

250

45%

42%

Adults

N/A

N/A

Kidney transplant

205

207

12%

14%

Adults

Extended release

Immediate release

Kidney transplant

268

275

12%

N/A

Adults

Extended release

Immediate release

Kidney transplant

162

162

Tacrolimus (Systemic): Adverse Reaction: Nephrotoxicity

Drug (Tacrolimus)

Comparator (various)

Population

Dosage Form (Tacrolimus)

Dosage Form (Comparator)

Indication

Number of Patients (Tacrolimus)

Number of Patients (Comparator)

40%

N/A

Adolescents & adults

N/A

N/A

Liver transplant

250

250

56%

N/A

Adults

N/A

N/A

Heart transplant

157

157

52%

N/A

Adults

N/A

N/A

Kidney transplant

205

207

Respiratory: Acute respiratory distress syndrome, asthma, atelectasis, bronchitis, decreased lung function, dyspnea, flu-like symptoms, increased cough, pharyngitis, pleural effusion, pneumonia, pneumothorax, productive cough, pulmonary edema, pulmonary emphysema, respiratory tract infection, rhinitis, sinusitis

Miscellaneous: Abnormal healing, crying, fever, postoperative wound complication, ulcer, wound healing impairment

1% to 10%:

Gastrointestinal: Gastrointestinal infection

Infection: Fungal infection (table 19)

Tacrolimus (Systemic): Adverse Reaction: Fungal Infection

Drug (Tacrolimus)

Comparator (various)

Population

Dosage Form (Tacrolimus)

Dosage Form (Comparator)

Indication

Number of Patients (Tacrolimus)

Number of Patients (Comparator)

9%

8%

Adults

Extended release

Immediate release

Kidney transplant

268

275

4%

4%

Adults

Extended release

Immediate release

Kidney transplant

162

162

Respiratory: Upper respiratory tract infection

Postmarketing:

Cardiovascular: Abnormal T waves on ECG, acute myocardial infarction, coronary artery vasospasm (Samer 2022), glomerular capillary thrombosis (Holman 1993), hypertrophic cardiomyopathy (Noda 2017), ischemic heart disease, pericardial effusion (including cardiac tamponade) (Doobay 2018), prolonged QT interval on ECG (Hodak 1998), pulmonary embolism, supraventricular extrasystole (Kim 2013), supraventricular tachycardia (Kim 2013), torsades de pointes (Hodak 1998), ventricular fibrillation, ventricular premature contractions

Dermatologic: Epidermal cyst of skin (Tiwari 2021), hyperpigmentation, malignant melanoma, Stevens-Johnson syndrome, toxic epidermal necrolysis, urticaria (Kamata 2009)

Endocrine & metabolic: Diabetic ketoacidosis (Maruyama 2019), hot flash, weight loss

Gastrointestinal: Biliary obstruction, colitis, delayed gastric emptying, enterocolitis, gastric ulcer, increased serum amylase, oral mucosa ulcer, pancreatitis (including acute pancreatitis, hemorrhagic pancreatitis, necrotizing pancreatitis) (Ding 2022; Xu 2019)

Genitourinary: Glycosuria, hemorrhagic cystitis, hypercalciuria (Farouk 2020)

Hematologic & oncologic: Agranulocytosis, decreased serum fibrinogen, disseminated intravascular coagulation, febrile neutropenia, hemolytic-uremic syndrome (Lin 2003), hepatosplenic T-cell lymphomas, immune thrombocytopenia (Gipson 2021), increased INR, leukemia, lymphoproliferative disorder (DiGiuseppe 1996), malignant lymphoma (Sekiguchi 2012), pancytopenia, prolonged partial thromboplastin time, pure red cell aplasia (Watanabe 2020), thrombotic thrombocytopenic purpura

Hepatic: Hepatic cirrhosis, hepatic cytolysis, hepatic failure, hepatic necrosis, hepatic sinusoidal obstruction syndrome (Shen 2015), hepatitis C (reactivation) (Liu 2014), liver steatosis

Immunologic: Epstein-Barr-associated lymphoproliferative disorder (Caillard 2006), graft- versus-host disease

Infection: Reactivation of HBV (Ridruejo 2010)

Nervous system: Aphasia, carpal tunnel syndrome, cerebral infarction (Kwun 2011), coma (Junna 2007), dysarthria (Vearrier 2011), dysphasia (Reyes 1990), encephalitis (autoimmune) (Spindel 2020), hemiparesis, intracranial hemorrhage (Shah 2022), jitteriness, leukoencephalopathy (Junna 2007), mutism (Vearrier 2011), neurotoxicity (Coe 2020), parkinsonism (Gmitterova 2018), progressive multifocal leukoencephalopathy (including JC virus-associated) (Bianchi 2020; Ischii 2019), reversible posterior leukoencephalopathy syndrome (including mental status changes) (Loar 2013), status epilepticus (Junna 2007)

Neuromuscular & skeletal: Inflammatory polyarthropathy, limb pain (including calcineurin-inhibitor induced pain syndrome) (Ayyala 2016), rhabdomyolysis

Ophthalmic: Blindness, cortical blindness (Steg 1999), optic atrophy, optic neuropathy (Nanda 2021), photophobia

Otic: Hearing loss (Gulleroglu 2013, Lakshmi 2020)

Respiratory: Allergic rhinitis, interstitial pulmonary disease, pulmonary hypertension, pulmonary infiltrates, respiratory distress, respiratory failure

Miscellaneous: Multiorgan failure

Contraindications

Hypersensitivity to tacrolimus, polyoxyl 60 hydrogenated castor oil (HCO-60), or any other component of the formulation.

Warnings/Precautions

Concerns related to adverse events:

• Cardiovascular: Myocardial hypertrophy has been reported; may be reversible with dose reduction or discontinuation. Prolongation of the QT/QTc and torsade de pointes may occur; avoid use in patients with congenital long QT syndrome.

• Gastrointestinal perforation: Gastrointestinal perforation may occur; all reported cases were considered to be a complication of transplant surgery or accompanied by infection, diverticulum, or malignant neoplasm.

Concurrent drug therapy:

• Everolimus combination therapy: In liver transplantation, the tacrolimus dose and target range should be reduced to minimize the risk of nephrotoxicity when used in combination with everolimus. Concurrent use may also increase the risk of thrombotic microangiopathy.

• Sirolimus combination therapy: ER tacrolimus in combination with sirolimus is not recommended in renal transplant recipients; the safety and efficacy of IR tacrolimus in combination with sirolimus has not been established in this patient population. Concomitant use was associated with increased mortality, graft loss, and hepatic artery thrombosis in liver transplant recipients, as well as increased risk of renal impairment, wound healing complications, and PTDM in heart transplant recipients. Concurrent use may also increase the risk of thrombotic microangiopathy.

Special populations:

• Patients with systemic lupus erythematosus (SLE) undergoing hip or knee replacement surgery: Patients with severe SLE (referring to patients with severe organ manifestations such as nephritis) should not interrupt therapy when undergoing hip or knee replacement surgery. For patients with SLE without severe disease, hold tacrolimus for at least 1 week prior to surgery to reduce infection risk; therapy can be restarted once surgical wound shows evidence of healing (eg, no swelling, erythema, or drainage), sutures/staples are removed, and no ongoing nonsurgical site infections (typically ~14 days to reduce infection risk) (ACR/AAHKS [Goodman 2022]).

Dosage form specific issues:

• Injection: Tacrolimus injection contains polyoxyl 60 hydrogenated castor oil (HCO-60), a castor oil derivative. HCO-60 is a solubilizer similar to polyoxyethylated castor oil (also known as polyoxyl 35 castor oil or Cremophor EL) (Nicolai 2012). Each mL of injection contains polyoxyl 60 hydrogenated castor oil (HCO-60) (200 mg) and dehydrated alcohol USP 80% v/v. Anaphylaxis has been reported with the injection, use should be reserved for those patients not able to take oral medications.

• Lactose: Oral formulations contain lactose.

Other warnings/precautions:

• Discontinuation of therapy: Myasthenia gravis: Abrupt cessation of this or any immunosuppressant, especially in clinically unstable individuals, may result in rapid deterioration of myasthenic symptoms and possibly myasthenic crisis (Melzer 2016).

• Error prevention: IR and ER capsules are NOT interchangeable or substitutable. The ER formulation is a once-daily preparation; and IR formulation is intended for twice-daily administration. Serious adverse events, including organ rejection, may occur if inadvertently substituted.

• Immunizations: Patients should be brought up to date with all immunizations before initiating therapy. Patients should not be immunized with live vaccines during or shortly after treatment and should avoid close contact with recently vaccinated (live vaccine) individuals. Inactivated vaccines may be administered (response may be diminished).

Dosage Forms Considerations

Prograf injection contains polyoxyl 60 hydrogenated castor oil (HCO-60)

Dosage Forms: US

Excipient information presented when available (limited, particularly for generics); consult specific product labeling.

Capsule, Oral:

Prograf: 0.5 mg, 1 mg, 5 mg

Generic: 0.5 mg, 1 mg, 5 mg

Capsule Extended Release 24 Hour, Oral:

Astagraf XL: 0.5 mg, 1 mg, 5 mg

Packet, Oral:

Prograf: 0.2 mg (50 ea); 1 mg (50 ea)

Solution, Intravenous:

Prograf: 5 mg/mL (1 mL) [contains alcohol, usp, peg-60 hydrog castor oil(cremophor rh60)]

Tablet Extended Release 24 Hour, Oral:

Envarsus XR: 0.75 mg, 1 mg, 4 mg

Generic Equivalent Available: US

May be product dependent

Pricing: US

Capsule ER 24 Hour Therapy Pack (Astagraf XL Oral)

0.5 mg (per each): $3.37

1 mg (per each): $6.73

5 mg (per each): $33.65

Capsules (Prograf Oral)

0.5 mg (per each): $4.31

1 mg (per each): $8.62

5 mg (per each): $43.10

Capsules (Tacrolimus Oral)

0.5 mg (per each): $1.26 - $2.23

1 mg (per each): $1.64 - $6.95

5 mg (per each): $10.80 - $22.30

Pack (Prograf Oral)

0.2 mg (per each): $6.24

1 mg (per each): $9.31

Solution (Prograf Intravenous)

5 mg/mL (per mL): $297.12

Tablet, 24-hour (Envarsus XR Oral)

0.75 mg (per each): $6.03

1 mg (per each): $8.04

4 mg (per each): $32.15

Disclaimer: A representative AWP (Average Wholesale Price) price or price range is provided as reference price only. A range is provided when more than one manufacturer's AWP price is available and uses the low and high price reported by the manufacturers to determine the range. The pricing data should be used for benchmarking purposes only, and as such should not be used alone to set or adjudicate any prices for reimbursement or purchasing functions or considered to be an exact price for a single product and/or manufacturer. Medi-Span expressly disclaims all warranties of any kind or nature, whether express or implied, and assumes no liability with respect to accuracy of price or price range data published in its solutions. In no event shall Medi-Span be liable for special, indirect, incidental, or consequential damages arising from use of price or price range data. Pricing data is updated monthly.

Dosage Forms: Canada

Excipient information presented when available (limited, particularly for generics); consult specific product labeling.

Capsule, Oral:

Prograf: 0.5 mg, 1 mg, 5 mg

Generic: 0.5 mg, 1 mg, 5 mg

Capsule Extended Release 24 Hour, Oral:

Advagraf: 0.5 mg, 1 mg, 3 mg, 5 mg

Solution, Intravenous:

Prograf: 5 mg/mL (1 mL) [contains alcohol, usp, polyoxyl/peg-35 castor oil(cremophor el)]

Tablet Extended Release 24 Hour, Oral:

Envarsus PA: 0.75 mg, 1 mg, 4 mg

Administration: Adult

IV: If IV administration is necessary, administer by continuous infusion (generally, over 24 hours). Do not use PVC tubing when administering diluted solutions. Do not mix with solutions with a pH ≥9 (eg, acyclovir or ganciclovir) due to chemical degradation of tacrolimus (use different ports in multilumen lines). Do not alter dose with concurrent T-tube clamping. Adsorption of the drug to PVC tubing may become clinically significant with low concentrations. Tacrolimus levels should not be drawn from a line that was used for a tacrolimus infusion. Falsely elevated tacrolimus levels may occur if drawn from the line used for the tacrolimus infusion; once a line (even if multi-lumen) is used for a tacrolimus IV infusion, it should be considered contaminated with tacrolimus (Ref).

Oral:

Immediate release: Administer with or without food; be consistent with timing and composition of meals if GI intolerance occurs and administration with food becomes necessary (per manufacturer). If dosed once daily, administer in the morning. If dosed twice daily, doses should be 12 hours apart. If the morning and evening doses differ, the larger dose (differences are never >0.5 to 1 mg) should be given in the morning. Some data suggest that the larger dose should be given in the evening because tacrolimus exposure may be reduced in the evening due to diurnal variation (Ref). If dosed 3 times daily, separate doses by 8 hours.

Combination therapy with everolimus for liver transplantation: Administer tacrolimus at the same time as everolimus.

Granules for oral suspension: Calculate dose based on patient weight; use the minimum whole number of packets for the required morning or evening dose. If the morning or evening dose is not covered by the whole number of packets, use one additional 0.2 mg packet to round up the dose. Do not use PVC-containing tubing, syringes, or cups to prepare or administer tacrolimus products; a non-PVC oral syringe (dispensed with prescription) may be used in younger patients. Do not sprinkle granules on food.

Empty the entire contents of each packet into a glass cup. Add 15 to 30 mL of room temperature drinking water; mix and administer the entire contents of the cup (granules will not completely dissolve). Administer immediately after preparation. For younger patients, the suspension can be drawn up via a non-PVC oral syringe (dispensed with prescription). Add additional 15 to 30 mL of water to rinse the cup or syringe to ensure all of the medication is taken.

Extended release: For all US preparations and Envarsus PA [Canadian product], administer on an empty stomach at least 1 hour before or 2 hours after a meal. Advagraf [Canadian product] labeling suggests that the capsule may be taken with food if necessary but should be administered consistently with or without food. Swallow whole, do not chew, crush, or divide. Take once daily in the morning at a consistent time each day. Missed doses may be taken up to 14 hours (15 hours for Envarsus XR) after scheduled time; if >14 hours (>15 hours for Envarsus XR), resume at next regularly scheduled time; do not double a dose to make up for a missed dose.

Bariatric surgery: Capsule and tablet, extended release: Some institutions may have specific protocols that conflict with these recommendations; refer to institutional protocols as appropriate. IR capsule, oral solution, and injectable formulations are available. If safety and efficacy can be effectively monitored, no change in formulation or administration is required after bariatric surgery; however, close clinical symptom monitoring and/or therapeutic drug monitoring is recommended in the immediate postoperative period and at regular follow-up intervals after bariatric surgery to validate drug absorption that may change with small bowel adaption or transit over time.

Nasogastric tube: If unable to swallow capsules, contents of IR capsule(s) may be mixed with water and flushed through a nasogastric tube; clamp nasogastric tube for 30 to 60 minutes after administration (Ref). Note: Dosing for IR capsules is not equivalent to dosing of Envarsus XR. When switching from Envarsus XR to IR capsules for nasogastric administration, consider administering a 20% to 30% higher total daily dose of IR tacrolimus.

Sublingual: If unable to swallow capsules, tacrolimus may be administered sublingually (at a reduced dose) by opening the IR capsules and placing the contents of the capsule(s) under the tongue, allowing contents to completely dissolve. The patient should avoid swallowing for 5 to 15 minutes and avoid oral intake for 15 to 30 minutes; also avoid mechanical suctioning for at least 30 minutes after administration (Ref). Caregivers should don 2 pairs of gloves while handling contents of capsules (Ref). Note: Absorption of a single dose of tacrolimus suspension given by the sublingual route was inadequate (Ref).

Administration: Pediatric

Oral:

Immediate release: Administer with or without food; be consistent with timing and composition of meals if GI intolerance occurs and administration with food becomes necessary (per manufacturer); do not administer with grapefruit juice; do not administer within 2 hours before or after antacids. If dosed twice daily, doses should be 12 hours apart. If the morning and evening doses differ, the larger dose (differences are never >0.5 to 1 mg) should be administered in the morning; however, some data suggests that the larger dose should be given in the evening since tacrolimus exposure may be reduced in the evening due to diurnal variation (Ref). If dosed 3 times daily, separate doses by 8 hours.

Oral granules for suspension: Empty the contents of each packet into a glass cup and mix with 15 to 30 mL of room temperature drinking water. Use whole packet(s) to prepare, adding additional 0.2 mg packets if necessary. The granules will not dissolve completely. Administer immediately after preparation. Dosing syringe or cup should be rinsed with an additional 15 to 30 mL of room temperature water to ensure all granules are taken. Do not use PVC-containing dosing cups or oral syringes to prepare. Do not sprinkle directly on food.

Oral capsules:

Nasogastric tube: If unable to swallow capsules, contents of immediate-release capsule(s) may be mixed with water and flushed through a nasogastric tube; clamp nasogastric tube for 30 to 60 minutes after administration (Ref). Note: If patient receiving an extended-release formulation, convert to an appropriate dose of the immediate release. Dosing for immediate-release capsules is not equivalent to dosing of Envarsus XR. When switching from Envarsus XR to immediate-release capsules for nasogastric administration, consider administering a 20% to 30% higher total daily dose of immediate-release tacrolimus.

Sublingual : Recommendations include opening capsules and applying contents under the tongue. Patient should be advised not to swallow for 5 to 15 minutes and eating or drinking anything for up to 30 minutes (Ref).

Extended release: Administer on an empty stomach at least 1 hour before or 2 hours after a meal. Swallow whole; do not chew, crush, or divide. Take once daily in the morning at a consistent time each day.

Missed doses: Astagraf XL: Children ≥4 years and Adolescents: May be taken up to 14 hours after scheduled time; if >14 hours, resume at next regularly scheduled time; do not double a dose to make up for a missed dose.

Parenteral: Administer by IV infusion as a continuous infusion (preferred) (generally over 24 hours) or may also be administered as an intermittent infusion given over 2 hours (Ref). Must be diluted prior to administration. Do not use PVC tubing when administering diluted solutions; PVC-free administration tubing should be used to minimize the potential for significant drug absorption onto the tubing; adsorption of the drug to PVC tubing may become clinically significant with low concentrations. Do not mix with solutions with a pH ≥9 (eg, acyclovir or ganciclovir) due to chemical degradation of tacrolimus (use different ports in multilumen lines). Do not alter dose with concurrent T-tube clamping. Continue only until oral medication can be tolerated.

Hazardous Drugs Handling Considerations

Hazardous agent (NIOSH 2016 [group 2]).

Use appropriate precautions for receiving, handling, storage, preparation, dispensing, transporting, administration, and disposal. Follow NIOSH and USP 800 recommendations and institution-specific policies/procedures for appropriate containment strategy (NIOSH 2016; USP-NF 2020).

Note: Facilities may perform risk assessment of some hazardous drugs to determine if appropriate for alternative handling and containment strategies (USP-NF 2020). Refer to institution-specific handling policies/procedures.

Medication Guide and/or Vaccine Information Statement (VIS)

An FDA-approved patient medication guide, which is available with the product information and as follows, must be dispensed with this medication:

Astagraf XL: https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/204096s011lbl.pdf#page=33

Envarsus XR: https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/206406s010lbl.pdf#page=29

Use: Labeled Indications

Organ rejection prophylaxis:

Advagraf [Canadian product]: Prevention of organ rejection in allogeneic kidney or liver transplant adult patients in combination with other immunosuppressants.

Astagraf XL: Prevention of organ rejection in adult and pediatric patients (who can swallow intact capsules) kidney transplant recipients in combination with other immunosuppressants.

Envarsus PA [Canadian product]: Prevention of organ rejection in allogeneic kidney or liver transplant adult patients in combination with other immunosuppressants.

Envarsus XR: Prevention of organ rejection in kidney transplant recipients in combination with other immunosuppressants.

Prograf: Prevention of organ rejection in adult and pediatric heart, kidney, liver, or lung transplant recipients in combination with other immunosuppressants.

Note: ER products (Advagraf [Canadian product], Astagraf XL, Envarsus PA [Canadian product], and Envarsus XR) are not interchangeable or substitutable with immediate release tacrolimus. In addition, the once-daily formulations (Advagraf [Canadian product], Astagraf XL, Envarsus PA [Canadian product], and Envarsus XR) are not interchangeable with each other due to significantly different pharmacokinetic properties.

Use: Off-Label: Adult

Graft-versus-host disease (prevention/treatment); Intestinal transplant; Myasthenia gravis, chronic immunosuppressive therapy; Pancreas transplant

Medication Safety Issues
Sound-alike/look-alike issues:

Prograf may be confused with Gengraf, Proscar, PROzac

Tacrolimus may be confused with everolimus, pimecrolimus, sirolimus (conventional), sirolimus (protein bound), tamsulosin, temsirolimus

High alert medication:

This medication is in a class the Institute for Safe Medication Practices (ISMP) includes among its list of drug classes that have a heightened risk of causing significant patient harm when used in error.

Other safety concerns:

The immediate release (Prograf) and extended release (Astagraf XL, Advagraf [Canadian product]) oral formulations are not interchangeable and are not substitutable.

Metabolism/Transport Effects

Substrate of CYP3A4 (major), P-glycoprotein/ABCB1 (major); Note: Assignment of Major/Minor substrate status based on clinically relevant drug interaction potential

Drug Interactions

Note: Interacting drugs may not be individually listed below if they are part of a group interaction (eg, individual drugs within “CYP3A4 Inducers [Strong]” are NOT listed). For a complete list of drug interactions by individual drug name and detailed management recommendations, use the Lexicomp drug interactions program by clicking on the “Launch drug interactions program” link above.

5-Aminosalicylic Acid Derivatives: May enhance the myelosuppressive effect of Myelosuppressive Agents. Risk C: Monitor therapy

Abrocitinib: May enhance the immunosuppressive effect of Immunosuppressants (Therapeutic Immunosuppressant Agents). Risk X: Avoid combination

Adalimumab: May decrease the serum concentration of CYP Substrates (Narrow Therapeutic Index/Sensitive with Inducers). Risk C: Monitor therapy

Adefovir: May enhance the nephrotoxic effect of Tacrolimus (Systemic). Risk C: Monitor therapy

Afatinib: Tacrolimus (Systemic) may increase the serum concentration of Afatinib. Management: Monitor for signs and symptoms of afatinib toxicity when these agents are combined. Consider administering tacrolimus simultaneously with, or after, the dose of afatinib. If the combination is not tolerated, consider reducing the afatinib dose by 10 mg. Risk C: Monitor therapy

Alcohol (Ethyl): May increase the absorption of Tacrolimus (Systemic). More specifically, the initial absorption rate may be increased, as alcohol may speed the release of tacrolimus from extended-release tablets. Management: Advise patients receiving extended-release tacrolimus (Astagraf XL or Envarsus XR brands) not to take the medication with alcoholic beverages. Risk D: Consider therapy modification

Aldesleukin: May increase the serum concentration of CYP Substrates (Narrow Therapeutic Index/Sensitive with Inhibitors). Risk C: Monitor therapy

Aluminum Hydroxide: May decrease the serum concentration of Tacrolimus (Systemic). Particularly, maximum concentration may decrease. Aluminum Hydroxide may increase the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Aminoglycosides: May enhance the nephrotoxic effect of Tacrolimus (Systemic). Risk C: Monitor therapy

Amiodarone: Tacrolimus (Systemic) may enhance the QTc-prolonging effect of Amiodarone. Amiodarone may increase the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Amphotericin B: May enhance the nephrotoxic effect of Tacrolimus (Systemic). Risk C: Monitor therapy

Androgens: Hypertension-Associated Agents may enhance the hypertensive effect of Androgens. Risk C: Monitor therapy

Angiotensin II Receptor Blockers: May enhance the hyperkalemic effect of Tacrolimus (Systemic). Risk C: Monitor therapy

Angiotensin-Converting Enzyme Inhibitors: May enhance the hyperkalemic effect of Tacrolimus (Systemic). Risk C: Monitor therapy

Antidiabetic Agents: Hyperglycemia-Associated Agents may diminish the therapeutic effect of Antidiabetic Agents. Risk C: Monitor therapy

Antithymocyte Globulin (Equine): Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the adverse/toxic effect of Antithymocyte Globulin (Equine). Specifically, these effects may be unmasked if the dose of immunosuppressive therapy is reduced. Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the immunosuppressive effect of Antithymocyte Globulin (Equine). Specifically, infections may occur with greater severity and/or atypical presentations. Risk C: Monitor therapy

Asciminib: May increase the serum concentration of P-glycoprotein/ABCB1 Substrates (Narrow Therapeutic Index/Sensitive with Inhibitors). Risk C: Monitor therapy

Azithromycin (Systemic): May increase the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Baricitinib: Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the immunosuppressive effect of Baricitinib. Risk X: Avoid combination

BCG (Intravesical): Myelosuppressive Agents may diminish the therapeutic effect of BCG (Intravesical). Risk X: Avoid combination

BCG Products: Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the adverse/toxic effect of BCG Products. Specifically, the risk of vaccine-associated infection may be increased. Immunosuppressants (Therapeutic Immunosuppressant Agents) may diminish the therapeutic effect of BCG Products. Risk X: Avoid combination

Betel Nut: May decrease the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Bimekizumab: May decrease the serum concentration of CYP Substrates (Narrow Therapeutic Index/Sensitive with Inducers). Risk C: Monitor therapy

Brincidofovir: Immunosuppressants (Therapeutic Immunosuppressant Agents) may diminish the therapeutic effect of Brincidofovir. Risk C: Monitor therapy

Brivudine: May enhance the adverse/toxic effect of Immunosuppressants (Therapeutic Immunosuppressant Agents). Risk X: Avoid combination

Calcium Channel Blockers (Dihydropyridine): May increase the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Cannabidiol: May increase the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Cannabis: May increase the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Chikungunya Vaccine (Live): Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the adverse/toxic effect of Chikungunya Vaccine (Live). Specifically, the risk of vaccine-associated infection may be increased. Immunosuppressants (Therapeutic Immunosuppressant Agents) may diminish the therapeutic effect of Chikungunya Vaccine (Live). Risk X: Avoid combination

Chloramphenicol (Ophthalmic): May enhance the adverse/toxic effect of Myelosuppressive Agents. Risk C: Monitor therapy

Chloramphenicol (Systemic): May increase the serum concentration of Tacrolimus (Systemic). Management: Reduce the tacrolimus dose and monitor tacrolimus whole blood concentrations frequently, beginning within 1 to 3 days of chloramphenicol initiation. Further tacrolimus dose adjustments should be guided by continued monitoring of tacrolimus levels. Risk D: Consider therapy modification

Cidofovir: May enhance the nephrotoxic effect of Tacrolimus (Systemic). Risk C: Monitor therapy

Cinacalcet: May decrease the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

CISplatin: May enhance the nephrotoxic effect of Tacrolimus (Systemic). Risk C: Monitor therapy

Cladribine: May enhance the myelosuppressive effect of Myelosuppressive Agents. Risk X: Avoid combination

Cladribine: Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the immunosuppressive effect of Cladribine. Risk X: Avoid combination

Clofazimine: May increase the serum concentration of CYP3A4 Substrates (High risk with Inhibitors). Risk C: Monitor therapy

Clotrimazole (Oral): May increase the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Clotrimazole (Topical): May increase the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

CloZAPine: Myelosuppressive Agents may enhance the adverse/toxic effect of CloZAPine. Specifically, the risk for neutropenia may be increased. Risk C: Monitor therapy

Coccidioides immitis Skin Test: Immunosuppressants (Therapeutic Immunosuppressant Agents) may diminish the diagnostic effect of Coccidioides immitis Skin Test. Management: Consider discontinuing therapeutic immunosuppressants several weeks prior to coccidioides immitis skin antigen testing to increase the likelihood of accurate diagnostic results. Risk D: Consider therapy modification

Colchicine: Tacrolimus (Systemic) may increase the serum concentration of Colchicine. Risk C: Monitor therapy

Corticosteroids (Systemic): May decrease the serum concentration of Tacrolimus (Systemic). Conversely, when discontinuing corticosteroid therapy, tacrolimus concentrations may increase. Risk C: Monitor therapy

COVID-19 Vaccines: Calcineurin Inhibitors (Systemic) may diminish the therapeutic effect of COVID-19 Vaccines. Management: Rheumatology guidelines recommend holding calcineurin inhibitors for 1 to 2 weeks after each vaccine dose as permitted by the underlying disease. This is specific to the use of calcineurin inhibitors for rheumatologic or musculoskeletal disease. Risk D: Consider therapy modification

CycloSPORINE (Systemic): Tacrolimus (Systemic) may enhance the nephrotoxic effect of CycloSPORINE (Systemic). CycloSPORINE (Systemic) may enhance the nephrotoxic effect of Tacrolimus (Systemic). Tacrolimus (Systemic) may increase the serum concentration of CycloSPORINE (Systemic). CycloSPORINE (Systemic) may increase the serum concentration of Tacrolimus (Systemic). Risk X: Avoid combination

CYP3A4 Inducers (Moderate): May decrease the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

CYP3A4 Inducers (Strong): May decrease the serum concentration of Tacrolimus (Systemic). Management: Monitor for decreased tacrolimus concentrations and effects when combined with strong CYP3A4 inducers. Tacrolimus dose increases will likely be needed during concomitant use. Risk D: Consider therapy modification

CYP3A4 Inducers (Weak): May decrease the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

CYP3A4 Inhibitors (Moderate): May increase the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

CYP3A4 Inhibitors (Strong): May increase the serum concentration of Tacrolimus (Systemic). Management: Reduce tacrolimus dose to one-third of the original dose if starting posaconazole or voriconazole. Coadministration with nelfinavir is not generally recommended. Tacrolimus dose reductions or prolongation of dosing interval will likely be required. Risk D: Consider therapy modification

CYP3A4 Inhibitors (Weak): May increase the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Deferasirox: May decrease the serum concentration of CYP3A4 Substrates (Narrow Therapeutic Index/Sensitive with Inducers). Risk C: Monitor therapy

Deferiprone: Myelosuppressive Agents may enhance the neutropenic effect of Deferiprone. Management: Avoid the concomitant use of deferiprone and myelosuppressive agents whenever possible. If this combination cannot be avoided, monitor the absolute neutrophil count more closely. Risk D: Consider therapy modification

Dengue Tetravalent Vaccine (Live): Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the adverse/toxic effect of Dengue Tetravalent Vaccine (Live). Specifically, the risk of vaccine-associated infection may be increased. Immunosuppressants (Therapeutic Immunosuppressant Agents) may diminish the therapeutic effect of Dengue Tetravalent Vaccine (Live). Risk X: Avoid combination

Denosumab: Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the immunosuppressive effect of Denosumab. Management: Consider the risk of serious infections versus the potential benefits of coadministration of denosumab and immunosuppressants. If combined, monitor for signs/symptoms of serious infections. Risk D: Consider therapy modification

Deucravacitinib: May enhance the immunosuppressive effect of Immunosuppressants (Therapeutic Immunosuppressant Agents). Risk X: Avoid combination

Dipyrone: May enhance the adverse/toxic effect of Myelosuppressive Agents. Specifically, the risk for agranulocytosis and pancytopenia may be increased Risk X: Avoid combination

Direct Acting Antiviral Agents (HCV): May decrease the serum concentration of Tacrolimus (Systemic). Direct Acting Antiviral Agents (HCV) may increase the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Dronedarone: Tacrolimus (Systemic) may enhance the QTc-prolonging effect of Dronedarone. Dronedarone may increase the serum concentration of Tacrolimus (Systemic). Management: Monitor for increased serum tacrolimus concentrations, tacrolimus toxicity, and QTc interval prolongation if combined with dronedarone. Tacrolimus dose adjustments may be needed. Risk D: Consider therapy modification

Efonidipine: May increase the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Elacestrant: May increase the serum concentration of P-glycoprotein/ABCB1 Substrates (Narrow Therapeutic Index/Sensitive with Inhibitors). Risk C: Monitor therapy

Elranatamab: May increase the serum concentration of CYP Substrates (Narrow Therapeutic Index/Sensitive with Inhibitors). Risk C: Monitor therapy

Epcoritamab: May increase the serum concentration of CYP Substrates (Narrow Therapeutic Index/Sensitive with Inhibitors). Risk C: Monitor therapy

Erdafitinib: May decrease the serum concentration of CYP3A4 Substrates (Narrow Therapeutic Index/Sensitive with Inducers). Risk X: Avoid combination

Erdafitinib: May increase the serum concentration of CYP3A4 Substrates (Narrow Therapeutic Index/Sensitive with Inhibitors). Risk X: Avoid combination

Ertapenem: May increase the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Erythromycin (Systemic): May increase the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Estrogen Derivatives: May increase the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Etrasimod: May enhance the immunosuppressive effect of Immunosuppressants (Therapeutic Immunosuppressant Agents). Risk X: Avoid combination

Everolimus: May decrease the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Fenofibrate and Derivatives: Tacrolimus (Systemic) may enhance the nephrotoxic effect of Fenofibrate and Derivatives. Risk C: Monitor therapy

Fexinidazole: May increase the serum concentration of CYP3A4 Substrates (High risk with Inhibitors). Risk X: Avoid combination

Fexinidazole: Myelosuppressive Agents may enhance the myelosuppressive effect of Fexinidazole. Risk X: Avoid combination

Filgotinib: May enhance the immunosuppressive effect of Immunosuppressants (Therapeutic Immunosuppressant Agents). Risk X: Avoid combination

Fluconazole: May increase the serum concentration of Tacrolimus (Systemic). Management: Monitor tacrolimus concentrations closely and adjust oral tacrolimus dose as necessary when concomitantly administered with fluconazole. Reduced doses of tacrolimus will likely be required. Risk D: Consider therapy modification

Foscarnet: May enhance the nephrotoxic effect of Tacrolimus (Systemic). Risk X: Avoid combination

Fusidic Acid (Systemic): May increase the serum concentration of CYP3A4 Substrates (High risk with Inhibitors). Risk X: Avoid combination

Futibatinib: May increase the serum concentration of P-glycoprotein/ABCB1 Substrates (Narrow Therapeutic Index/Sensitive with Inhibitors). Risk C: Monitor therapy

Ganciclovir (Systemic): May enhance the nephrotoxic effect of Tacrolimus (Systemic). Risk C: Monitor therapy

Gilteritinib: May increase the serum concentration of P-glycoprotein/ABCB1 Substrates (Narrow Therapeutic Index/Sensitive with Inhibitors). Risk C: Monitor therapy

Glofitamab: May increase the serum concentration of CYP Substrates (Narrow Therapeutic Index/Sensitive with Inhibitors). Risk C: Monitor therapy

Grapefruit Juice: May decrease the metabolism of Tacrolimus (Systemic). Risk X: Avoid combination

Haloperidol: QT-prolonging Agents (Indeterminate Risk - Caution) may enhance the QTc-prolonging effect of Haloperidol. Risk C: Monitor therapy

Inebilizumab: Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the immunosuppressive effect of Inebilizumab. Risk C: Monitor therapy

Influenza Virus Vaccines: Immunosuppressants (Therapeutic Immunosuppressant Agents) may diminish the therapeutic effect of Influenza Virus Vaccines. Management: Administer influenza vaccines at least 2 weeks prior to initiating immunosuppressants if possible. If vaccination occurs less than 2 weeks prior to or during therapy, revaccinate 2 to 3 months after therapy discontinued if immune competence restored. Risk D: Consider therapy modification

Inhibitors of the Proton Pump (PPIs and PCABs): May increase the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Interleukin-6 (IL-6) Inhibiting Therapies: May decrease the serum concentration of CYP3A4 Substrates (Narrow Therapeutic Index/Sensitive with Inducers). Risk C: Monitor therapy

Interleukin-6 (IL-6) Inhibiting Therapies: May decrease the serum concentration of CYP Substrates (Narrow Therapeutic Index/Sensitive with Inducers). Risk C: Monitor therapy

Itraconazole: May increase the serum concentration of Tacrolimus (Systemic). Management: Monitor tacrolimus concentrations closely during therapy with itraconazole; tacrolimus dose reductions will likely be required. The magnitude of this interaction may be greater in older patients or those with one or more CYP3A5*3 alleles. Risk D: Consider therapy modification

Ivosidenib: May decrease the serum concentration of CYP3A4 Substrates (Narrow Therapeutic Index/Sensitive with Inducers). Management: Consider alternatives to this combination when possible. If combined, monitor for decreased effectiveness of these CYP3A4 substrates if combined with ivosidenib. Risk D: Consider therapy modification

Ketoconazole (Systemic): May increase the serum concentration of Tacrolimus (Systemic). Management: Tacrolimus dose adjustment may be required; empiric dose reductions of 50% have been recommended. Monitor tacrolimus concentrations and clinical response closely. Risk D: Consider therapy modification

Lasmiditan: May increase the serum concentration of P-glycoprotein/ABCB1 Substrates (Narrow Therapeutic Index/Sensitive with Inhibitors). Risk X: Avoid combination

Leflunomide: Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the immunosuppressive effect of Leflunomide. Management: Increase the frequency of chronic monitoring of platelet, white blood cell count, and hemoglobin or hematocrit to monthly, instead of every 6 to 8 weeks, if leflunomide is coadministered with immunosuppressive agents. Risk D: Consider therapy modification

LevoFLOXacin (Systemic): May enhance the QTc-prolonging effect of Tacrolimus (Systemic). LevoFLOXacin (Systemic) may increase the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Magnesium Hydroxide: May increase the serum concentration of Tacrolimus (Systemic). Magnesium Hydroxide may decrease the serum concentration of Tacrolimus (Systemic). Specifically, maximum concentration may decrease. Risk C: Monitor therapy

Maribavir: May increase the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Mavacamten: May decrease the serum concentration of CYP3A4 Substrates (Narrow Therapeutic Index/Sensitive with Inducers). Risk C: Monitor therapy

Metoclopramide: May increase the serum concentration of Tacrolimus (Systemic). Specifically, treatment of gastroparesis may increase tacrolimus concentrations. Risk C: Monitor therapy

Mifamurtide: Tacrolimus (Systemic) may diminish the therapeutic effect of Mifamurtide. Risk X: Avoid combination

Mitapivat: May increase the serum concentration of P-glycoprotein/ABCB1 Substrates (Narrow Therapeutic Index/Sensitive with Inhibitors). Risk C: Monitor therapy

Mosunetuzumab: May increase the serum concentration of CYP Substrates (Narrow Therapeutic Index/Sensitive with Inhibitors). Risk C: Monitor therapy

Mumps- Rubella- or Varicella-Containing Live Vaccines: Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the adverse/toxic effect of Mumps- Rubella- or Varicella-Containing Live Vaccines. Specifically, the risk of vaccine-associated infection may be increased. Immunosuppressants (Therapeutic Immunosuppressant Agents) may diminish the therapeutic effect of Mumps- Rubella- or Varicella-Containing Live Vaccines. Risk X: Avoid combination

Nadofaragene Firadenovec: Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the adverse/toxic effect of Nadofaragene Firadenovec. Specifically, the risk of disseminated adenovirus infection may be increased. Risk X: Avoid combination

Natalizumab: Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the immunosuppressive effect of Natalizumab. Risk X: Avoid combination

Nelfinavir: May increase the serum concentration of Tacrolimus (Systemic). Management: Avoid this combination unless benefits outweigh risks. If used, tacrolimus dose reductions will likely be required; monitor response and tacrolimus concentrations closely, beginning within 1 to 3 days of coadministration. Risk D: Consider therapy modification

Nirmatrelvir and Ritonavir: May increase the serum concentration of Tacrolimus (Systemic). Management: Consider avoiding this combination, if possible, through use of alternative anti-COVID-19 therapy. If combined, hold tacrolimus during nirmatrelvir/ritonavir treatment and for at least 2 to 3 days after completion. Monitor tacrolimus levels closely. Risk D: Consider therapy modification

Nirogacestat: May increase the serum concentration of CYP3A4 Substrates (Narrow Therapeutic Index/Sensitive with Inhibitors). Risk X: Avoid combination

Nonsteroidal Anti-Inflammatory Agents: May enhance the nephrotoxic effect of Tacrolimus (Systemic). Risk C: Monitor therapy

Nonsteroidal Anti-Inflammatory Agents (Topical): May enhance the nephrotoxic effect of Tacrolimus (Systemic). Risk C: Monitor therapy

Ocrelizumab: Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the immunosuppressive effect of Ocrelizumab. Risk C: Monitor therapy

Ofatumumab: Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the immunosuppressive effect of Ofatumumab. Risk C: Monitor therapy

Olaparib: Myelosuppressive Agents may enhance the myelosuppressive effect of Olaparib. Risk C: Monitor therapy

Olutasidenib: May decrease the serum concentration of CYP3A4 Substrates (Narrow Therapeutic Index/Sensitive with Inducers). Management: Avoid use of olutasidenib with sensitive or narrow therapeutic index CYP3A4 substrates when possible. If concurrent use with olutasidenib is unavoidable, monitor closely for evidence of decreased concentrations of the CYP3A4 substrates. Risk D: Consider therapy modification

Ombitasvir, Paritaprevir, and Ritonavir: May increase the serum concentration of Tacrolimus (Systemic). Risk X: Avoid combination

Ombitasvir, Paritaprevir, Ritonavir, and Dasabuvir: May increase the serum concentration of Tacrolimus (Systemic). Risk X: Avoid combination

Pacritinib: May increase the serum concentration of CYP3A4 Substrates (Narrow Therapeutic Index/Sensitive with Inhibitors). Risk X: Avoid combination

Pacritinib: May increase the serum concentration of P-glycoprotein/ABCB1 Substrates (Narrow Therapeutic Index/Sensitive with Inhibitors). Risk X: Avoid combination

P-glycoprotein/ABCB1 Inhibitors: May increase the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Pidotimod: Immunosuppressants (Therapeutic Immunosuppressant Agents) may diminish the therapeutic effect of Pidotimod. Risk C: Monitor therapy

Pimecrolimus: Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the immunosuppressive effect of Pimecrolimus. Risk X: Avoid combination

Pitolisant: May decrease the serum concentration of CYP3A4 Substrates (Narrow Therapeutic Index/Sensitive with Inducers). Risk C: Monitor therapy

Pneumococcal Vaccines: Immunosuppressants (Therapeutic Immunosuppressant Agents) may diminish the therapeutic effect of Pneumococcal Vaccines. Risk C: Monitor therapy

Poliovirus Vaccine (Live/Trivalent/Oral): Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the adverse/toxic effect of Poliovirus Vaccine (Live/Trivalent/Oral). Specifically, the risk of vaccine-associated infection may be increased. Immunosuppressants (Therapeutic Immunosuppressant Agents) may diminish the therapeutic effect of Poliovirus Vaccine (Live/Trivalent/Oral). Risk X: Avoid combination

Polymethylmethacrylate: Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the potential for allergic or hypersensitivity reactions to Polymethylmethacrylate. Management: Use caution when considering use of bovine collagen-containing implants such as the polymethylmethacrylate-based Bellafill brand implant in patients who are receiving immunosuppressants. Consider use of additional skin tests prior to administration. Risk D: Consider therapy modification

Posaconazole: May increase the serum concentration of Tacrolimus (Systemic). Management: Reduce tacrolimus dose to approximately one-third of original dose when starting posaconazole. Tacrolimus blood concentrations should be monitored closely beginning within 1 to 3 days of coadministration. Risk D: Consider therapy modification

Potassium-Sparing Diuretics: May enhance the hyperkalemic effect of Tacrolimus (Systemic). Risk C: Monitor therapy

Pretomanid: May increase the serum concentration of P-glycoprotein/ABCB1 Substrates (Narrow Therapeutic Index/Sensitive with Inhibitors). Risk C: Monitor therapy

Promazine: May enhance the myelosuppressive effect of Myelosuppressive Agents. Risk C: Monitor therapy

Protease Inhibitors: May enhance the nephrotoxic effect of Tacrolimus (Systemic). Protease Inhibitors may decrease the metabolism of Tacrolimus (Systemic). Management: Consider reducing the tacrolimus dose to 1 mg once or twice per week if coadministered with protease inhibitors that are strong inhibitors of CYP3A4. Monitor response, plasma concentrations (as appropriate), and for signs of toxicity. Risk D: Consider therapy modification

QT-prolonging Agents (Highest Risk): QT-prolonging Agents (Indeterminate Risk - Caution) may enhance the QTc-prolonging effect of QT-prolonging Agents (Highest Risk). Management: Monitor for QTc interval prolongation and ventricular arrhythmias when these agents are combined. Patients with additional risk factors for QTc prolongation may be at even higher risk. Risk C: Monitor therapy

Rabies Vaccine: Immunosuppressants (Therapeutic Immunosuppressant Agents) may diminish the therapeutic effect of Rabies Vaccine. Management: Complete rabies vaccination at least 2 weeks before initiation of immunosuppressant therapy if possible. If combined, check for rabies antibody titers, and if vaccination is for post exposure prophylaxis, administer a 5th dose of the vaccine. Risk D: Consider therapy modification

Remdesivir: May increase the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Ritlecitinib: Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the immunosuppressive effect of Ritlecitinib. Risk X: Avoid combination

Ritonavir: May increase the serum concentration of Tacrolimus (Systemic). Management: Tacrolimus dose reductions may be needed with concurrent ritonavir. Monitor tacrolimus concentrations closely to determine dose; doses of tacrolimus 0.5 mg to 1 mg every week may be adequate. Risk D: Consider therapy modification

Ropeginterferon Alfa-2b: Myelosuppressive Agents may enhance the myelosuppressive effect of Ropeginterferon Alfa-2b. Management: Avoid coadministration of ropeginterferon alfa-2b and other myelosuppressive agents. If this combination cannot be avoided, monitor patients for excessive myelosuppressive effects. Risk D: Consider therapy modification

Ruxolitinib (Topical): Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the immunosuppressive effect of Ruxolitinib (Topical). Risk X: Avoid combination

Saquinavir: May increase the serum concentration of Tacrolimus (Systemic). Risk X: Avoid combination

Schisandra: May increase the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Sevelamer: May decrease the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Simeprevir: May decrease the serum concentration of Tacrolimus (Systemic). Tacrolimus (Systemic) may increase the serum concentration of Simeprevir. Risk C: Monitor therapy

Sipuleucel-T: Immunosuppressants (Therapeutic Immunosuppressant Agents) may diminish the therapeutic effect of Sipuleucel-T. Management: Consider reducing the dose or discontinuing the use of immunosuppressants prior to initiating sipuleucel-T therapy. Risk D: Consider therapy modification

Sirolimus Products: Tacrolimus (Systemic) may enhance the adverse/toxic effect of Sirolimus Products. Sirolimus Products may enhance the adverse/toxic effect of Tacrolimus (Systemic). Sirolimus Products may decrease the serum concentration of Tacrolimus (Systemic). Risk X: Avoid combination

Sodium Zirconium Cyclosilicate: May decrease the serum concentration of Tacrolimus (Systemic). Management: Separate the administration of sodium zirconium cyclosilicate and tacrolimus by at least 2 hours. Risk D: Consider therapy modification

Solriamfetol: May enhance the hypertensive effect of Hypertension-Associated Agents. Risk C: Monitor therapy

Sotagliflozin: May decrease the serum concentration of Tacrolimus (Systemic). Sotagliflozin may increase the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Sparsentan: May increase the serum concentration of P-glycoprotein/ABCB1 Substrates (Narrow Therapeutic Index/Sensitive with Inhibitors). Risk X: Avoid combination

Sphingosine 1-Phosphate (S1P) Receptor Modulator: May enhance the immunosuppressive effect of Immunosuppressants (Therapeutic Immunosuppressant Agents). Risk C: Monitor therapy

Spironolactone: May increase the serum concentration of CYP3A4 Substrates (Narrow Therapeutic Index/Sensitive with Inhibitors). Risk C: Monitor therapy

St John's Wort: May decrease the serum concentration of Tacrolimus (Systemic). Management: Consider avoiding this combination. If coadministered, monitor for reduced serum concentrations of tacrolimus when St Johns Wort is initiated/dose increased, and increased serum concentrations if St Johns Wort is discontinued/dose decreased. Risk D: Consider therapy modification

Tacrolimus (Topical): Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the immunosuppressive effect of Tacrolimus (Topical). Risk X: Avoid combination

Talimogene Laherparepvec: Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the adverse/toxic effect of Talimogene Laherparepvec. Specifically, the risk of infection from the live, attenuated herpes simplex virus contained in talimogene laherparepvec may be increased. Risk X: Avoid combination

Talquetamab: May increase the serum concentration of CYP Substrates (Narrow Therapeutic Index/Sensitive with Inhibitors). Risk C: Monitor therapy

Taurursodiol: May increase the serum concentration of P-glycoprotein/ABCB1 Substrates (Narrow Therapeutic Index/Sensitive with Inhibitors). Risk X: Avoid combination

Taurursodiol: May decrease the serum concentration of CYP3A4 Substrates (Narrow Therapeutic Index/Sensitive with Inducers). Risk X: Avoid combination

Teclistamab: May increase the serum concentration of CYP Substrates (Narrow Therapeutic Index/Sensitive with Inhibitors). Risk C: Monitor therapy

Temsirolimus: Tacrolimus (Systemic) may enhance the adverse/toxic effect of Temsirolimus. Temsirolimus may enhance the adverse/toxic effect of Tacrolimus (Systemic). Temsirolimus may decrease the serum concentration of Tacrolimus (Systemic). Temsirolimus may increase the serum concentration of Tacrolimus (Systemic). Risk X: Avoid combination

Tenofovir Products: May enhance the nephrotoxic effect of Tacrolimus (Systemic). Risk C: Monitor therapy

Tertomotide: Immunosuppressants (Therapeutic Immunosuppressant Agents) may diminish the therapeutic effect of Tertomotide. Risk X: Avoid combination

Tigecycline: May increase the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Tofacitinib: Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the immunosuppressive effect of Tofacitinib. Management: Coadministration of tofacitinib with potent immunosuppressants is not recommended. Use with non-biologic disease-modifying antirheumatic drugs (DMARDs) was permitted in psoriatic arthritis clinical trials. Risk X: Avoid combination

Tofisopam: May increase the serum concentration of Tacrolimus (Systemic). Risk C: Monitor therapy

Treosulfan: May increase the serum concentration of CYP3A4 Substrates (Narrow Therapeutic Index/Sensitive with Inhibitors). Risk X: Avoid combination

Trofinetide: May increase the serum concentration of CYP3A4 Substrates (Narrow Therapeutic Index/Sensitive with Inhibitors). Risk C: Monitor therapy

Typhoid Vaccine: Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the adverse/toxic effect of Typhoid Vaccine. Specifically, the risk of vaccine-associated infection may be increased. Immunosuppressants (Therapeutic Immunosuppressant Agents) may diminish the therapeutic effect of Typhoid Vaccine. Risk X: Avoid combination

Ublituximab: Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the immunosuppressive effect of Ublituximab. Risk C: Monitor therapy

Upadacitinib: Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the immunosuppressive effect of Upadacitinib. Risk X: Avoid combination

Ustekinumab: May decrease the serum concentration of CYP Substrates (Narrow Therapeutic Index/Sensitive with Inducers). Risk C: Monitor therapy

Vaccines (Inactivated/Non-Replicating): Immunosuppressants (Therapeutic Immunosuppressant Agents) may diminish the therapeutic effect of Vaccines (Inactivated/Non-Replicating). Management: Give inactivated vaccines at least 2 weeks prior to initiation of immunosuppressants when possible. Patients vaccinated less than 14 days before initiating or during therapy should be revaccinated at least 2 to 3 months after therapy is complete. Risk D: Consider therapy modification

Vaccines (Live): Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the adverse/toxic effect of Vaccines (Live). Specifically, the risk of vaccine-associated infection may be increased. Immunosuppressants (Therapeutic Immunosuppressant Agents) may diminish the therapeutic effect of Vaccines (Live). Risk X: Avoid combination

Voriconazole: May increase the serum concentration of Tacrolimus (Systemic). Management: Reduce tacrolimus dose to approximately one-third of the original dose when starting concurrent voriconazole. Tacrolimus whole blood trough concentrations should be monitored closely beginning within 1 to 3 days of concomitant use. Risk D: Consider therapy modification

Yellow Fever Vaccine: Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the adverse/toxic effect of Yellow Fever Vaccine. Specifically, the risk of vaccine-associated infection may be increased. Immunosuppressants (Therapeutic Immunosuppressant Agents) may diminish the therapeutic effect of Yellow Fever Vaccine. Risk X: Avoid combination

Food Interactions

Ethanol: Alcohol may increase the rate of release of extended-release tacrolimus and adversely affect tacrolimus safety and/or efficacy. Management: Avoid alcohol.

Food: Food decreases rate and extent of absorption. High-fat meals have most pronounced effect (37% and 25% decrease in AUC, respectively, and 77% and 25% decrease in Cmax, respectively, for immediately release and extended release formulations). Grapefruit juice, a CYP3A4 inhibitor, may increase serum level and/or toxicity of tacrolimus. Management: Administer with or without food (immediate release), but be consistent. Administer extended release on an empty stomach. Avoid concurrent use of grapefruit juice.

Reproductive Considerations

Family planning and contraceptive options for patients who can become pregnant and patients with partners who can become pregnant should be evaluated prior to starting treatment.

Tacrolimus is an acceptable immunosuppressant for use in kidney, heart, and liver transplant recipients planning a pregnancy. Conception may be considered for patients who can become pregnant who are on a stable/low maintenance dose for ≥1 year following transplant (AASLD [Lucey 2013]; EBPG 2002; ISHLT [Velleca 2023]; López 2014). Tacrolimus has also been used as an immunosuppressant in patients undergoing uterine transplant (Jones 2019).

Based on limited data, tacrolimus may be used in patients with rheumatic and musculoskeletal diseases who are planning to become pregnant and are not able to use alternative therapies; however, BP monitoring is recommended. Conception should be planned during a period of quiescent/low disease activity (ACR [Sammaritano 2020]).

Based on limited data, tacrolimus may be used in patients with rheumatic and musculoskeletal diseases who are planning to father a child (ACR [Sammaritano 2020]).

Pregnancy Considerations

Tacrolimus crosses the human placenta and is measurable in the cord blood, amniotic fluid, and newborn serum. Tacrolimus also accumulates in the placenta in concentrations that may be higher than the maternal serum (Freriksen 2018; Jain 1997). Infants with lower birth weights have been found to have higher tacrolimus concentrations, possibly due to slower metabolism (Bramham 2013).

Miscarriage, preterm delivery, low birth weight, birth defects (including cardiac, craniofacial, neurologic, renal/urogenital, and skeletal abnormalities), renal dysfunction, transient neonatal hyperkalemia, and fetal distress have been reported following in utero exposure to tacrolimus in infants of organ transplant recipients; however, pregnant patients were also taking a concomitant medication known to cause adverse pregnancy outcomes. Tacrolimus may exacerbate hypertension and hyperglycemia in pregnant patients with preexisting disease. Adverse pregnancy outcomes may also be associated with an organ transplant, including preterm delivery and low birth weight in the infant and hypertension and preeclampsia in the mother. Cholestasis of pregnancy may be increased following liver transplant.

Due to pregnancy-induced physiologic changes, some pharmacokinetic properties of tacrolimus are altered. Increased monitoring of blood levels in the pregnant patient is recommended, and dosage adjustments may be required during pregnancy and immediately postpartum (AASLD [Lucey 2013]; EBPG 2002; ISHLT [Velleca 2023]; López 2014). Whole blood concentrations of tacrolimus decrease as pregnancy progresses; however, unbound tacrolimus concentrations increase. Measuring unbound concentrations may be preferred, especially in patients with anemia or hypoalbuminemia. If unbound concentration measurement is not available, interpretation of whole blood concentrations should account for RBC count and serum albumin concentration (Hebert 2013; Zheng 2012).

Tacrolimus monotherapy is the preferred immunosuppressant in patients who become pregnant following a liver transplant (AASLD [Lucey 2013]). Tacrolimus may also be used in pregnant patients who have had a kidney (EBPG 2002; López 2014), heart (ISHLT [Velleca 2023]), or uterine (Jones 2019; Perni 2022; Richards 2021; Wilson 2023) transplant.

Based on limited data, tacrolimus may be used in pregnant patients with rheumatic and musculoskeletal diseases who are not able to use alternative therapies; however, close monitoring of blood pressure is recommended (ACR [Sammaritano 2020]). Tacrolimus is not one of the recommended agents for the treatment of myasthenia gravis during pregnancy (Sanders 2016).

The Transplant Pregnancy Registry International (TPRI) is a registry that follows pregnancies that occur in maternal transplant recipients or those fathered by male transplant recipients. The TPRI encourages reporting of pregnancies following solid organ transplant by contacting them at 1-877-955-6877 or https://www.transplantpregnancyregistry.org.

Breastfeeding Considerations

Tacrolimus is present in breast milk.

Authors of several small studies and case reports have calculated the relative exposure of tacrolimus to the breastfeeding infant to be in acceptable concentrations (≤0.5% of the weight-adjusted maternal dose) (Akamine 2021; Bramham 2013; French 2003; Gardiner 2006; Hiramatsu 2018; Kociszewska-Najman 2018; Richards 2021; Zheng 2013).

A study describes the course of 15 infants (12 receiving exclusively human milk) exposed to tacrolimus throughout pregnancy and after birth. Serial blood levels evaluated in 13 of the infants over the first 72 hours of life showed a decrease of tacrolimus concentrations over time regardless of breastfeeding status. Although present in cord blood, tacrolimus was undetectable in infant blood between 11 to 22 days after birth (n=8). Breast milk concentrations were variable but correlated with maternal blood levels (Bramham 2013). A second study evaluated tacrolimus concentrations in breast milk and infant serum between 1 and 3 months' postpartum. Although tacrolimus was measurable in breast milk, infant serum concentrations were below the limit of quantification in the 13 infants tested (Hiramatsu 2018). Information is also available from six breastfed children whose mothers were taking tacrolimus. Children were breastfed for 1.5 to 6 months (four exclusively breastfed) and monitored for 2 to 30 months. No adverse outcomes related to tacrolimus exposure were observed (Gouraud 2012).

According to the manufacturer, the decision to breastfeed during therapy should consider the risk of infant exposure, the benefits of breastfeeding to the infant, and the benefits of treatment to the breastfeeding patient. Recommendations for breastfeeding in patients taking tacrolimus following a kidney transplant differ; generally breastfeeding may be considered with maternal use of maintenance doses (Constantinescu 2014; EBPG 2002; López 2014). Tacrolimus is acceptable for use in lactating patients following a heart transplant (ISHLT [Velleca 2023]) and may be continued or initiated in patients with rheumatic and musculoskeletal diseases who are breastfeeding (ACR [Sammaritano 2020]).

Dietary Considerations

Avoid grapefruit and grapefruit juice. Avoid alcohol.

Monitoring Parameters

Renal function, hepatic function, serum electrolytes (calcium, magnesium, phosphorus, potassium; periodically in patients with heart failure, bradyarrhythmias, or concomitant medications known to prolong the QT interval), blood glucose (frequently). Monitor BP (3 times/week for first few weeks, then gradually decrease frequency as patient stabilizes). Patients receiving IV tacrolimus should be monitored for signs/symptoms of hypersensitivity/anaphylactic reactions for at least the first 30 minutes following the start of the infusion, and frequently thereafter. Monitor for QT prolongation; consider echocardiographic evaluation in patients who develop renal failure, electrolyte abnormalities, with heart failure, bradyarrhythmias, or concomitant medications known to prolong the QT interval or clinical manifestations of ventricular dysfunction. Monitor for signs/symptoms of infection, neurotoxicity, and secondary malignancy.

Closely monitor tacrolimus levels to assist in dose adjustment, monitor compliance, prevent organ rejection, and reduce drug-related toxicity. Whole blood concentrations should be used for monitoring (trough for oral therapy drawn typically within 30 minutes prior to the next dose); frequency varies depending on transplant type, time since transplantation, and clinical situation. Tacrolimus serum levels may be falsely elevated in infected liver transplant patients due to interference from beta-galactosidase antibodies, or if tacrolimus levels were drawn from IV lines that tacrolimus was infused through (Grouzmann 2008). Timing for obtaining tacrolimus trough levels depend on the dosage form as follows:

Oral:

Immediate release: ~12 hours after previous dose or immediately prior to next dose (Jusko 1995).

Extended release: Immediately prior to next dose; steady state takes 7 days to achieve. Obtain 2 trough levels on separate days during the first week after initiation.

IV: For intermittent dosing, ~12 hours after previous dose or immediately prior to next dose; levels drawn during continuous infusion will not represent trough concentrations (Jusko 1995). Do not draw levels from IV lines where tacrolimus has been infused (Grouzmann 2008).

Reference Range

Note: Blood concentrations may differ depending on the laboratory assay used; refer also to institutional protocols when interpreting tacrolimus concentrations.

Heart transplant: Typical whole blood trough concentrations (in combination with azathioprine or mycophenolate), goal levels may vary in patients on mTOR inhibitors (ISHLT [Velleca 2023]):

Days 0 to 60: 10 to 15 ng/mL.

Months 3 to 6: 8 to 12 ng/mL.

>6 months: 5 to 10 ng/mL; when combined with mTOR inhibitors 3 to 8 ng/mL (ISHLT [Velleca 2023]).

Kidney transplant: Whole blood trough concentrations:

Immediate release:

In combination with azathioprine (if used):

Months 1 to 3: 7 to 20 ng/mL.

Months 4 to 12: 5 to 15 ng/mL.

In combination with mycophenolate mofetil/IL-2 receptor antagonist (eg, basiliximab): 4 to 11 ng/mL.

In combination with mTOR inhibitor (everolimus): 4 to 8 ng/mL for the first 2 months post-transplant followed by 3 to 5 ng/mL thereafter (Sommerer 2016).

Extended release (Astagraf XL):

Adult:

With basiliximab induction:

Month 1: 7 to 15 ng/mL.

Months 2 to 6: 5 to 15 ng/mL.

>6 months: 5 to 10 ng/mL.

Without basiliximab induction:

Month 1: 10 to 15 ng/mL.

Months 2 to 6: 5 to 15 ng/mL.

>6 months: 5 to 10 ng/mL.

Pediatric:

With basiliximab induction:

Month 1: 10 to 20 ng/mL.

> Month 1: 5 to 15 ng/mL.

Extended release (Advagraf [Canadian product]):

Months 1 to 3: 7 to 16 ng/mL.

> Month 3: 5 to 15 ng/mL.

Extended release (Envarsus XR):

Month 1: 6 to 11 ng/mL.

> Month 1: 4 to 11 ng/mL.

Extended release (Envarsus PA [Canadian product]):

Months 1 to 3: 7 to 20 ng/mL.

> Month 3: 5 to 15 ng/mL.

Liver transplant: Whole blood trough concentrations:

Immediate release: Months 1 to 12: 5 to 20 ng/mL.

Recommended therapeutic ranges when administered in combination with everolimus for liver transplant (Zortress product labeling 2013): By 3 weeks after first everolimus dose and through month 12 post-transplant: 3 to 5 ng/mL.

Extended release (Advagraf [Canadian product], Envarsus PA [Canadian product]):

Months 1 to 2: 5 to 20 ng/mL.

> Month 2: 5 to 15 ng/mL.

Intestinal transplant (off-label use): Months 1 to 3: 10 to 20 ng/mL followed by gradual dose reduction; concentrations allowed to decrease over the first year so that by 12 months, trough levels range from 5 to 10 ng/mL (Abu-Elmagd 2009b; Horslen 2006).

Lung transplant:

Immediate release:

In combination with azathioprine or mycophenolate mofetil:

Months 1 to 3: 10 to 15 ng/mL.

Months 4 to 12: 8 to 12 ng/mL.

Or: Whole blood trough concentrations: 5 to 15 ng/mL (ACCP [Baughman 2012]).

Pancreas transplant (off-label use): Whole blood trough concentration: 8 to 15 ng/mL ≤6 months post transplant; 5 to 10 ng/mL >6 months post transplant (Bechstein 2004).

Myasthenia gravis (off-label use): Whole blood trough concentrations: 8 to 9 ng/mL (Sanders 2016).

Prevention of graft-versus-host disease (off-label use): Whole blood trough concentrations: 10 to 20 ng/mL (Uberti 1999), although some institutions use a lower limit of 5 ng/mL and an upper limit of 15 ng/mL (Przepiorka 1999; Yanik 2000).

Mechanism of Action

Tacrolimus suppresses cellular immunity (inhibits T-lymphocyte activation) by binding to an intracellular protein, FKBP-12 and complexes with calcineurin dependent proteins to inhibit calcineurin phosphatase activity

Pharmacokinetics (Adult Data Unless Noted)

Absorption: Better in resected patients with a closed stoma; unlike cyclosporine, clamping of the T-tube in liver transplant recipients does not alter trough concentrations or AUC; Oral: Incomplete and variable (5% to 67%); the rate and extent of absorption is decreased (27%) by food (particularly a high-fat meal). Oral absorption may be variable in hematopoietic cell transplant recipients with mucositis due to the conditioning regimen. Systemic exposure (AUC) of single dose granules was ~16% higher than single dose capsules.

Distribution: Distributes to erythrocytes, lung, kidneys, pancreas, liver, heart, and spleen; Vd: Mean: Infants and Children: 2.6 L/kg in liver transplant recipients; Adults: 0.85 to 1.41 L/kg in liver and kidney transplant recipients.

Protein binding: ~99% primarily to albumin and alpha-1 acid glycoprotein

Metabolism: Extensively hepatic via CYP3A4 to eight possible metabolites (major metabolite, 31-demethyl tacrolimus, shows same activity as tacrolimus in vitro)

Bioavailability: Oral: Incomplete and variable: Immediate release:

Infants and Children: Kidney transplant: 19% ± 14%, liver transplant: 31% ± 24%

Adults: Kidney transplant: 17% ± 10%, liver transplant: 22% ± 6%, heart transplant: 23% ± 9%

Half-life elimination:

Children: Kidney transplant: 10.2 ± 5 hours; Infants and Children: Liver transplant: 11.5 ± 3.8 hours

Adults:

Immediate release: Variable, 23 to 46 hours in healthy volunteers; 2.1 to 36 hours in transplant recipients; prolonged in patients with severe hepatic impairment.

Extended release: 38 ± 3 hours; prolonged in patients with severe hepatic impairment

Time to peak: Oral: 0.5 to 6 hours

Excretion: Feces (~93%); urine (<1% as unchanged drug)

Clearance: 7 to 103 mL/minute/kg (average: 30 mL/minute/kg); clearance higher in children

Pharmacokinetics: Additional Considerations (Adult Data Unless Noted)

Hepatic function impairment: The mean clearance was substantially lower in patients with severe hepatic impairment.

Race/Ethnicity: Cmax is lower, Tmax is prolonged, and bioavailability is decreased following oral (but not IV) administration in African American kidney transplant recipients.

Cystic fibrosis: Lower tacrolimus bioavailability has been reported in patients with cystic fibrosis.

Brand Names: International
International Brand Names by Country
For country code abbreviations (show table)

  • (AE) United Arab Emirates: Advagraf;
  • (AR) Argentina: Prograf | Tacrolimus sandoz | Tartrime;
  • (AT) Austria: Advagraf | Envarsus | Modigraf | Prograf | Tacforius | Tacni transplant | Tacrolimus sandoz;
  • (AU) Australia: Advagraf xl | Pacrolim | Pharmacor tacrolimus | Prograf | Tacrograf | Tacrolimus sandoz;
  • (BE) Belgium: Adoport | Prograft | Tacni | Tacrolimus EG | Tacrolimus sandoz;
  • (BG) Bulgaria: Advagraf | Envarsus | Prograf | Tacni;
  • (BR) Brazil: Prograf | Tacrofort | Tacrolil | Tacrolimo | Tarfic;
  • (CH) Switzerland: Advagraf | Envarsus | Modigraf | Prograf | Tacni | Tacrolimus Actavis | Tacrolimus sandoz | Tacrolimus Teva;
  • (CL) Chile: Cidimus | Cidimus xl | Crolim | Prograf | T inmun | Tacni | Tacni xr;
  • (CN) China: Fu mei xin | Prograf;
  • (CO) Colombia: Framebin | Prejotac | Prograf | Prograf xl | Taclim;
  • (CZ) Czech Republic: Apo tacrolimus | Envarsus | Gecrol | Modigraf | Prograf | Tacforius | Tacni | Tacrolimus accord | Tacrolimus Mylan | Tacrolimus sandoz;
  • (DE) Germany: Adoport | Advagraf | Crilomus | Dailiport | Envarsus | Modigraf | Prograf | Tacni | Tacpan | Tacro cell | Tacrolimus accord | Tacrolimus Aliud | Tacrolimus Heumann | Tacrolimus Hexal | Vivadex;
  • (DO) Dominican Republic: Prograf | Prograf xl | T inmun;
  • (EC) Ecuador: T inmun;
  • (EE) Estonia: Advagraf | Envarsus | Modigraf | Prograf;
  • (EG) Egypt: Adport | Advagraf | Prograf;
  • (ES) Spain: Adoport | Conferoport | Envarsus | Modigraf | Prograf | Tacforius | Tacni | Tacrolimus accord | Tacrolimus Mylan | Tacrolimus sandoz | Tacrolimus Stada | Tartrime;
  • (ET) Ethiopia: Tacrolimus;
  • (FI) Finland: Adport | Advagraf | Envarsus | Modigraf | Prograf | Tacni | Tacrolimus accord | Tacrolimus sandoz;
  • (FR) France: Adoport | Advagraf | Conferoport | Envarsus | Modigraf | Prograf | Tacrolimus EG;
  • (GB) United Kingdom: Adoport | Capexion | Envarsus | Modigraf | Prograf | Tacni | Tacrolimus Teva | Vivadex;
  • (GR) Greece: Advagraf | Prograf | Tacrolimus/generics;
  • (HK) Hong Kong: Advagraf | Prograf | Tacrolimus sandoz;
  • (HR) Croatia: Advagraf | Envarsus | Prograf | Tacrocel;
  • (HU) Hungary: Envarsus | Modigraf | Prograf | Tacforius | Tacni | Tacrolimus Mylan | Tacrolimus sandoz | Tacrolimus Stada;
  • (ID) Indonesia: Prograf;
  • (IE) Ireland: Envarsus | Modigraf | Prograf;
  • (IL) Israel: Prograf;
  • (IN) India: Advagraf | Amigraf | Biomus | Crolim | Emgraf | Imunotac | Loprolif | Mylimus | Od venta | Olmis | Pangraf | Prograf | Rengraf | Rolitrans | Seegraf | Taclicept | Tacrograf | Tacromus | Tacroren | Tacrosan | Tacrotec | Tacrotor | Tacsant | Tacva | Takfa | Vingraf;
  • (IT) Italy: Adoport | Conferoport | Envarsus | Prograf | Tacforius | Tacni | Tacrolimus Mylan;
  • (JO) Jordan: Advagraf | Pangraf | Prograf;
  • (JP) Japan: Graceptor | Prograf;
  • (KE) Kenya: Adoport | Advagraf | Pangraf | Tacrocord | Tacrograf | Tacrotas | Tacroz | Tcoglob | Vingraf;
  • (KR) Korea, Republic of: Advagraf | Cellogra | Celogra | Newromis | Prograf | Tacrobell | Tacrobell sr | Tacroin | Tacroli | Tacromin | Tacros | Tacross | Tarimus | Tarsin | Unirori;
  • (KW) Kuwait: Advagraf | Modigraf | Prograf;
  • (LB) Lebanon: Advagraf | Prograf | Rolitac;
  • (LT) Lithuania: Advagraf | Envarsus | Modigraf | Prograf | Tacrolimus accord;
  • (LU) Luxembourg: Prograft | Tacni;
  • (LV) Latvia: Advagraf | Envarsus | Modigraf | Prograf | Tacrolimus pharmaswiss;
  • (MX) Mexico: Akrocell | Framebin | Limustin | Octralin | Profelam | Prograf | Prograf xl | Tevacrol | Tinmacrol;
  • (MY) Malaysia: Advagraf | Prograf;
  • (NL) Netherlands: Adport | Advagraf | Envarsus | Modigraf | Prograf | Prograft | Tacforius | Tacni | Tacrolimus accord | Tacrolimus sandoz;
  • (NO) Norway: Adport | Advagraf | Envarsus | Modigraf | Prograf | Tacni | Tacrolimus sandoz;
  • (NZ) New Zealand: Advagraf xl | Prograf | Tacrolimus sandoz;
  • (PE) Peru: Norgraf | T inmun;
  • (PH) Philippines: Advagraf | Cidimus | Imugraft | Prograf | Tagraf | Vingraf;
  • (PK) Pakistan: Cyonse | Tacgraf | Tacogen | Tacrobell | Tacrograf | Tacrolim | Tacromed | Tacromus | Tacrosan | Taklida | Talimus | Unigraf;
  • (PL) Poland: Cidimus | Envarsus | Modigraf | Prograf | Tacni | Taliximun;
  • (PR) Puerto Rico: Astagraf Xl | Hecoria | Prograf;
  • (PT) Portugal: Advagraf | Modigraf | Prograf | Tacforius | Tacni;
  • (PY) Paraguay: Armonil | Graftac | Granmac | Prograf | Prograf xl | T inmun | Tacrolil | Tacrolimus bauel top | Tacrolimus imedic | Tacrolimus libra | Tacrolimus pisa | Tacrolimus prosalud | Tacrolimus sandoz | Tenacrine;
  • (QA) Qatar: Advagraf | Prograf | Rolitac;
  • (RO) Romania: Advagraf | Prograf | Tacni | Tacrolimus sandoz;
  • (RU) Russian Federation: Advagraf | Grastiva | Pangraf | Priluxid | Prograf | Redinesp | Tacrolimus | Tacrolimus akri | Tacrolimus richter | Tacrolimus Stada | Tacrolimus Teva | Tacrosel;
  • (SA) Saudi Arabia: Advagraf | Envarsus xr | Prograf | Rolitac;
  • (SE) Sweden: Adoport | Adport | Advagraf | Envarsus | Modigraf | Prograf | Tacforius | Tacni | Tacrolimus 2care4 | Tacrolimus accord | Tacrolimus eql | Tacrolimus sandoz | Takrolimus ebb;
  • (SG) Singapore: Advagraf | Prograf;
  • (SI) Slovenia: Advagraf | Envarsus | Modigraf | Prograf | Tacforius | Tacni;
  • (SK) Slovakia: Gecrol | Modigraf | Prograf | Tacforius | Takrolimus sandoz;
  • (TH) Thailand: Advagraf | Prograf;
  • (TN) Tunisia: Advagraf | Prograf | Tacrolimus medis;
  • (TR) Turkey: Adoport | Advagraf | Oligma | Panolimus | Prograf;
  • (TW) Taiwan: Advagraf | Prograf | Zygraf;
  • (UA) Ukraine: Advagraf | Envarsus | Modigraf | Prograf;
  • (UY) Uruguay: Prograf;
  • (ZA) South Africa: Advagraf | Graftac | Prograf | Tacrum | Talomune | Tarograf
  1. <800> Hazardous Drugs—Handling in Healthcare Settings. United States Pharmacopeia and National Formulary (USP 43-NF 38). Rockville, MD: United States Pharmacopeia Convention; 2020:74-92.
  2. Abu-Elmagd KM, Costa G, Bond GJ, et al. Evolution of the immunosuppressive strategies for the intestinal and multivisceral recipients with special reference to allograft immunity and achievement of partial tolerance. Transpl Int. 2009a;22(1):96-109. [PubMed 18954362]
  3. Abu-Elmagd KM, Costa G, Bond GJ, et al. Five hundred intestinal and multivisceral transplantations at a single center: major advances with new challenges. Ann Surg. 2009b;250(4):567-581. [PubMed 19730240]
  4. Acott P, Babel N. BK virus replication following kidney transplant: does the choice of immunosuppressive regimen influence outcomes? Ann Transplant. 2012;17(1):86-99. doi:10.12659/aot.882640 [PubMed 22466913]
  5. Advagraf (tacrolimus) [product monograph]. Markham, Ontario, Canada: Astellas Pharma Canada Inc; December 2022.
  6. Advagraf XL (tacrolimus) [product monograph]. Markham, Ontario, Canada: Astellas Pharma Canada Inc; December 2020.
  7. Afshar K. Future direction of immunosuppression in lung transplantation. Curr Opin Organ Transplant. 2014;19(6):583-590. [PubMed 25260152]
  8. Akamine Y, Fujiyama N, Kagaya H, et al. Tacrolimus concentrations after renal transplantation in a mother-neonate dyad: maternal, neonatal and breast milk measurements. J Clin Pharm Ther. 2021;46(6):1800-1803. doi:10.1111/jcpt.1345 [PubMed 34041772]
  9. Al-Nouri ZL, Reese JA, Terrell DR, Vesely SK, George JN. Drug-induced thrombotic microangiopathy: a systematic review of published reports. Blood. 2015;125(4):616-618. doi:10.1182/blood-2014-11-611335 [PubMed 25414441]
  10. Al-Uzri A, Yorgin PD, Kling PJ. Anemia in children after transplantation: etiology and the effect of immunosuppressive therapy on erythropoiesis. Pediatr Transplant. 2003;7(4):253-264. doi:10.1034/j.1399-3046.2003.00042.x [PubMed 12890002]
  11. Anghel D, Tanasescu R, Campeanu A, Lupescu I, Podda G, Bajenaru O. Neurotoxicity of immunosuppressive therapies in organ transplantation. Maedica (Bucur). 2013;8(2):170-175. [PubMed 24371481]
  12. Arikan C, Kilic M, Tokat Y, Aydogdu S. Allergic disease after pediatric liver transplantation with systemic tacrolimus and cyclosporine a therapy. Transplant Proc. 2003;35(8):3039-41. doi:10.1016/j.transproceed.2003.10.033 [PubMed 14697973]
  13. Artz MA, Boots JM, Ligtenberg G, et al. Conversion from cyclosporine to tacrolimus improves quality-of-life indices, renal graft function and cardiovascular risk profile. Am J Transplant. 2004;4(6):937-945. doi:10.1111/j.1600-6143.2004.00427.x [PubMed 15147428]
  14. Asante-Korang A, Boyle GJ, Webber SA, et al, “Experience of FK506 Immune Suppression in Pediatric Heart Transplantation: A Study of Long-Term Adverse Effects,” J Heart Lung Transplant, 1996, 15(4):415-22. [PubMed 8732602]
  15. ASHP. Standardize 4 Safety Initiative Compounded Oral Liquid Version 1.01. July 2017. https://www.ashp.org/-/media/assets/pharmacy-practice/s4s/docs/s4s-ashp-oral-compound-liquids.ashx?la=en&hash=4C2E4F370B665C028981B61F6210335AD5D0D1D6.
  16. Astagraf XL (tacrolimus) [prescribing information]. Northbrook, IL: Astellas Pharma US Inc; February 2014.
  17. Astagraf XL (tacrolimus) [prescribing information]. Northbrook, IL: Astellas Pharma US Inc; November 2022.
  18. Atkison P, Joubert G, Barron A, et al, “Hypertrophic Cardiomyopathy Associated With Tacrolimus in Paediatric Transplant Patients,” Lancet, 1995, 345(8954):894-6. [PubMed 7535875]
  19. Ayyala RS, Arnold SD, Bhatia M, Dastgir J. Imaging findings in a child with calcineurin inhibitor-induced pain syndrome after bone marrow transplant for beta thalassemia major. Pediatr Radiol. 2016;46(11):1618-1621. doi:10.1007/s00247-016-3644-5 [PubMed 27324395]
  20. Bagg A, Dunphy CH. Immunosuppressive and immunomodulatory therapy-associated lymphoproliferative disorders. Semin Diagn Pathol. 2013;30(2):102-212. doi:10.1053/j.semdp.2012.08.005 [PubMed 23541274]
  21. Bartynski WS, Tan HP, Boardman JF, Shapiro R, Marsh JW. Posterior reversible encephalopathy syndrome after solid organ transplantation. AJNR Am J Neuroradiol. 2008;29(5):924-930. doi:10.3174/ajnr.A0960 [PubMed 18272559]
  22. Baughman RP, Meyer KC, Nathanson I, et al. Monitoring of nonsteroidal immunosuppressive drugs in patients with lung disease and lung transplant recipients: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 2012;142(5):e1S-e111S. [PubMed 23131960]
  23. Bechstein WO, Malaise J, Saudek F, et al; EuroSPK Study Group. Efficacy and safety of tacrolimus compared with cyclosporine microemulsion in primary simultaneous pancreas-kidney transplantation: 1-year results of a large multicenter trial. Transplantation. 2004;77(8):1221-1228. doi:10.1097/01.tp.0000120865.96360.df [PubMed 15114089]
  24. Bechstein WO. Neurotoxicity of calcineurin inhibitors: impact and clinical management. Transpl Int. 2000;13(5):313-326. doi:10.1007/s001470050708 [PubMed 11052266]
  25. Bellón T. Mechanisms of severe cutaneous adverse reactions: recent advances. Drug Saf. 2019;42(8):973-992. doi:10.1007/s40264-019-00825-2 [PubMed 31020549]
  26. Bentata Y. Tacrolimus: 20 years of use in adult kidney transplantation. What we should know about its nephrotoxicity. Artif Organs. 2020;44(2):140-152. doi:10.1111/aor.13551 [PubMed 31386765]
  27. Bianchi A, Ragonese P, Banco MA, et al. Four cases of progressive multifocal leukoencephalopathy in iatrogenic immunocompromised patients. eNeurologicalSci. 2020;19:100243. doi:10.1016/j.ensci.2020.100243 [PubMed 32478179]
  28. Bloom RD, Rao V, Weng F, Grossman RA, Cohen D, Mange KC. Association of hepatitis C with posttransplant diabetes in renal transplant patients on tacrolimus. J Am Soc Nephrol. 2002;13(5):1374-1380. doi:10.1097/01.asn.0000012382.97168.e0 [PubMed 11961026]
  29. Bock ME, Cohn RA, Ali FN. Treatment of childhood nephrotic syndrome with long-term, low-dose tacrolimus. Clin Nephrol. 2013;79(6):432-438. [PubMed 23380387]
  30. Boyer NL, Niven A, Edelman J. Tacrolimus-associated thrombotic microangiopathy in a lung transplant recipient. BMJ Case Rep. 2013;2013:bcr2012007351. doi:10.1136/bcr-2012-007351 [PubMed 23396921]
  31. Braithwaite HE, Darley DR, Brett J, Day RO, Carland JE. Identifying the association between tacrolimus exposure and toxicity in heart and lung transplant recipients: A systematic review. Transplant Rev (Orlando). 2021;35(2):100610. doi:10.1016/j.trre.2021.100610 [PubMed 33756310]
  32. Bramham K, Chusney G, Lee J, et al, "Breastfeeding and Tacrolimus: Serial Monitoring in Breast-Fed and Bottle-Fed Infants," Clin J Am Soc Nephrol, 2013, Apr;8(4):563-7. [PubMed 23349333]
  33. Caillard S, Dharnidharka V, Agodoa L, Bohen E, Abbott K. Posttransplant lymphoproliferative disorders after renal transplantation in the United States in era of modern immunosuppression. Transplantation. 2005;80(9):1233-1243. doi:10.1097/01.tp.0000179639.98338.39 [PubMed 16314791]
  34. Caillard S, Lelong C, Pessione F, Moulin B; French PTLD Working Group. Post-transplant lymphoproliferative disorders occurring after renal transplantation in adults: report of 230 cases from the French Registry. Am J Transplant. 2006;6(11):2735-2742. doi:10.1111/j.1600-6143.2006.01540.x [PubMed 17049061]
  35. Chakkera HA, Kudva Y, Kaplan B. Calcineurin inhibitors: Pharmacologic mechanisms impacting both insulin resistance and insulin secretion leading to glucose dysregulation and diabetes mellitus. Clin Pharmacol Ther. 2017;101(1):114-120. doi:10.1002/cpt.546 [PubMed 27804122]
  36. Chiurchiu C, Ruggenenti P, Remuzzi G. Thrombotic microangiopathy in renal transplantation. Ann Transplant. 2002;7(1):28-33. [PubMed 12221901]
  37. Chockalingam R, Downing C, Tyring SK. Cutaneous squamous cell carcinomas in organ transplant recipients. J Clin Med. 2015;4(6):1229-1239. doi:10.3390/jcm4061229 [PubMed 26239556]
  38. Coe CL, Horst SN, Izzy MJ. Neurologic toxicities associated with tumor necrosis factor inhibitors and calcineurin inhibitors. Neurol Clin. 2020;38(4):937-951. doi:10.1016/j.ncl.2020.07.009 [PubMed 33040870]
  39. Collin C, Boussaud V, Lefeuvre S, et al. Sublingual tacrolimus as an alternative to intravenous route in patients with thoracic transplant: a retrospective study. Transplant Proc. 2010;42(10):4331-4337. [PubMed 21168693]
  40. Constantinescu S, Pai A, Coscia LA, Davison JM, Moritz MJ, Armenti VT. Breast-feeding after transplantation. Best Pract Res Clin Obstet Gynaecol. 2014;28(8):1163‐1173. doi:10.1016/j.bpobgyn.2014.09.001 [PubMed 25271063]
  41. Cowlrick I, Delventhal H, Kaipainen K, Krcmar C, Petan J, Schleibner S. Three-year follow-up of malignancies in tacrolimus-treated renal recipients--an analysis of European multicentre studies. Clin Transplant. 2008;22(3):372-377. doi:10.1111/j.1399-0012.2008.00796.x [PubMed 18279418]
  42. Cox KL, Lawrence-Miyasaki LS, Garcia-Kennedy R, et al. An increased incidence of Epstein-Barr virus infection and lymphoproliferative disorder in young children on FK506 after liver transplantation. Transplantation. 1995;59(4):524-529. [PubMed 7533344]
  43. Cruz JL, Wolff ML, Vanderman AJ, Brown JN. The emerging role of tacrolimus in myasthenia gravis. Ther Adv Neurol Disord. 2015;8(2):92-103. doi: 10.1177/1756285615571873. [PubMed 25922621]
  44. Dai C, Walker JT, Shostak A, et al. Tacrolimus- and sirolimus-induced human β cell dysfunction is reversible and preventable. JCI Insight. 2020;5(1):e130770. doi:10.1172/jci.insight.130770 [PubMed 31941840]
  45. Darlenski R. Probable contact urticaria caused by tacrolimus-containing ointment in the treatment of atopic dermatitis. J Allergy Clin Immunol Pract. 2019;7(5):1665-1667. doi:10.1016/j.jaip.2018.11.025 [PubMed 30513360]
  46. Davidson JA, Wilkinson A; International Expert Panel on New-Onset Diabetes after Transplantation. New-onset diabetes after transplantation 2003 International Consensus Guidelines: an endocrinologist's view. Diabetes Care. 2004;27(3):805-812. doi:10.2337/diacare.27.3.805 [PubMed 14988309]
  47. Dayton JD, Richmond ME, Weintraub RG, Shipp AT, Orjuela M, Addonizio LJ. Role of immunosuppression regimen in post-transplant lymphoproliferative disorder in pediatric heart transplant patients. J Heart Lung Transplant. 2011;30(4):420-425. doi:10.1016/j.healun.2010.10.004 [PubMed 21147001]
  48. De Waele L, Van Gaal PJ, Abramowicz D. Electrolytes disturbances after kidney transplantation. Acta Clin Belg. 2019;74(1):48-52. doi:10.1080/17843286.2018.1549193 [PubMed 30482110]
  49. Dharnidharka VR, Ho PL, Stablein DM, Harmon WE, Tejani AH. Mycophenolate, tacrolimus and post-transplant lymphoproliferative disorder: a report of the North American Pediatric Renal Transplant Cooperative Study. Pediatr Transplant. 2002;6(5):396-399. doi:10.1034/j.1399-3046.2002.00021.x [PubMed 12390426]
  50. DiGiuseppe JA, Bastacky SI, Shirey RS, Silberman MA, Hutchins GM, Ness PM. Tacrolimus-related posttransplant lymphoproliferative disorder presenting as autoimmune hemolytic anemia. Arch Pathol Lab Med. 1996;120(3):282-285. [PubMed 8629906]
  51. Ding Y, Qu C, He H, Cao F, Ou T, Li F. Case report: acute pancreatitis associated with tacrolimus in kidney transplantation and a review of the literature. Front Med (Lausanne). 2022;9:843870. doi:10.3389/fmed.2022.843870 [PubMed 35530036]
  52. Doligalski CT, Liu EC, Sammons CM, Silverman A, Logan AT. Sublingual administration of tacrolimus: current trends and available evidence. Pharmacotherapy. 2014;34(11):1209-1219. [PubMed 25251980]
  53. Doobay R, Gambhir HS. Cardiac tamponade induced by tacrolimus toxicity. Am J Ther. 2018;25(6):e683-e684. doi:10.1097/MJT.0000000000000630 [PubMed 28708698]
  54. EBPG Expert Group on Renal Transplantation, "European Best Practice Guidelines For Renal Transplantation. Section IV: Long-Term Management of the Transplant Recipient, IV.10. Pregnancy in Renal Transplant Recipients," Nephrol Dial Transplant, 2002, 17 Suppl 4:50-5. [PubMed 12091650]
  55. Egeland EJ, Reisaeter AV, Robertsen I, et al. High tacrolimus clearance - a risk factor for development of interstitial fibrosis and tubular atrophy in the transplanted kidney: a retrospective single-center cohort study. Transpl Int. 2019;32(3):257-269. doi:10.1111/tri.13356 [PubMed 30252957]
  56. Ekberg H, Bernasconi C, Tedesco-Silva H, et al. Calcineurin inhibitor minimization in the Symphony study: observational results 3 years after transplantation. Am J Transplant. 2009;9(8):1876-1885. doi:10.1111/j.1600-6143.2009.02726.x [PubMed 19563339]
  57. ElChaki R, Ettenger R, Lee S, et al. Envarsus XR pharmacokinetics in adolescents post-kidney transplantation - A pilot study. Pediatr Transplant. 2023;27(3):e14480. doi:10.1111/petr.14480 [PubMed 36732080]
  58. Elefante A, Muindi J, West K, et al, “Long-Term Stability of a Patient-Convenient 1 mg/mL Suspension of Tacrolimus for Accurate Maintenance of Stable Therapeutic Levels,” Bone Marrow Transplant, 2006, 37(8):781-4. [PubMed 16518428]
  59. Elfeky R, Lucchini G, Lum SH, et al. New insights into risk factors for transplant-associated thrombotic microangiopathy in pediatric HSCT. Blood Adv. 2020;4(11):2418-2429. doi:10.1182/bloodadvances.2019001315 [PubMed 32492158]
  60. Ellis D, Jaffe R, Green M, et al. Epstein-Barr virus-related disorders in children undergoing renal transplantation with tacrolimus-based immunosuppression. Transplantation. 1999;68(7):997-1003. doi:10.1097/00007890-199910150-00017 [PubMed 10532541]
  61. Envarsus PA (tacrolimus) [product monograph]. Saint-Laurent, Quebec, Canada: Paladin Labs Inc; May 2020.
  62. Envarsus XR (tacrolimus) [prescribing information]. Cary, NC: Veloxis Pharmaceuticals Inc; September 2020.
  63. Esquivel C, So S, McDiarmid S, Andrews W, and Colombani PM, “Suggested Guidelines for the Use of Tacrolimus in Pediatric Liver Transplant Patients,” Transplantation, 1996, 61(5):847-8. [PubMed 8607198]
  64. Expert opinion. Senior Renal Editorial Team: Bruce Mueller, PharmD, FCCP, FASN, FNKF; Jason A. Roberts, PhD, BPharm (Hons), B App Sc, FSHP, FISAC; Michael Heung, MD, MS.
  65. Farouk SS, Rein JL. The many faces of calcineurin inhibitor toxicity-what the FK? Adv Chronic Kidney Dis. 2020;27(1):56-66. doi:10.1053/j.ackd.2019.08.006 [PubMed 32147003]
  66. Ferrari U, Empl M, Kim KS, Sostak P, Förderreuther S, Straube A. Calcineurin inhibitor-induced headache: clinical characteristics and possible mechanisms. Headache. 2005;45(3):211-214. doi:10.1111/j.1526-4610.2005.05046.x [PubMed 15836594]
  67. Filler G, Webb NJ, Milford DV, et al. Four-year data after pediatric renal transplantation: a randomized trial of tacrolimus vs cyclosporin microemulsion. Pediatr Transplant. 2005; 9(4):498-503. [PubMed 16048603]
  68. Foster JA, Jacobson PA, Johnson CE, et al, “Stability of Tacrolimus in an Extemporaneously Compounded Oral Liquid (Abstract of Meeting Presentation),” American Society of Health-System Pharmacists Annual Meeting, 1996, 53:P-52(E). [PubMed 9117806]
  69. French AE, Soldin SJ, Soldin OP, et al, “Milk Transfer and Neonatal Safety of Tacrolimus,” Ann Pharmacother, 2003, 37(6):815-8. [PubMed 12773068]
  70. Freriksen JJM, Feyaerts D, van den Broek PHH, et al. Placental disposition of the immunosuppressive drug tacrolimus in renal transplant recipients and in ex vivo perfused placental tissue. Eur J Pharm Sci. 2018;119:244‐248. doi:10.1016/j.ejps.2018.04.017 [PubMed 29655601]
  71. Fuchs U, Klein S, Zittermann A, Ensminger SM, Schulz U, Gummert JF. Incidence of malignant neoplasia after heart transplantation--a comparison between cyclosporine a and tacrolimus. Ann Transplant. 2014;19:300-304. doi:10.12659/AOT.890199 [PubMed 24953848]
  72. Fuchs U, Zittermann A, Ensminger SM, Schulz U, Gummert JF. Clinical outcome in heart transplant recipients receiving everolimus in combination with dosage reduction of the calcineurin inhibitor cyclosporine A or tacrolimus. Transpl Immunol. 2014;31(2):87-91. [PubMed 24932812]
  73. Funch DP, Ko HH, Travasso J, Brady J, Kew CE 2nd, Nalesnik MA, Walker AM. Posttransplant lymphoproliferative disorder among renal transplant patients in relation to the use of mycophenolate mofetil. Transplantation. 2005;80(9):1174-1180. doi:10.1097/01.tp.0000169035.10572.c6 [PubMed 16314782]
  74. Furlan V, Perello L, Jacquemin E, et al, “Interactions Between FK506 and Rifampicin or Erythromycin in Pediatric Liver Recipients,” Transplantation, 1995, 59(8):1217-8. [PubMed 7537398]
  75. Furlong T, Storb R, Anasetti C, et al, “Clinical Outcome After Conversion to FK 506 (Tacrolimus) Therapy for Acute Graft-Versus-Host Disease Resistant to Cyclosporine or for Cyclosporine-Associated Toxicities,” Bone Marrow Transplant, 2000, 26(9):985-91. [PubMed 11100278]
  76. Gardiner SJ and Begg EJ, “Breastfeeding During Tacrolimus Therapy,” Obstet Gynecol, 2006, 107(2 Pt 2):453-5. [PubMed 16449146]
  77. Garrity ER Jr, Hertz MI, Trulock EP, Keenan R, Love R. Suggested guidelines for the use of tacrolimus in lung-transplant recipients. J Heart Lung Transplant. 1999;18(3):175-176. [PubMed 10328140]
  78. Geetha D, Zachary JB, Baldado HM, Kronz JD, Kraus ES. Pure red cell aplasia caused by Parvovirus B19 infection in solid organ transplant recipients: a case report and review of literature. Clin Transplant. 2000;14(6):586-591. doi:10.1034/j.1399-0012.2000.140612.x [PubMed 11127313]
  79. George JN, Nester CM. Syndromes of thrombotic microangiopathy. N Engl J Med. 2014;371(7):654-666. doi:10.1056/NEJMra1312353 [PubMed 25119611]
  80. Gipson DR, Larkin T, Seifert R, Black LV. Romiplostim as a therapeutic intervention for tacrolimus-induced immune thrombocytopenia in a pediatric cardiac transplant patient. J Pediatr Hematol Oncol. 2021;43(6):e777-e779. doi:10.1097/MPH.0000000000001994 [PubMed 33181586]
  81. Gmitterová K, Minár M, Žigrai M, Košutzká Z, Kušnírová A, Valkovič P. Tacrolimus-induced parkinsonism in a patient after liver transplantation - case report. BMC Neurol. 2018;18(1):44. doi:10.1186/s12883-018-1052-1 [PubMed 29678162]
  82. Goodman SM, Springer BD, Chen AF, et al. 2022 American College of Rheumatology/American Association of Hip and Knee Surgeons guideline for the perioperative management of antirheumatic medication in patients with rheumatic diseases undergoing elective total hip or total knee arthroplasty. Arthritis Care Res (Hoboken). 2022;74(9):1399-1408. doi:10.1002/acr.24893 [PubMed 35718887]
  83. Gouraud A, Bernard N, Millaret A, et al. Follow-up of tacrolimus breastfed babies. Transplantation. 2012;94(6):e38‐e40. doi:10.1097/TP.0b013e3182654d36 [PubMed 22996303]
  84. Granot E, Yakobovich E, Bardenstein R. Tacrolimus immunosuppression - an association with asymptomatic eosinophilia and elevated total and specific IgE levels. Pediatr Transplant. 2006;10(6):690-693. doi:10.1111/j.1399-3046.2006.00542.x [PubMed 16911492]
  85. Green M. Introduction: Infections in solid organ transplantation. Am J Transplant. 2013;13 Suppl 4:3-8. doi:10.1111/ajt.12093 [PubMed 23464993]
  86. Grimer M and Caring for Australians with Renal Impairment (CARI), “The CARI Guidelines. Calcineurin Inhibitors in Renal Transplantation: Pregnancy, Lactation and Calcineurin Inhibitors,” Nephrology (Carlton), 2007, 12(Suppl 1):98-105. [PubMed 17316288]
  87. Groetzner J, Reichart B, Roemer U, et al. Cardiac transplantation in pediatric patients: fifteen-year experience of a single center. Ann Thorac Surg. 2005;79(1):53-60. [PubMed 15620914]
  88. Grouzmann E, Buclin T, Biollaz J. Misleading tacrolimus concentration value in blood taken from a catheter used for tacrolimus administration. Am J Health Syst Pharm. 2008;65(3):226-228. doi:10.2146/ajhp070054 [PubMed 18216007]
  89. Gruber S, Tiringer K, Dehlink E, et al. Allergic sensitization in kidney-transplanted patients prevails under tacrolimus treatment. Clin Exp Allergy. 2011;41(8):1125-1132. doi:10.1111/j.1365-2222.2011.03761.x [PubMed 21545550]
  90. Gruber SA, Hewitt JM, Sorenson AL, et al. Pharmacokinetics of FK506 after intravenous and oral administration in patients awaiting renal transplantation. J Clin Pharmacol. 1994;34(8):859-864. doi:10.1002/j.1552-4604.1994.tb02052.x [PubMed 7525661]
  91. Guethoff S, Stroeh K, Grinninger C, et al. De novo sirolimus with low-dose tacrolimus versus full-dose tacrolimus with mycophenolate mofetil after heart transplantation--8-year results. J Heart Lung Transplant. 2015;34(5):634-642. [PubMed 25701373]
  92. Gulati A, Sinha A, Gupta A, et al. Treatment with tacrolimus and prednisolone is preferable to intravenous cyclophosphamide as the initial therapy for children with steroid-resistant nephrotic syndrome. Kidney Int. 2012;82(10):1130-1135. [PubMed 22763815]
  93. Gulleroglu K, Baskin E, Bayrakci U, et al. Sudden hearing loss associated with tacrolimus after pediatric renal transplant. Exp Clin Transplant. 2013;11(6):562-564. doi:10.6002/ect.2012.0241 [PubMed 24344948]
  94. Hachem RR, Yusen RD, Chakinala MM, et al. A randomized controlled trial of tacrolimus versus cyclosporine after lung transplantation. J Heart Lung Transplant. 2007;26(10):1012-1018. [PubMed 17919621]
  95. Hebert MF, Zheng S, Hays K, et al, "Interpreting Tacrolimus Concentrations During Pregnancy and Postpartum," Transplantation. 2013, 95(7):908-15. [PubMed 23274970]
  96. Higgins R, Ramaiyan K, Dasgupta T, et al. Hyponatraemia and hyperkalaemia are more frequent in renal transplant recipients treated with tacrolimus than with cyclosporin. Further evidence for differences between cyclosporin and tacrolimus nephrotoxicities. Nephrol Dial Transplant. 2004;19(2):444-450. doi:10.1093/ndt/gfg515 [PubMed 14736972]
  97. Hiramatsu Y, Yoshida S, Kotani T, et al. Changes in the blood level, efficacy, and safety of tacrolimus in pregnancy and the lactation period in patients with systemic lupus erythematosus. Lupus. 2018;27(14):2245‐2252. doi:10.1177/0961203318809178 [PubMed 30394835]
  98. Hodak SP, Moubarak JB, Rodriguez I, Gelfand MC, Alijani MR, Tracy CM. QT prolongation and near fatal cardiac arrhythmia after intravenous tacrolimus administration: a case report. Transplantation. 1998;66(4):535-537. doi:10.1097/00007890-199808270-00021 [PubMed 9734501]
  99. Holman MJ, Gonwa TA, Cooper B, et al. FK506-associated thrombotic thrombocytopenic purpura. Transplantation. 1993;55(1):205-206. [PubMed 7678357]
  100. Hoorn EJ, Walsh SB, McCormick JA, et al. The calcineurin inhibitor tacrolimus activates the renal sodium chloride cotransporter to cause hypertension. Nat Med. 2011;17(10):1304-1309. doi:10.1038/nm.2497 [PubMed 21963515]
  101. Hoorn EJ, Walsh SB, McCormick JA, Zietse R, Unwin RJ, Ellison DH. Pathogenesis of calcineurin inhibitor-induced hypertension. J Nephrol. 2012;25(3):269-275. doi:10.5301/jn.5000174 [PubMed 22573529]
  102. Hörl MP, Schmitz M, Ivens K, Grabensee B. Opportunistic infections after renal transplantation. Curr Opin Urol. 2002;12(2):115-123. doi:10.1097/00042307-200203000-00006 [PubMed 11859257]
  103. Horslen SP. Optimal management of the post-intestinal transplant patient. Gastroenterology. 2006;130(2 Suppl 1):S163-169. [PubMed 16473067]
  104. Hosakoppal SS, Bryce PJ. Transplant-acquired food allergy: current perspectives. J Asthma Allergy. 2017;10:307-315. doi:10.2147/JAA.S136319 [PubMed 29238209]
  105. Ierardi E, Principi M, Rendina M, et al. Oral tacrolimus (FK 506) in Crohn's disease complicated by fistulae of the perineum. J Clin Gastroenterol. 2000;30(2):200-202. [PubMed 10730928]
  106. Ishii K, Yamamoto F, Homma S, et al. Probable progressive multifocal leukoencephalopathy-immune reconstitution inflammatory syndrome with immunosuppressant dose reduction following lung transplantation: a case report and literature review. BMC Neurol. 2019;19(1):263. doi:10.1186/s12883-019-1493-1 [PubMed 31672142]
  107. Iwahori T, Takeuchi H, Matsuno N, et al. Pharmacokinetic differences between morning and evening administration of cyclosporine and tacrolimus therapy. Transplant Proc. 2005;37(4):1739-1740. [PubMed 15919450]
  108. Jahan A, Prabha R, Chaturvedi S, Mathew B, Fleming D, Agarwal I. Clinical efficacy and pharmacokinetics of tacrolimus in children with steroid-resistant nephrotic syndrome. Pediatr Nephrol. 2015;30(11):1961-1967. [PubMed 26135137]
  109. Jain A, Venkataramanan R, Fung JJ, et al, “Pregnancy After Liver Transplantation Under Tacrolimus,” Transplantation, 1997, 64(4):559-65. [PubMed 9293865]
  110. Johannesson L, Wall A, Putman JM, Zhang L, Testa G, Diaz-Garcia C. Rethinking the time interval to embryo transfer after uterus transplantation - DUETS (Dallas UtErus Transplant Study). BJOG. 2019;126(11):1305‐1309. doi:10.1111/1471-0528.15860 [PubMed 31286643]
  111. Johnson MC, So S, March JW, et al, “QT Prolongation and Torsades de Pointes After Administration of FK506,” Transplantation, 1992, 53(4):929-30. [PubMed 1373538]
  112. Jones BP, Saso S, Bracewell-Milnes T, et al. Human uterine transplantation: a review of outcomes from the first 45 cases. BJOG. 2019;126(11):1310‐1319. doi:10.1111/1471-0528.15863 [PubMed 31410987]
  113. Jumani A, Hala K, Tahir S, et al. Causes of acute thrombotic microangiopathy in patients receiving kidney transplantation. Exp Clin Transplant. 2004;2(2):268-272. [PubMed 15859940]
  114. Junna MR, Rabinstein AA. Tacrolimus induced leukoencephalopathy presenting with status epilepticus and prolonged coma. J Neurol Neurosurg Psychiatry. 2007;78(12):1410-1411. doi:10.1136/jnnp.2007.121806 [PubMed 18024699]
  115. Jusko WJ, Piekoszewski W, Klintmalm GB, et al, “Pharmacokinetics of Tacrolimus in Liver Transplant Patients,” Clin Pharmacol Ther, 1995, 57(3):281-96. [PubMed 7535213]
  116. Jusko WJ, Thomson AW, Fung J, et al. Consensus document: therapeutic monitoring of tacrolimus (FK-506). Ther Drug Monit. 1995;17(6):606-614. doi:10.1097/00007691-199512000-00011 [PubMed 8588229]
  117. Kainz A, Harabacz I, Cowlrick IS, et al, “Analysis of 100 Pregnancy Outcomes in Women Treated Systemically With Tacrolimus,” Transpl Int, 2000, 13(Suppl 1):299-300. [PubMed 11112018]
  118. Kamata Y, Iwamoto M, Minota S. Treatment of rheumatoid arthritis with tacrolimus: tacrolimus-induced urticaria. J Clin Rheumatol. 2009;15(4):213. doi:10.1097/RHU.0b013e3181a7b0ec [PubMed 19502910]
  119. Kang SY, Sohn KH, Lee JO, Kim SH, Cho SH, Chang YS. Intravenous tacrolimus and cyclosporine induced anaphylaxis: what is next? Asia Pac Allergy. 2015;5(3):181-186. doi:10.5415/apallergy.2015.5.3.181 [PubMed 26240796]
  120. Karolin A, Genitsch V, Sidler D. Calcineurin inhibitor toxicity in solid organ transplantation. Pharmacology. 2021;106(7-8):347-355. doi:10.1159/000515933 [PubMed 34130291]
  121. Katari SR, Magnone M, Shapiro R, et al. Clinical features of acute reversible tacrolimus (FK 506) nephrotoxicity in kidney transplant recipients. Clin Transplant. 1997;11(3):237-242. [PubMed 9193849]
  122. Kaufman DB, Kaplan B, Kanwar YS, et al, “The Successful Use of Tacrolimus (FK506) in a Pancreas/Kidney Transplant Recipient With Recurrent Cyclosporine-Associated Hemolytic Uremic Syndrome,” Transplantation, 1995, 59(12):1737-9. [PubMed 7541579]
  123. Kelly PA, Burckart GJ, and Venkataramanan R, “Tacrolimus: A New Immunosuppressive Agent,” Am J Health Syst Pharm, 1995, 52(14):1521-35. [PubMed 7552894]
  124. Keven K, Ozturk R, Sengul S, et al. Renal tubular acidosis after kidney transplantation--incidence, risk factors and clinical implications. Nephrol Dial Transplant. 2007;22(3):906-910. doi:10.1093/ndt/gfl714 [PubMed 17210594]
  125. Kidney Disease: Improving Global Outcomes (KDIGO) Glomerular Diseases Work Group. KDIGO 2021 clinical practice guideline for the management of glomerular diseases. Kidney Int. 2021;100(4S):S1-S276. doi:10.1016/j.kint.2021.05.021 [PubMed 34556256]
  126. Kidney Disease: Improving Global Outcomes (KDIGO) Glomerulonephritis Work Group. KDIGO Clinical Practice Guideline for Glomerulonephritis. Kidney Int. Suppl. 2012;139-274.
  127. Kidney Disease: Improving Global Outcomes (KDIGO) Transplant Work Group. KDIGO clinical practice guideline for the care of kidney transplant recipients. Am J Transplant. 2009;9(Suppl 3):S1-S155. [PubMed 19845597]
  128. Kim JS, Aviles DH, Silverstein DM, et al. Effect of age, ethnicity, and glucocorticoid use on tacrolimus pharmacokinetics in pediatric renal transplant patients. Pediatr Transplant. 2005;9(2):162-169. [PubMed 15787787]
  129. Kim BR, Shin HS, Jung YS, Rim H. A case of tacrolimus-induced supraventricular arrhythmia after kidney transplantation. Sao Paulo Med J. 2013;131(3):205-207. doi:10.1590/1516-3180.2013.1313472 [PubMed 23903271]
  130. Kishida N, Shinoda M, Itano O, et al. Increased incidence of thrombotic microangiopathy after ABO-incompatible living donor liver transplantation. Ann Transplant. 2016;21:755-764. doi:10.12659/aot.900915 [PubMed 27956735]
  131. Kishino S, Takekuma Y, Sugawara M, et al. Influence of continuous venovenous haemodiafiltration on the pharmacokinetics of tacrolimus in liver transplant recipients with small-for-size grafts. Clin Transplant. 2003;17(5):412-416. doi:10.1034/j.1399-0012.2003.00048.x [PubMed 14703922]
  132. Knorr JP, Grewal KS, Balasubramanian M, et al, “Falsely Elevated Tacrolimus Levels Caused by Immunoassay Interference Secondary to Beta-Galactosidase Antibodies in an Infected Liver Transplant Recipient,” Pharmacotherapy, 2010, 30(9):954. [PubMed 20812435]
  133. Kociszewska-Najman B, Mazanowska N, Pietrzak B, et al. Low transfer of tacrolimus and its metabolites into colostrum of graft recipient mothers. Nutrients. 2018;10(3). pii: E267. doi: 10.3390/nu10030267. [PubMed 29495430]
  134. Kolb M, Offer K, Jin Z, et al. Risk factors for subtherapeutic tacrolimus levels after conversion from continuous intravenous infusion to oral in children after allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2016;22(5):957-961. [PubMed 26880117]
  135. Krämer BK, Del Castillo D, Margreiter R, et al; European Tacrolimus versus Ciclosporin Microemulsion Renal Transplantation Study Group. Efficacy and safety of tacrolimus compared with ciclosporin A in renal transplantation: three-year observational results. Nephrol Dial Transplant. 2008;23(7):2386-2392. doi:10.1093/ndt/gfn004 [PubMed 18258740]
  136. Kulkarni M. Type 4 renal tubular acidosis in a kidney transplant recipient. Biomed J. 2016;39(1):85-86. doi:10.1016/j.bj.2015.08.008 [PubMed 27105603]
  137. Kwun WH. Tacrolimus related neurologic complication after pediatric kidney transplantation. J Korean Surg Soc. 2011;81(3):225-8. doi:10.4174/jkss.2011.81.3.225 [PubMed 22066126]
  138. Lakshmi BS, Vidya B, Reddy MHK, Kumar ACV, Ram R, Kumar VS. Sensorineural deafness following tacrolimus use. Exp Clin Transplant. 2020;18(1):110-111. doi:10.6002/ect.2017.0114 [PubMed 29969081]
  139. Lamas S. Cellular mechanisms of vascular injury mediated by calcineurin inhibitors. Kidney Int. 2005;68(2):898-907. doi:10.1111/j.1523-1755.2005.00472.x [PubMed 16014073]
  140. Langone A, Steinberg SM, Gedaly R, et al; STRATO Investigators. Switching study of kidney transplant patients with tremor to LCP-TacrO (STRATO): an open-label, multicenter, prospective phase 3b study. Clin Transplant. 2015;29(9):796-805. doi:10.1111/ctr.12581 [PubMed 26113208]
  141. Laskow DA, Vincenti F, Neylan JF, Mendez R, Matas AJ. An open-label, concentration-ranging trial of FK506 in primary kidney transplantation: a report of the United States Multicenter FK506 Kidney Transplant Group. Transplantation. 1996;62(7):900-905. doi:10.1097/00007890-199610150-00005 [PubMed 8878381]
  142. Lee A, Bridges LR, Lloyd M, et al. Epstein-Barr virus associated CNS lymphoproliferative disorder after long-term immunosuppression. Pract Neurol. 2020;20(1):83-86. doi:10.1136/practneurol-2019-002356 [PubMed 31467148]
  143. Lee WS, Lee SI, Lee MS, Kim SI, Lee SS, Yoo WH. Efficacy and safety of low-dose tacrolimus for active rheumatoid arthritis with an inadequate response to methotrexate. Korean J Intern Med. 2016;31(4):779-787. doi:10.3904/kjim.2015.066 [PubMed 26961485]
  144. Lichtenstein GR, Loftus EV, Isaacs KL, et al. ACG clinical guideline: management of Crohn's disease in adults. Am J Gastroenterol. 2018;113(4):481-517. doi: 10.1038/ajg.2018.27. [PubMed 29610508]
  145. Lin CC, King KL, Chao YW, Yang AH, Chang CF, Yang WC. Tacrolimus-associated hemolytic uremic syndrome: a case analysis. J Nephrol. 2003;16(4):580-585. [PubMed 14696762]
  146. Liu Z, Chen Y, Tao R, Xv J, Meng J, Yong X. Tacrolimus-based versus cyclosporine-based immunosuppression in hepatitis C virus-infected patients after liver transplantation: a meta-analysis and systematic review. PLoS One. 2014;9(9):e107057. doi:10.1371/journal.pone.0107057 [PubMed 25198195]
  147. Liu D, Yang Y, Kuang F, Qing S, Hu B, Yu X. Risk of infection with different immunosuppressive drugs combined with glucocorticoids for the treatment of idiopathic membranous nephropathy: A pairwise and network meta-analysis. Int Immunopharmacol. 2019;70:354-361. doi:10.1016/j.intimp.2019.03.002 [PubMed 30852290]
  148. Loar RW, Patterson MC, O'Leary PW, Driscoll DJ, Johnson JN. Posterior reversible encephalopathy syndrome and hemorrhage associated with tacrolimus in a pediatric heart transplantation recipient. Pediatr Transplant. 2013;17(2):E67-E70. doi:10.1111/petr.12039 [PubMed 23331314]
  149. López LF, Martínez CJ, Castañeda DA, Hernández AC, Pérez HC, Lozano E. Pregnancy and kidney transplantation, triple hazard? Current concepts and algorithm for approach of preconception and perinatal care of the patient with kidney transplantation. Transplant Proc. 2014;46(9):3027‐3031. doi:10.1016/j.transproceed.2014.07.013 [PubMed 25420815]
  150. Lucey MR, Terrault N, Ojo L, et al. Long-term management of the successful adult liver transplant: 2012 practice guideline by the American Association for the Study of Liver Diseases and the American Society of Transplantation. Liver Transpl. 2013;19(1):3‐26. doi:10.1002/lt.23566 [PubMed 23281277]
  151. Lué A, Martinez E, Navarro M, et al. Donor age predicts calcineurin inhibitor induced neurotoxicity after liver transplantation. Transplantation. 2019;103(8):e211-e215. doi:10.1097/TP.0000000000002750 [PubMed 30985573]
  152. MacDonald AS and Sketris IS, “Tacrolimus in Transplantation,” Am J Health Syst Pharm, 1995, 52(14):1569-71. [PubMed 7552906]
  153. Maes BD, Kuypers D, Messiaen T, et al. Posttransplantation diabetes mellitus in FK-506-treated renal transplant recipients: analysis of incidence and risk factors. Transplantation. 2001;72(10):1655-1661. doi:10.1097/00007890-200111270-00014 [PubMed 11726827]
  154. Mahadevan U, Robinson C, Bernasko N, et al. Inflammatory bowel disease in pregnancy clinical care pathway: a report from the American Gastroenterological Association IBD Parenthood Project Working Group. Gastroenterology. 2019;156(5):1508‐1524. doi:10.1053/j.gastro.2018.12.022 [PubMed 30658060]
  155. Mallat SG, Tanios BY, Itani HS, et al. CMV and BKPyV infections in renal Transplant recipients receiving an mTOR inhibitor-based regimen versus a CNI-based regimen: A systematic review and meta-analysis of randomized, controlled trials. Clin J Am Soc Nephrol. 2017;12(8):1321-1336. doi:10.2215/CJN.13221216 [PubMed 28576905]
  156. Margreiter R; European Tacrolimus vs Ciclosporin Microemulsion Renal Transplantation Study Group. Efficacy and safety of tacrolimus compared with ciclosporin microemulsion in renal transplantation: a randomised multicentre study. Lancet. 2002;359(9308):741-746. doi:10.1016/S0140-6736(02)07875-3 [PubMed 11888584]
  157. Maruyama K, Chujo D. Tacrolimus-induced diabetic ketoacidosis with subsequent rapid recovery of endogenous insulin secretion after cessation of tacrolimus: a case report with review of literature. Medicine (Baltimore). 2019;98(36):e16992. doi:10.1097/MD.0000000000016992 [PubMed 31490380]
  158. Mavroudi A, Imvrios G, Xinias I, Cassimos D, Vagiotas L, Giouleme O. Tacrolimus immunosuppression causes drug-induced hypersensitivity reactions in liver-transplant patients: A 3-case report. Transplant Proc. 2021;53(5):1731-1735. doi:10.1016/j.transproceed.2021.03.031 [PubMed 33962771]
  159. McDiarmid SV, Colonna JO, Shaked A, et al, “Differences in Oral FK506 Dose Requirements Between Adults and Pediatric Liver Transplant Patients,” Transplantation, 1993, 55(6):1328-32. [PubMed 7685933]
  160. Melzer N, Ruck T, Fuhr P, et al. Clinical features, pathogenesis, and treatment of myasthenia gravis: a supplement to the guidelines of the German Neurological Society. J Neurol. 2016;263(8):1473-1494. doi: 10.1007/s00415-016-8045-z. [PubMed 26886206]
  161. Méndez A, Berastegui C, López-Meseguer M, et al. Pharmacokinetic study of conversion from tacrolimus twice-daily to tacrolimus once-daily in stable lung transplantation. Transplantation. 2014;97(3):358-362. [PubMed 24492423]
  162. Menegaux F, Keeffe EB, Andrews BT, et al, “Neurological Complications of Liver Transplantation in Adult Versus Pediatric Patients,” Transplantation, 1994, 58(4):447-50. [PubMed 8073514]
  163. Messina C, Faraci M, de Fazio V, et al, “Paediatric Working Party. Prevention and Treatment of Acute GvHD,” Bone Marrow Transplant, 2008, 41(Suppl 2):65-70. [PubMed 18545247]
  164. Meyer KC, Raghu G, Verleden GM, et al; ISHLT/ATS/ERS BOS Task Force Committee. An international ISHLT/ATS/ERS clinical practice guideline: diagnosis and management of bronchiolitis obliterans syndrome. Eur Respir J. 2014;44(6):1479-1503. doi:10.1183/09031936.00107514 [PubMed 25359357]
  165. Min DI, Chen HY, Lee MK, Ashton K, Martin MF. Time-dependent disposition of tacrolimus and its effect on endothelin-1 in liver allograft recipients. Pharmacotherapy. 1997;17(3):457-463. [PubMed 9165550]
  166. Moes AD, Hesselink DA, Zietse R, van Schaik RH, van Gelder T, Hoorn EJ. Calcineurin inhibitors and hypertension: a role for pharmacogenetics? Pharmacogenomics. 2014;15(9):1243-1251. doi:10.2217/pgs.14.87 [PubMed 25141899]
  167. Montero N, Pascual J. Immunosuppression and post-transplant hyperglycemia. Curr Diabetes Rev. 2015;11(3):144-154. doi:10.2174/1573399811666150331160846 [PubMed 25824238]
  168. Montini G, Ujka F, Varagnolo C, et al. The pharmacokinetics and immunosuppressive response of tacrolimus in paediatric renal transplant recipients. Pediatr Nephrol. 2006;21(5):719-724. [PubMed 16550361]
  169. Naesens M, Kuypers DR, Sarwal M. Calcineurin inhibitor nephrotoxicity. Clin J Am Soc Nephrol. 2009;4(2):481-508. doi:10.2215/CJN.04800908 [PubMed 19218475]
  170. Nanda T, Rasool N, Bearelly S. Tacrolimus induced optic neuropathy in post-lung transplant patients: a series of 3 patients. Am J Ophthalmol Case Rep. 2021;22:101056. doi:10.1016/j.ajoc.2021.101056 [PubMed 33778180]
  171. Nankivell BJ, Borrows RJ, Fung CL, O'Connell PJ, Chapman JR, Allen RD. Calcineurin inhibitor nephrotoxicity: longitudinal assessment by protocol histology. Transplantation. 2004;78(4):557-565. doi:10.1097/01.tp.0000128636.70499.6e [PubMed 15446315]
  172. Nankivell BJ, PʼNg CH, OʼConnell PJ, Chapman JR. Calcineurin inhibitor nephrotoxicity through the lens of longitudinal histology: Comparison of cyclosporine and tacrolimus eras. Transplantation. 2016;100(8):1723-1731. doi:10.1097/TP.0000000000001243 [PubMed 27306529]
  173. Nasiri-Toosi Z, Dashti-Khavidaki S, Nasiri-Toosi M, et al. Clinical pharmacokinetics of oral versus sublingual administration of tacrolimus in adult liver transplant recipients. Exp Clin Transplant. 2012;10(6):586-591. [PubMed 22770208]
  174. Natazuka T, Ogawa R, Kizaki T, et al, “Immunosuppressive Drugs and Hypertrophic Cardiomyopathy,” Lancet, 1995, 345(8965):1644. [PubMed 7540241]
  175. National Institute for Occupational Safety and Health (NIOSH). NIOSH List of Antineoplastic and Other Hazardous Drugs in Healthcare Settings 2012. Available at http://www.cdc.gov/niosh/docs/2012-150/pdfs/2012-150.pdf. Accessed January 21, 2013.
  176. Nelson J, Alvey N, Bowman L, et al. Consensus recommendations for use of maintenance immunosuppression in solid organ transplantation: endorsed by the American College of Clinical Pharmacy, American Society of Transplantation, and the International Society for Heart and Lung Transplantation. Pharmacotherapy. 2022;42(8):599-633. doi:10.1002/phar.2716 [PubMed 36032031]
  177. Nemecek BD, Hammond DA, eds. Demystifying Drug Dosing in Renal Dysfunction. American Society of Health-System Pharmacists; 2019.
  178. Nicolai S and Bunyavanich S. Hypersensitivity reaction to intravenous but not oral tacrolimus. Transplantation. 2012;94(9):e61-63. doi:10.1097/TP.0b013e31826e5995 [PubMed 23128975]
  179. Noda K, Ukichi T, Furuya K, et al. Tacrolimus-induced hypertrophic cardiomyopathy in a patient with dermatomyositis. Rheumatology (Oxford). 2017;56(11):2037-2038. doi:10.1093/rheumatology/kex310 [PubMed 28977571]
  180. Notaro E, Brown L, Albers E, et al. Dose conversion factor between cyclosporine and tacrolimus in pediatric heart transplant recipients. J Heart Lung Transplant. 2014;33(7):766-768. doi: 10.1016/j.healun.2014.04.004. [PubMed 24854566]
  181. Nwaba A, MacQuillan G, Adams LA, et al. Tacrolimus-induced thrombotic microangiopathy in orthotopic liver transplant patients: case series of four patients. Intern Med J. 2013;43(3):328-333. doi:10.1111/imj.12048 [PubMed 23441660]
  182. Obayashi N, Suzuki M, Yokokura T, et al. Management of tacrolimus-associated food allergy after liver transplantation. Pediatr Int. 2015;57(6):1205-1207. doi:10.1111/ped.12721 [PubMed 26541649]
  183. Offer K, Kolb M, Jin Z, et al. Efficacy of tacrolimus/mycophenolate mofetil as acute graft-versus-host disease prophylaxis and the impact of subtherapeutic tacrolimus levels in children after matched sibling donor allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2015;21(3):496-502. [PubMed 25536217]
  184. Opelz G, Döhler B. Lymphomas after solid organ transplantation: a collaborative transplant study report. Am J Transplant. 2004;4(2):222-230. doi:10.1046/j.1600-6143.2003.00325.x [PubMed 14974943]
  185. Ozdemir O, Arrey-Mensah A, Sorensen RU. Development of multiple food allergies in children taking tacrolimus after heart and liver transplantation. Pediatr Transplant. 2006;10(3):380-383. doi:10.1111/j.1399-3046.2005.00474.x [PubMed 16677366]
  186. Park SI, Felipe CR, Pinheiro-Machado PG, Garcia R, Tedesco-Silva H Jr, Medina-Pestana JO. Circadian and time-dependent variability in tacrolimus pharmacokinetics. Fundam Clin Pharmacol. 2007;21(2):191-197. [PubMed 17391292]
  187. Parissis H, Gould K, Dark J. Dangerous drug interactions leading to hemolytic uremic syndrome following lung transplantation. J Cardiothorac Surg. 2010;5:70. doi:10.1186/1749-8090-5-70 [PubMed 20813025]
  188. Patel N, Perez C, Taber DJ, Kalbavi V, Gonzales H, Rohan V. Safety and efficacy of perioperative sublingual tacrolimus in pancreas transplant compared with oral tacrolimus. Exp Clin Transplant. 2021;19(6):592-595. doi:10.6002/ect.2020.0391 [PubMed 33952179]
  189. Peddi VR, Wiseman A, Chavin K, Slakey D. Review of combination therapy with mTOR inhibitors and tacrolimus minimization after transplantation. Transplant Rev (Orlando). 2013;27(4):97-107. [PubMed 23932018]
  190. Penninga L, Penninga EI, Moller CH, Iversen M, Steinbruchel DA, Gluud C. Tacrolimus versus cyclosporin as primary immunosuppression for lung transplant recipients. Cochrane Database Syst Rev. 2013;5:Cd008817. doi:10.1002/14651858.CD008817.pub [PubMed 23728681]
  191. Pennington CA, Park JM. Sublingual tacrolimus as an alternative to oral administration for solid organ transplant recipients. Am J Health Syst Pharm. 2015;72(4):277-284. [PubMed 25631834]
  192. Perni UC, Wang EY, Gregg AR. Antepartum care of the uterus transplant patient: the experience of 3 successful US centers. Clin Obstet Gynecol. 2022;65(1):84-91. doi: 10.1097/GRF.0000000000000682 [PubMed 35045029]
  193. Pirsch JD, Miller J, Deierhoi MH, Vincenti F, Filo RS. A comparison of tacrolimus (FK506) and cyclosporine for immunosuppression after cadaveric renal transplantation. FK506 Kidney Transplant Study Group. Transplantation. 1997;63(7):977-983. doi:10.1097/00007890-199704150-00013 [PubMed 9112351]
  194. Podesser BK, Rinaldi M, Yona NA, et al, “Comparison of Low and High Initial Tacrolimus Dosing in Primary Heart Transplant Recipients: A Prospective European Multicenter Study,” Transplantation, 2005, 79(1):65-71. [PubMed 15714171]
  195. Pollock-Barziv SM, Dipchand AI. McCrindle BW, et al. Randomized clinical trial of tacrolimus- vs cyclosporine-based immunosuppression in pediatric heart transplantation: preliminary results at 15-month follow-up. J Heart Lung Transplant. 2005;24(2):190-194. [PubMed 15701436]
  196. Ponseti JM, Gamez J, Azem J, López-Cano M, Vilallonga R, Armengol M. Tacrolimus for myasthenia gravis: a clinical study of 212 patients. Ann N Y Acad Sci. 2008;1132:254-263. [PubMed 18096852]
  197. Porrini EL, Díaz JM, Moreso F, et al. Clinical evolution of post-transplant diabetes mellitus. Nephrol Dial Transplant. 2016;31(3):495-505. doi:10.1093/ndt/gfv368 [PubMed 26538615]
  198. Prograf (tacrolimus) [prescribing information]. Northbrook, IL: Astellas Pharma US Inc; November 2022.
  199. Prograf (tacrolimus) [product monograph]. Markham, Ontario, Canada: Astellas Pharma Canada Inc; December 2022.
  200. Przepiorka D, Devine S, Fay J, et al, “Practical Considerations in the Use of Tacrolimus for Allogeneic Marrow Transplantation,” Bone Marrow Transplant, 1999, 24(10):1053-6. [PubMed 10578154]
  201. Przepiorka D, Suzuki J, Ippoliti C, et al, “Blood Tacrolimus Concentration Unchanged by Plasmapheresis,” Am J Hosp Pharm, 1994, 51(13):1708. [PubMed 7524318]
  202. Randhawa PS, Tsamandas AC, Magnone M, et al. Microvascular changes in renal allografts associated with FK506 (Tacrolimus) therapy. Am J Surg Pathol. 1996;20(3):306-312. doi:10.1097/00000478-199603000-00007 [PubMed 8772784]
  203. Reams BD, Palmer SM. Sublingual tacrolimus for immunosuppression in lung transplantation: a potentially important therapeutic option in cystic fibrosis. Am J Respir Med. 2002;1(2):91-98. [PubMed 14720063]
  204. Refer to manufacturer's labeling.
  205. Remuzzi G, Cravedi P, Perna A, et al; Dual Kidney Transplant Group. Long-term outcome of renal transplantation from older donors. N Engl J Med. 2006;354(4):343-352. doi:10.1056/NEJMoa052891 [PubMed 16436766]
  206. Reyes J, Gayowski T, Fung J, Todo S, Alessiani M, Starzl TE. Expressive dysphasia possibly related to FK506 in two liver transplant recipients. Transplantation. 1990;50(6):1043-1045. doi:10.1097/00007890-199012000-00028 [PubMed 1701571]
  207. Richards EG, Farrell RM, Ricci S, et al. Uterus transplantation: state of the art in 2021. J Assist Reprod Genet. 2021;38(9):2251-2259. doi:10.1007/s10815-021-02245-7 [PubMed 34057644]
  208. Ridruejo E, Díaz C, Michel MD, et al. Short and long term outcome of kidney transplanted patients with chronic viral hepatitis B and C. Ann Hepatol. 2010;9(3):271-277. [PubMed 20720267]
  209. Riley L, Mudd L, Baize T, Herzig R. Cross-sensitivity reaction between tacrolimus and macrolide antibiotics. Bone Marrow Transplant. 2000;25(8):907-908. doi:10.1038/sj.bmt.1702223 [PubMed 10808214]
  210. Rubik J, Debray D, Kelly D, et al. Efficacy and safety of prolonged-release tacrolimus in stable pediatric allograft recipients converted from immediate-release tacrolimus - a phase 2, open-label, single-arm, one-way crossover study. Transpl Int. 2019;32(11):1182-1193. doi:10.1111/tri.13479 [PubMed 31325368]
  211. Samer A, Almehmadi F, Krimly A, Alrajhi A. Tacrolimus-induced diffuse coronary artery spasm. Cureus. 2022;14(6):e25748. doi: 10.7759/cureus.25748 [PubMed 35812568]
  212. Sammaritano LR, Bermas BL, Chakravarty EE, et al. 2020 American College of Rheumatology guideline for the management of reproductive health in rheumatic and musculoskeletal diseases. Arthritis Rheumatol. 2020;72(4):529‐556. doi:10.1002/art.41191
  213. Samonakis DN, Germani G, Burroughs AK. Immunosuppression and HCV recurrence after liver transplantation. J Hepatol. 2012;56(4):973-983. doi:10.1016/j.jhep.2011.06.031 [PubMed 21963518]
  214. Sandborn WJ, Present DH, Isaacs KL, et al. Tacrolimus for the treatment of fistulas in patients with Crohn's disease: a randomized, placebo-controlled trial. Gastroenterology. 2003;125(2):380-388. [PubMed 12891539]
  215. Sanders DB, Wolfe GI, Benatar M, et al. International consensus guidance for management of myasthenia gravis: executive summary. Neurology. 2016;87(4):419-425. doi:10.1212/WNL.0000000000002790 [PubMed 27358333]
  216. Saudek F, Malaise J, Boucek P, Adamec M; Euro-SPK Study Group. Efficacy and safety of tacrolimus compared with cyclosporin microemulsion in primary SPK transplantation: 3-year results of the Euro-SPK 001 trial. Nephrol Dial Transplant. 2005;20(suppl 2):ii3-10, ii62. doi:10.1093/ndt/gfh1076 [PubMed 15814547]
  217. Scherrer M, Araujo MG, Farah K. Tacrolimus-induced symmetric drug-related intertriginous and flexural exanthema (SDRIFE). Contact Dermatitis. 2018;78(6):414-416. doi:10.1111/cod.12954 [PubMed 29722025]
  218. Sekiguchi Y, Shimada A, Imai H, et al. Epstein-Barr virus-negative, CD5-positive diffuse large B-cell lymphoma developing after treatment with oral tacrolimus for mixed connective tissue disease : a case report and review of the literature. J Clin Exp Hematop. 2012;52(3):211-218. doi:10.3960/jslrt.52.211. [PubMed 23269082]
  219. Shah S, Ojha R, Chamlagain R, et al. Therapeutic drug level of tacrolimus causing intracranial hemorrhage in a patient with renal transplant. Clin Case Rep. 2022;10(4):e05788. doi:10.1002/ccr3.5788 [PubMed 35498342]
  220. Sharif A, Hecking M, de Vries AP, et al. Proceedings from an international consensus meeting on posttransplantation diabetes mellitus: recommendations and future directions. Am J Transplant. 2014;14(9):1992-2000. doi: 10.1111/ajt.12850 [PubMed 25307034]
  221. Shaw DW, Eichenfield LF, Shainhouse T, Maibach HI. Allergic contact dermatitis from tacrolimus. J Am Acad Dermatol. 2004;50(6):962-965. doi:10.1016/j.jaad.2003.09.013 [PubMed 15153904]
  222. Shen T, Feng XW, Geng L, Zheng SS. Reversible sinusoidal obstruction syndrome associated with tacrolimus following liver transplantation. World J Gastroenterol. 2015;21(20):6422-6426. doi:10.3748/wjg.v21.i20.6422 [PubMed 26034381]
  223. Singh JA, Hossain A, Kotb A, Wells G. Risk of serious infections with immunosuppressive drugs and glucocorticoids for lupus nephritis: a systematic review and network meta-analysis. BMC Med. 2016;14(1):137. doi:10.1186/s12916-016-0673-8 [PubMed 27623861]
  224. Singh RK, Humlicek T, Jeewa A, Fester K. Pediatric cardiac intensive care society 2014 consensus statement: pharmacotherapies in cardiac critical care immune therapy. Pediatr Crit Care Med. 2016;17(3 Suppl 1):S69-S76. [PubMed 26945331]
  225. Skeens M, Pai V, Garee A, et al. Twice daily i.v. bolus tacrolimus infusion for GVHD prophylaxis in children undergoing stem cell transplantation. Bone Marrow Transplant. 2012;47(11):1415-1418. [PubMed 22484323]
  226. Smith JM, Rudser K, Gillen D, et al. Risk of lymphoma after renal transplantation varies with time: an analysis of the United States Renal Data System. Transplantation. 2006;81(2):175-180. doi:10.1097/01.tp.0000188687.18972.a8 [PubMed 16436959]
  227. Snanoudj R, Royal V, Elie C, et al. Specificity of histological markers of long-term CNI nephrotoxicity in kidney-transplant recipients under low-dose cyclosporine therapy. Am J Transplant. 2011;11(12):2635-2646. doi:10.1111/j.1600-6143.2011.03718.x [PubMed 21883915]
  228. Snell GI, Westall GP, Paraskeva MA. Immunosuppression and allograft rejection following lung transplantation: evidence to date. Drugs. 2013a;73(16):1793-1813. [PubMed 24142409]
  229. Snell GI, Ivulich S, Mitchell L, Westall GP, Levvey BJ. Evolution to twice daily bolus intravenous tacrolimus: optimizing efficacy and safety of calcineurin inhibitor delivery early post lung transplant. Ann Transplant. 2013b;18:399-407. doi:10.12659/AOT.883993 [PubMed 23921892]
  230. Sofue T, Inui M, Kiyomoto H, et al. Excess fluid distribution affects tacrolimus absorption in peritoneal dialysis patients. Clin Exp Nephrol. 2013;17(5):743-749. doi:10.1007/s10157-012-0764-6 [PubMed 23269423]
  231. Sommerer C, Suwelack B, Draqun D, et al. Design and rationale of the ATHENA study--A 12-month, multicentre, prospective study evaluating the outcomes of a de novo everolimus-based regimen in combination with reduced cyclosporine or tacrolimus versus a standard regimen in kidney transplant patients: study protocol for a randomised controlled trial. Trials. 2016;17:92. [PubMed 26888217]
  232. Soubhia RM, Mendes GE, Mendonça FZ, Baptista MA, Cipullo JP, Burdmann EA. Tacrolimus and nonsteroidal anti-inflammatory drugs: an association to be avoided. Am J Nephrol. 2005;25(4):327-334. doi:10.1159/000086569 [PubMed 15976495]
  233. Spindel J, Heckroth M, Marsano L. Antibody-negative autoimmune encephalitis as a complication of long-term immune-suppression for liver transplantation. BMJ Case Rep. 2020;13(9):e235777. doi:10.1136/bcr-2020-235777 [PubMed 32933909]
  234. Staatz CE, Tett SE. Clinical pharmacokinetics and pharmacodynamics of tacrolimus in solid organ transplantation. Clin Pharmacokinet. 2004;43(10):623-653. doi:10.2165/00003088-200443100-00001 [PubMed 15244495]
  235. Staatz CE, Tett SE. Clinical pharmacokinetics of once-daily tacrolimus in solid-organ transplant patients. Clin Pharmacokinet. 2015;54(10):993-1025. doi:10.1007/s40262-015-0282-2 [PubMed 26038096]
  236. Starzl TE, Fung J, Jordan M, et al, “Kidney Transplantation Under FK506,” JAMA, 1990, 264(1):63-7. [PubMed 1693970]
  237. Steg RE, Kessinger A, Wszolek ZK. Cortical blindness and seizures in a patient receiving FK506 after bone marrow transplantation. Bone Marrow Transplant. 1999;23(9):959-962. doi:10.1038/sj.bmt.1701732 [PubMed 10338054]
  238. Suwelack B, Malyar V, Koch M, Sester M, Sommerer C. The influence of immunosuppressive agents on BK virus risk following kidney transplantation, and implications for choice of regimen. Transplant Rev (Orlando). 2012;26(3):201-211. doi:10.1016/j.trre.2011.05.002 [PubMed 21940156]
  239. Swenson JM, Fricker FJ, Armitage JM. Immunosuppression switch in pediatric heart transplant recipients: cyclosporine to FK 506. J Am Coll Cardiol. 1995;25(5):1183-1188. [PubMed 7534779]
  240. Swerdlow SH, Campo E, Pileri SA, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood. 2016;127(20):2375-2390. doi:10.1182/blood-2016-01-643569 [PubMed 26980727]
  241. Taghvaye-Masoumi H, Sadeghi K, Hadjibabaie M. Successful switch to oral tacrolimus in a patient with hypersensitivity reaction to parenteral vitamin K, cyclosporine, and tacrolimus: A case report. J Oncol Pharm Pract. 2020;26(4):986-988. doi:10.1177/1078155219869442 [PubMed 31446869]
  242. Taormina D, Abdallah HY, Venkataramanan R, et al, "Stability and Sorption of FK 506 in 5% Dextrose Injection and 0.9% Sodium Chloride Injection in Glass, Polyvinyl Chloride, and Polyolefin Containers," Am J Hosp Pharm, 1992, 49(1):119-22. [PubMed 1373936]
  243. Taylor DO, Barr ML, Meiser BM, Pham SM, Mentzer RM, Gass AL. Suggested guidelines for the use of tacrolimus in cardiac transplant recipients. J Heart Lung Transplant. 2001;20(7):734-738. [PubMed 11448799]
  244. Tiwari V, Gupta A, Gothwal C, et al. Tacrolimus-induced epidermoid cysts in the renal transplant patient. Indian J Nephrol. 2021;31(6):571-573. doi:10.4103/ijn.IJN_318_19 [PubMed 35068767]
  245. Trautmann A, Vivarelli M, Samuel S, et al. IPNA clinical practice recommendations for the diagnosis and management of children with steroid-resistant nephrotic syndrome. Pediatr Nephrol. 2020;35(8):1529-1561. doi:10.1007/s00467-020-04519-1 [PubMed 32382828]
  246. Treede H, Glanville AR, Klepetko W, et al; European and Australian Investigators in Lung Transplantation. Tacrolimus and cyclosporine have differential effects on the risk of development of bronchiolitis obliterans syndrome: results of a prospective, randomized international trial in lung transplantation. J Heart Lung Transplant. 2012;31(8):797-804. doi:10.1016/j.healun.2012.03.008 [PubMed 22554673]
  247. Treede H, Klepetko W, Reichenspurner H, et al; Munich and Vienna Lung Transplant Group. Tacrolimus versus cyclosporine after lung transplantation: a prospective, open, randomized two-center trial comparing two different immunosuppressive protocols. J Heart Lung Transplant. 2001;20(5):511-517. doi:10.1016/s1053-2498(01)00244-3 [PubMed 11343977]
  248. Tremblay S, Nigro V, Weinberg J, Woodle ES, Alloway RR. A steady-state head-to-head pharmacokinetic comparison of all FK-506 (tacrolimus) formulations (ASTCOFF): an open-label, prospective, randomized, two-arm, three-period crossover study. Am J Transplant. 2017;17(2):434-442. [PubMed 27340950]
  249. Trofe-Clark J, Doshi M, Fadugba O, Lim MA. Evaluation of allergy to tacrolimus in kidney transplant candidates and recipients with a history of macrolide antibiotic allergy. Am J Transplant. 2018;18(7):1831-1832. doi:10.1111/ajt.14879 [PubMed 29673079]
  250. Trompeter R, Filler G, Webb NJ, et al. Randomized trial of tacrolimus versus cyclosporin microemulsion in renal transplantation. Pediatr Nephrol. 2002;17(3):141-149. [PubMed 11956848]
  251. Tsapepas D, Saal S, Benkert S, et al. Sublingual tacrolimus: a pharmacokinetic evaluation pilot study. Pharmacotherapy. 2013;33(1):31-37. [PubMed 23307542]
  252. Uberti JP, Cronin S, Ratanatharathorn V. Optimum use of tacrolimus in the prophylaxis of graft versus host disease. BioDrugs. 1999;11(5):343-358. [PubMed 18031144]
  253. US Department of Health and Human Services; Centers for Disease Control and Prevention; National Institute for Occupational Safety and Health. NIOSH list of antineoplastic and other hazardous drugs in healthcare settings 2016. https://www.cdc.gov/niosh/docs/2016-161/default.html. Updated September 2016. Accessed October 5, 2016.
  254. US Multicenter FK506 Liver Study Group. A comparison of tacrolimus (FK 506) and cyclosporine for immunosuppression in liver transplantation. N Engl J Med. 1994 Oct 27;331(17):1110-1115. doi:10.1056/NEJM199410273311702 [PubMed 7523946]
  255. Van Hooff J, Van der Walt I, Kallmeyer J, et al. Pharmacokinetics in stable kidney transplant recipients after conversion from twice-daily to once-daily tacrolimus formulations. Ther Drug Monit. 2012;34(1):46-52. [PubMed 22249344]
  256. Vasudevan A, Thergaonkar R, Mantan M, et al. Consensus guidelines on management of steroid-resistant nephrotic syndrome. Indian Pediatr. 2021;58(7):650-666. [PubMed 33408286]
  257. Vearrier D, Simpson SE, Greenberg MI. Mutism and persistent dysarthria due to tacrolimus-based immunosuppression following allogeneic liver transplantation. Am J Ther. 2011;18(6):e274-e276. doi:10.1097/MJT.0b013e3181debc6f [PubMed 20535006]
  258. Velleca A, Shullo MA, Dhital K, et al. The International Society for Heart and Lung Transplantation (ISHLT) guidelines for the care of heart transplant recipients. J Heart Lung Transplant. 2023;42(5):e1-e141. doi:10.1016/j.healun.2022.10.015 [PubMed 37080658]
  259. Velvet AJJ, Bhutani S, Papachristos S, et al. A single-center experience of post-transplant lymphomas involving the central nervous system with a review of current literature. Oncotarget. 2019;10(4):437-448. doi:10.18632/oncotarget.26522 [PubMed 30728897]
  260. Vincenti F, Friman S, Scheuermann E, et al; DIRECT (Diabetes Incidence after Renal Transplantation: Neoral C Monitoring Versus Tacrolimus) Investigators. Results of an international, randomized trial comparing glucose metabolism disorders and outcome with cyclosporine versus tacrolimus. Am J Transplant. 2007;7(6):1506-1514. doi:10.1111/j.1600-6143.2007.01749.x [PubMed 17359512]
  261. Watanabe S, Sakamoto R, Yamamoto H, et al. Pediatric pure red cell aplasia caused by tacrolimus after living-donor liver transplant. Exp Clin Transplant. 2020;18(7):838-841. doi:10.6002/ect.2019.0075 [PubMed 31724928]
  262. Watkins KD, Boettger RF, Hanger KM, et al. Use of sublingual tacrolimus in lung transplant recipients. J Heart Lung Transplant. 2012;31(2):127-132. [PubMed 22177691]
  263. Webb NJA, Baumann U, Camino M, Frauca E, Undre N; OPTION Study Group. Pharmacokinetics of tacrolimus granules in pediatric de novo liver, kidney, and heart transplantation: the OPTION study. Pediatr Transplant. 2019;23(1):e13328. doi:10.1111/petr.13328 [PubMed 30665258]
  264. Weir MR, Burgess ED, Cooper JE, et al. Assessment and management of hypertension in transplant patients. J Am Soc Nephrol. 2015;26(6):1248-1260. doi:10.1681/ASN.2014080834 [PubMed 25653099]
  265. Wilson NK, Schulz P, Wall A, et al. Immunosuppression in uterus transplantation: experience from the Dallas Uterus transplant study. Transplantation. 2023;107(3):729-736. doi:10.1097/TP.0000000000004437 [PubMed 36445981]
  266. Winkel E, DiSesa VJ, and Costanzo MR, “Advances in Heart Transplantation,” Dis Mon, 1999, 45(3):62-87. [PubMed 10080968]
  267. Witt CA, Hachem RR. Immunosuppression: what's standard and what's new? Semin Respir Crit Care Med. 2013;34(3):405-413. [PubMed 23821513]
  268. Wong TY, Chan PK, Leung CB, Szeto CC, Tam JS, Li PK. Parvovirus B19 infection causing red cell aplasia in renal transplantation on tacrolimus. Am J Kidney Dis. 1999;34(6):1132-1136. doi:10.1016/S0272-6386(99)70021-1 [PubMed 10585325]
  269. Wu Z, Xu Q, Qiu X, Jiao Z, Zhang M, Zhong M. FOXP3 rs3761548 polymorphism is associated with tacrolimus-induced acute nephrotoxicity in renal transplant patients. Eur J Clin Pharmacol. 2017;73(1):39-47. doi:10.1007/s00228-016-2140-z [PubMed 27747372]
  270. Xu J, Xu L, Wei X, Li X, Cai M. A case report: acute pancreatitis associated with tacrolimus in kidney transplantation. BMC Nephrol. 2019;20(1):209. doi:10.1186/s12882-019-1395-x [PubMed 31174507]
  271. Yamamoto R, Saito M, Abe K, et al. Anaphylaxis induced by intravenous tacrolimus administration during kidney transplant surgery: A case report. Transplant Proc. 2021;53(4):1292-1294. doi:10.1016/j.transproceed.2021.02.010 [PubMed 33714607]
  272. Yanik G, Levine JE, Ratanatharathorn V, et al, “Tacrolimus (FK506) and Methotrexate as Prophylaxis for Acute Graft-Versus-Host Disease in Pediatric Allogeneic Stem Cell Transplantation,” Bone Marrow Transplant, 2000, 26(2):161-7. [PubMed 10918426]
  273. Yocum DE, Furst DE, Kaine JL, et al. Efficacy and safety of tacrolimus in patients with rheumatoid arthritis: a double-blind trial. Arthritis Rheum. 2003;48(12):3328-3337. doi:10.1002/art.11363 [PubMed 14673984]
  274. Yoshikawa H, Kiuchi T, Saida T, Takamori M. Randomised, double-blind, placebo-controlled study of tacrolimus in myasthenia gravis. J Neurol Neurosurg Psychiatry. 2011;82(9):970-977. doi:10.1136/jnnp-2011-300148 [PubMed 21784757]
  275. Yucel OK, Alemdar MS, Akkaya B, Kocak H, Suleymanlar G, Yilmaz VT. Tacrolimus-associated pure red cell aplasia in a patient with renal transplant. Exp Clin Transplant. 2020 Nov 27. doi:10.6002/ect.2020.0290 [PubMed 33272158]
  276. Zheng S, Easterling TR, Hays K, et al. Tacrolimus placental transfer at delivery and neonatal exposure through breast milk. Br J Clin Pharmacol. 2013;76(6):988‐996. doi:10.1111/bcp.12122 [PubMed 23528073]
  277. Zheng S, Easterling TR, Umans JG, et al, "Pharmacokinetics of Tacrolimus During Pregnancy," Ther Drug Monit, 2012, 34(6):660-70. [PubMed 23007747]
  278. Zolota A, Miserlis G, Solonaki F, et al. New-onset diabetes after transplantation: comparison between a cyclosporine-based and a tacrolimus-based immunosuppressive regimen. Transplant Proc. 2018;50(10):3386-3391. doi:10.1016/j.transproceed.2018.08.037 [PubMed 30577210]
  279. Zuckermann A, Reichenspurner H, Birsan T, et al. Cyclosporine A versus tacrolimus in combination with mycophenolate mofetil and steroids as primary immunosuppression after lung transplantation: one-year results of a 2-center prospective randomized trial. J Thorac Cardiovasc Surg. 2003;125(4):891-900. doi:10.1067/mtc.2003.71 [PubMed 12698153]
Topic 9920 Version 572.0

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟