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

Cyclosporine (ciclosporin) (systemic): Drug information

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

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

Only health care providers experienced in the management of systemic immunosuppressive therapy for the indicated disease should prescribe cyclosporine. At doses used in solid organ transplantation, only health care providers experienced in immunosuppressive therapy and management of organ transplant recipients should prescribe cyclosporine. Patients receiving cyclosporine should be managed in facilities equipped and staffed with adequate laboratory and supportive medical resources. The health care provider responsible for maintenance therapy should have complete information requisite for the follow-up of the patient.

Immunosuppression:

Cyclosporine may increase the susceptibility to infection and the development of neoplasia. In kidney, liver, and heart transplant patients, Gengraf and Neoral may be administered with other immunosuppressive agents. Increased susceptibility to infection and the possible development of lymphoma and other neoplasms may result from the increase in the degree of immunosuppression in transplant patients.

Bioavailability:

The absorption of Sandimmune capsules and oral solution during long-term administration was found to be erratic. It is recommended that patients taking Sandimmune capsules or oral solution over a period of time be monitored at repeated intervals for cyclosporine blood levels and that subsequent dose adjustments be made to avoid toxicity from high levels and possible organ rejection from low absorption of cyclosporine. This is of special importance in liver transplants. Numerous assays are being developed to measure blood levels of cyclosporine.

Sandimmune capsules and oral solution have decreased bioavailability in comparison with Gengraf and Neoral capsules and Gengraf and Neoral oral solution. Gengraf and Neoral have increased bioavailability compared to Sandimmune capsules and oral solution and are not bioequivalent to Sandimmune and cannot be used interchangeably without the supervision of a health care provider. For a given trough concentration, cyclosporine exposure will be greater with Neoral and Gengraf than with Sandimmune. If a patient who is receiving exceptionally high doses of Sandimmune is converted to Neoral or Gengraf, particular caution should be exercised. Cyclosporine blood concentrations should be monitored in transplant and rheumatoid arthritis (RA) patients taking Gengraf and Neoral to avoid toxicity due to high concentrations. Dose adjustments should be made in transplant patients to minimize possible organ rejection due to low concentrations. Comparison of blood concentrations in the published literature with blood concentrations obtained using current assays must be done with detailed knowledge of the assay methods employed.

Psoriasis (Neoral, Gengraf):

Psoriasis patients previously treated with psoralens plus ultraviolet A (PUVA) and, to a lesser extent, methotrexate or other immunosuppressive agents, ultraviolet B (UVB), coal tar, or radiation therapy, are at an increased risk of developing skin malignancies when taking cyclosporine. Also see boxed warnings above.

Hypertension/nephrotoxicity:

Cyclosporine, in recommended doses, can cause systemic hypertension and nephrotoxicity. The risk increases with increasing dose and duration of cyclosporine therapy. Renal dysfunction, including structural kidney damage, is a potential consequence of cyclosporine and, therefore, renal function must be monitored during therapy.

Brand Names: US
  • Gengraf;
  • Neoral;
  • SandIMMUNE
Brand Names: Canada
  • Neoral;
  • PMS-Cyclosporine;
  • SandIMMUNE IV;
  • SANDOZ CycloSPORINE
Pharmacologic Category
  • Calcineurin Inhibitor;
  • Immunosuppressant Agent
Dosing: Adult

Neoral/Gengraf (cyclosporine modified) and Sandimmune (cyclosporine non-modified) are not bioequivalent and cannot be used interchangeably. Use caution when selecting, dispensing, and administrating cyclosporine products; in general, cyclosporine (modified) is more commonly used clinically.

Aplastic anemia

Aplastic anemia (off-label use):

Severe: Oral: 5 mg/kg/day in 2 divided doses (in combination with antithymocyte globulin [equine] and eltrombopag) for a minimum of 12 months; begin tapering off cyclosporine over the next 12 months to discontinue by month 24 (Ref) or 10 mg/kg in 2 divided doses (every 12 hours) (in combination with antithymocyte globulin [equine]) for 6 months then tapered over 18 months; target cyclosporine trough levels were maintained between 200 to 400 ng/mL (Ref).

Non-severe: Oral: 2.5 mg/kg every 12 hours (with antithymocyte globulin [equine]) for at least 6 months or until blood cell count plateaus, followed by gradual taper; target cyclosporine trough levels were maintained between 75 to 200 ng/mL (Ref).

Focal segmental glomerulosclerosis

Focal segmental glomerulosclerosis (off-label use):

Note: Cyclosporine should be administered for at least 4 to 6 months before considering the disease to be resistant. To minimize relapse, continue cyclosporine for at least 12 months if there is an initial response, unless eGFR declines to <30 mL/minute/1.73 m2 (Ref).

Oral: Initial: 3 to 5 mg/kg/day in 2 divided doses (every 12 hours; with or without corticosteroids) (Ref). Target trough levels of 100 to 175 ng/mL are published but only used to verify adherence or to minimize toxicity; dose should be titrated to achieve a reduction in proteinuria. Reduce dose if serum creatinine increases to >30% of baseline; if serum creatinine fails to improve after dose reduction, discontinuation of therapy should be considered (Ref).

Graft-versus-host disease, acute, prevention

Graft-versus-host disease, acute, prevention (off-label use): IV followed by oral:

Initial: IV: 3 mg/kg/day 1 day prior to transplant; may convert to oral therapy when oral intake is tolerated using an IV-to-oral ratio of ~1:2 to 1:4 (depending on protocol); titrate dose to appropriate cyclosporine trough concentration (in combination with methotrexate); taper per protocol (refer to specific references and institutional protocols for tapering and target trough details); discontinue 6 months post-transplant in the absence of acute GVHD (Ref).

or

Initial: IV: 5 mg/kg/day (as a continuous infusion over 20 hours) for 6 days (loading dose) starting 2 days prior to transplant, then 3 mg/kg/day over 20 hours for 11 days starting on post-transplant day 4, then 3.75 mg/kg/day over 20 hours for 21 days starting on day 15, then oral (in 2 divided daily doses): 10 mg/kg/day days 36 to 83, then 8 mg/kg/day days 84 to 97, then 6 mg/kg/day days 98 to 119, then 4 mg/kg/day days 120 to 180, then discontinue (in combination with methotrexate +/- corticosteroid) (Ref).

Graft-versus-host disease, chronic, treatment

Graft-versus-host disease, chronic, treatment (off-label use): Oral: 6 mg/kg twice daily every other day (in combination with prednisone) until week 40, followed by a gradual cyclosporine taper (Ref).

Immune thrombocytopenia, refractory

Immune thrombocytopenia, refractory (off-label use): Oral: 2.5 to 3 mg/kg/day as monotherapy or in combination with other agents (eg, prednisone) (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).

Oral: Initial: 2.5 mg/kg/day in 2 divided doses given every 12 hours; may increase total daily dose in increments of 0.5 mg/kg/day every 4 to 8 weeks to a maximum of 5 mg/kg/day based on tolerance, efficacy, and target trough concentration (maximum total dose is not established). Onset of clinical response to cyclosporine may take 1 to 3 months, with a maximum effect apparent at 7 months (Ref).

Nephrotic syndrome

Nephrotic syndrome (Canadian labeling): Oral: Cyclosporine (modified):

Initial: 3.5 mg/kg/day in 2 divided doses (every 12 hours); titrate for induction of remission and renal function. Adjunct therapy with low-dose oral corticosteroids is recommended for patients with an inadequate response to cyclosporine (particularly if steroid-resistant).

Maintenance: Dose is individualized based on proteinuria, serum creatinine, and tolerability but should be maintained at lowest effective dose; maximum dose: 5 mg/kg daily. Discontinue if no improvement is observed after 3 months.

Psoriasis

Psoriasis: Oral: Cyclosporine (modified): Initial dose: 1 to 3 mg/kg/day in 2 divided doses (Ref).

Titration:

Increase by 0.5 mg/kg/day if insufficient response is seen after 4 weeks of treatment. Additional dosage increases may be made every 2 weeks if needed (maximum dose: 4 mg/kg/day).

Discontinue if no benefit is seen by 6 weeks of therapy at the maximum dose. Once patients are adequately controlled, the dose should be decreased to the lowest effective dose. Treatment longer than 1 year is not recommended.

Note: Increase the frequency of blood pressure monitoring after each alteration in dosage of cyclosporine. Cyclosporine dosage should be decreased by 25% to 50% in patients with no history of hypertension who develop sustained hypertension during therapy and, if hypertension persists, treatment with cyclosporine should be discontinued.

Solid organ transplantation

Solid organ transplantation: Cyclosporine is commonly used in combination with an antiproliferative immunosuppressive agent and a corticosteroid. Although cyclosporine may be initiated preoperatively, it is more frequently started postoperatively (depending on concomitant renal function); adjust dose to achieve desired plasma concentration.

Note: Cyclosporine therapeutic drug monitoring in solid organ transplant recipients is frequently individualized based on disease, concurrent therapy, type of assay used, and institution-specific protocols. Trough concentrations are commonly used for convenience to facilitate dose adjustments for efficacy and toxicity; however, pharmacokinetic studies suggest that the 2-hour post dose level (C2) is a more consistent predictor of overall cyclosporine exposure and may be considered in some clinical scenarios (Ref).

Oral: Dose is dependent upon type of transplant and formulation; refer to institutional protocol for specific dosing:

Cyclosporine (modified): Manufacturer's labeling for newly transplanted patients:

Renal: 9 ± 3 mg/kg/day in 2 divided doses.

Liver: 8 ± 4 mg/kg/day in 2 divided doses.

Heart: 7 ± 3 mg/kg/day in 2 divided doses.

Lung (off label): Initial (conversion from IV post transplant): Usual dosage ranges from 2.5 to 5 mg/kg/day in 2 divided doses but is dependent on IV dose administered and cyclosporine plasma concentrations (Griffith 1994; Treede 2001; Zuckermann 2003).

Cyclosporine (nonmodified): Refer to institutional protocol for specific dosing; dosing in clinical practice may differ greatly compared to the manufacturer's labeling. Note: When using the nonmodified formulation, cyclosporine levels may increase in liver transplant recipients when the T-tube is closed; dose may need to be decreased.

IV: Cyclosporine (nonmodified):

Heart, liver, renal: Manufacturer's labeling for newly transplanted patients: Initial dose: 5 to 6 mg/kg/day or one-third of the oral dose as a single dose, infused over 2 to 6 hours; use should be limited to patients unable to take capsules or oral solution; patients should be switched to an oral dosage form as soon as possible.

Lung (off label): Initial: 1 to 2.5 mg/kg/day beginning immediately after transplantation (in combination with other transplant immunosuppressants); convert to oral cyclosporine with conversion dose based on blood C2 or trough levels as soon as possible post extubation (Griffith 1994; Treede 2001; Zuckermann 2003).

Note: Many transplant centers administer cyclosporine as "divided dose" infusions (in 2 doses daily) or as a continuous (24-hour) infusion. Specific institutional protocols should be consulted.

Conversion to cyclosporine (modified) from cyclosporine (nonmodified): Until the blood trough concentration attains the preconversion value, it is strongly recommended that the cyclosporine blood-trough concentration be monitored every 4 to 7 days after conversion. In addition, monitor clinical safety parameters, such as serum creatinine and BP, every 2 weeks during the first 2 months after conversion. If the blood trough concentrations are outside the desired range and/or if the clinical safety parameters worsen, adjust the dosage accordingly.

T-cell large granular lymphocytic leukemia, symptomatic

T-cell large granular lymphocytic leukemia, symptomatic (off-label use): Oral: 5 to 10 mg/kg/day (in 2 divided doses), adjust dose to maintain therapeutic level of 200 to 400 ng/mL; after 3 months, slowly taper to cyclosporine dose required to maintain response; discontinue if no response or if relapse occurs (Ref) or 1 to 6.7 mg/kg/day (in 2 divided doses), adjust dose to maintain level of ≤250 ng/mL; continue indefinitely in the absence of toxicity (Ref).

Ulcerative colitis, severe, steroid-refractory

Ulcerative colitis, severe, steroid-refractory (off-label use):

IV: Cyclosporine (non-modified): 2 to 4 mg/kg/day, infused continuously over 24 hours (Ref). Note: Some studies suggest no therapeutic difference between low-dose (2 mg/kg) and high-dose (4 mg/kg) cyclosporine regimens; therefore, 2 mg/kg/day is the target treatment dose (Ref).

Oral: Cyclosporine (modified): 2.3 to 3 mg/kg every 12 hours (Ref).

Note: Patients responsive to IV therapy should be switched to oral therapy when possible.

Uveitis

Uveitis (off-label use): Oral: 2.5 to 5 mg/kg/day in 2 divided doses; gradually decrease to maintenance dose; used alone or in conjunction with other corticosteroids (Ref). An expert panel recommends initial dose of 3 to 5 mg/kg/day; reducing dose, once inflammation was under control, to 2 to 3 mg/kg/day until a maintenance dose of 1 mg/kg/day is achieved (Ref).

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 A. Roberts, PhD, BPharm (Hons), B App Sc, FSHP, FISAC; Michael Heung, MD, MS.

Note: Kidney impairment does not significantly alter the pharmacokinetics of cyclosporine (Ref); however, cyclosporine may cause acute or chronic kidney dysfunction. Routine monitoring of cyclosporine concentrations should be considered when therapeutic drug monitoring guidance is available; consider targeting the lower end of the therapeutic range in patients with preexisting kidney impairment (eg, CrCl <60 mL/minute) and avoid concurrent nephrotoxins (eg, nonsteroidal anti-inflammatory drugs) when possible (Ref).

Oral, IV:

Kidney impairment prior to treatment initiation:

Altered kidney function:

CrCl ≥60 mL/minute: No dosage adjustment necessary.

CrCl <60 mL/minute: No dosage adjustment necessary (0.1% excreted in the urine unchanged) (Ref). For nontransplant indications (eg, autoimmune disease), the manufacturer's labeling states use is contraindicated in patients with abnormal renal function (not defined); however, when potential benefits outweigh the risks, may consider cautious use with frequent monitoring of kidney function, or consider use of an alternative agent due to increased risk of worsening kidney function, especially for patients with more severe impairment (Ref).

Hemodialysis, intermittent (thrice weekly): Not dialyzable (Ref): No supplemental dose or dosage adjustment necessary (Ref). For nontransplant indications (eg, autoimmune disease) the manufacturer's labeling states use is contraindicated in patients with abnormal renal function (not defined); however, may use with extreme caution if benefits outweigh risks, or consider use of an alternative agent, especially if the patient has residual kidney function (Ref).

Peritoneal dialysis: Unlikely to be significantly dialyzable (large Vd): No dosage adjustment necessary (Ref). For nontransplant indications (eg, autoimmune disease) the manufacturer's labeling states use is contraindicated in patients with abnormal renal function (not defined); however, may use with extreme caution if benefits outweigh risks, or consider use of an alternative agent, especially if patient has residual kidney function (Ref).

CRRT: No dosage adjustment necessary (Ref). However, cyclosporine can potentially worsen acute kidney injury; therefore, avoid use unless benefits outweigh the risks. Monitor kidney function closely (Ref).

PIRRT (eg, sustained, low-efficiency diafiltration): No dosage adjustment necessary (Ref). However, cyclosporine can potentially worsen acute kidney injury; therefore, avoid use unless benefits outweigh the risks. Monitor kidney function closely (Ref).

Nephrotoxicity or acute kidney injury during treatment:

Altered kidney function:

Nontransplant indications (eg, autoimmune disease): The following general recommendations may be considered; individualize therapy according to risks/benefits and institutional protocols, when available:

If serum creatinine increases 25% to 30% above baseline (measured on 2 separate occasions at least 2 weeks apart), or by ≥50% at any time during therapy, reduce dose by 25% to 50% and monitor serum creatinine every 2 weeks for 1 month. If serum creatinine does not decrease to within 25% to 30% of baseline, further reduce dose by 25% to 50% and monitor serum creatinine every 2 weeks for 1 month. If serum creatinine does not decrease to within 25% to 30% of baseline, discontinue cyclosporine (Ref).

Transplant indications: There are no specific dosage adjustments recommended; therapy should be individualized by the patient's transplant team (Ref).

Patients receiving renal replacement therapies (eg, hemodialysis, peritoneal dialysis, CRRT):

Nontransplant indications (eg, autoimmune disease): Consider temporary interruption of therapy or switching to an alternative agent to help promote renal recovery and preserve residual kidney function if other factors (eg, concurrent nephrotoxins, dehydration) contributing to decreased kidney function cannot be mitigated. Continued use should only be considered if benefits outweigh risks of further kidney injury (Ref).

Transplant indications: There are no specific dosage adjustments recommended; therapy should be individualized by the patient's transplant team (Ref).

Dosing: Hepatic Impairment: Adult

Mild-to-moderate impairment: There are no dosage adjustments provided in the manufacturer’s labeling; monitor blood concentrations.

Severe impairment: There are no dosage adjustments provided in the manufacturer’s labeling; however, metabolism is extensively hepatic (exposure is increased). Monitor blood concentrations; may require dose reduction.

Dosing: Older Adult

Refer to adult dosing.

Dosing: Pediatric

(For additional information see "Cyclosporine (ciclosporin) (systemic): Pediatric drug information")

Note: Cyclosporine (modified) (Gengraf, Neoral) and cyclosporine (non-modified) (Sandimmune) are not bioequivalent and cannot be used interchangeably. Initial dosing provided; adjust initial dose to achieve desired target cyclosporine concentration for indication; specific institutional protocols should be consulted.

Hepatitis, autoimmune, second-line therapy

Hepatitis, autoimmune, second-line therapy: Limited data available:

Children ≥2 years and Adolescents: Oral: Initial: 3 to 5 mg/kg/day in divided doses every 8 to 12 hours; titrate to targeted cyclosporine trough concentrations; concentrations reported in the literature range between 150 to 300 ng/mL for induction of remission with lower goals (100 to 200 ng/mL) following remission, and further reduced after 1 year of therapy to attain goals of 50 to 70 ng/mL. When clinically indicated, cyclosporine may be tapered off (over ~14 days) (Ref).

Graft-versus-host disease, prevention

Graft-versus-host disease (GVHD), prevention: Limited data available, optimal regimen not established:

Infants, Children, and Adolescents: IV followed by oral: Initial: IV: 3 to 5 mg/kg/day in divided doses every 12 hours administered over 2 hours (Ref) beginning 1 day prior to transplant; convert to oral therapy when tolerating oral intake; titrate dose to appropriate cyclosporine trough concentration, reported doses ranged from 6 to 12.5 mg/kg/day in divided doses every 12 hours; taper per protocol (refer to specific references for tapering and target trough details); duration of therapy variable and dependent upon clinical parameters including underlying disease (malignant vs nonmalignant); in reports, 100 days post-hematopoietic stem-cell transplantation to 6 months has been reported (Ref). Lower doses of 1 mg/kg/day divided every 12 hours have also been reported; a study of 59 pediatric patients (median age: 8 years; range: 1 to 18 years) with acute leukemia compared low-dose IV cyclosporine (1 mg/kg/day divided every 12 hours) with standard IV dose (3 mg/kg/day divided every 12 hours); although patients receiving the lower initial IV dose experienced a higher incidence of GVHD there was a statistically significant decrease in the rate of leukemia relapse (15% vs 41% in the standard dose group) (Ref).

Kawasaki disease, refractory; alternative therapy

Kawasaki disease, refractory; alternative therapy:

Note: Consider use in refractory Kawasaki disease when a second intravenous immune globulin (IVIG) infusion, infliximab, or a course of steroids has failed (Ref). Dose varies based on route of administration.

IV: Infants ≥2 months and Children: IV: 3 mg/kg/day in divided doses every 12 hours; adjust dose to achieve the following cyclosporine concentrations: 2-hour peak of 300 to 600 ng/mL and trough of 50 to 150 ng/mL. Transition to oral cyclosporine (modified) when clinically feasible (Ref).

Oral: Infants ≥2 months and Children: Cyclosporine (modified) (Gengraf, Neoral): Oral: 4 to 8 mg/kg/day in divided doses every 12 hours. Higher doses of 10 mg/kg/day have also been reported; adjust dose to achieve the following cyclosporine concentrations: 2-hour peak of 300 to 600 ng/mL and trough of 50 to 150 ng/mL. Once patient is afebrile, clinically improving, and C-reactive protein is ≤1 mg/dL or after 2 weeks of therapy (whichever is longer), taper by 10% every 3 days. May discontinue once dose is down to 1 mg/kg/day (Ref).

Lupus nephritis

Lupus nephritis: Limited data available: Note: Cyclosporine may be considered as maintenance therapy in patients with Class I or II who are at risk for relapse, or as initial induction and subsequent maintenance therapy for use in patients with at least Class III lupus nephritis for whom cyclophosphamide or standard-dose mycophenolic acid analogs (MPAA) are not an option; use in combination with MPAA at a reduced doses and glucocorticoids (Ref).

Children and Adolescents: Oral: 3 to 5 mg/kg/day in divided doses; adjust dose to maintain patient-specific targeted trough concentration (eg, ~60 to 100 ng/mL); continue therapy for at least 1 to 2 years; some experts suggest a duration of >36 months for proliferative cases (Ref). Reported range from trials: 2 to 5 mg/kg/day divided every 12 hours adjusted to maintain targeted trough concentrations. In one trial of 40 children and adolescents (age range: 9 to 14 years), a dosage range of 3.5 to 5 mg/kg/day was shown to effectively decrease proteinuria and allow for reduction of steroid dose; study duration 1 year (Ref). A smaller trial of 7 adolescents (14 to 18 years) used an initial dose of 1.5 to 3 mg/kg/day and adjusted to clinical response; after 1 year of therapy, final dosage range was 2 to 4 mg/kg/day (Ref). Case reports have suggested that lower doses (1.8 to 2.5 mg/kg/day) are effective to induce remission (Ref). In a small trial (n=13; age: 4 to 16 years), combination therapy with mycophenolate for induction of severe disease has been reported using cyclosporine 3 to 6 mg/kg/day in 2 to 3 divided doses (Ref).

Nephrotic syndrome

Nephrotic syndrome: Limited data available:

Frequently relapsing (with or without steroid dependency):

Weight-directed dosing: Children and Adolescents: Oral: 4 to 5 mg/kg/day in divided doses twice daily; titrate doses to achieve target trough concentration of 60 to 150 ng/mL. In children <6 years of age, daily dose may be divided 3 times daily to achieve goals (Ref). Doses as low as 3 mg/kg/day divided twice daily have also been recommended (Ref). Treatment should continue for at least 12 months to reduce risk of relapse (Ref).

BSA-directed dosing: Children ≥3 years and Adolescents: Cyclosporine (modified): Oral: 150 mg/m2/day in divided doses twice daily; titrate doses to achieve a target trough concentration of 80 to 100 ng/mL; dose based on a randomized, open-label, crossover study of 60 pediatric patients (age range: 3 to 18 years) (Ref). Note: KDIGO guidelines recommend goal trough concentration of 60 to 150 ng/mL (Ref).

Steroid resistant:

Weight-directed dosing: Cyclosporine: Children and Adolescents: Oral: 3 to 6 mg/kg/day in divided doses twice daily; to achieve target trough concentration of 60 to 150 ng/mL. After 6 months of therapy, assess continuation. If no remission (partial or complete), then discontinue therapy; if remission is achieved (partial or complete), then continue for a minimum of 12 months (Ref).

BSA-directed dosing: Cyclosporine (modified): Children and Adolescents: Oral: 150 mg/m2/day in divided doses twice daily; titrate doses to achieve a target trough concentration of 120 to 180 ng/mL; in one study, if remission was not achieved by 12 weeks, doses were increased to achieve a target trough concentration of 300 to 400 ng/mL (Ref). Note: KDIGO guidelines recommend goal trough concentration of 60 to 150 ng/mL (Ref).

Solid organ transplantation, rejection prophylaxis

Solid organ transplantation , rejection prophylaxis:

IV: Cyclosporine (nonmodified): Note: Limit parenteral use to patients unable to take capsules or oral solution; patients should be switched to an oral dosage form as soon as possible.

Intermittent IV infusion:

Manufacturer's labeling: Infants ≥6 months, Children, and Adolescents: Initial: 5 to 6 mg/kg/day or one-third (1/3) of the oral dose as a single daily dose over 2 to 6 hours; administer 4 to 12 hours prior to organ transplantation or may be given postoperatively

Alternate dosing: Limited data available: Infants ≥3 months, Children, and Adolescents: 2 to 6 mg/kg/day in divided doses every 8 to 12 hours (Ref)

Continuous IV infusion: Limited data available: Children and Adolescents: 2 to 4 mg/kg/day; doses as high as 4.5 mg/kg/day have been used in patients ≥6 years old (Ref)

Oral: Note: Dose is dependent upon type of transplant and formulation. Adjust dose to achieve desired target cyclosporine concentration; specific institutional protocols should be consulted.

Cyclosporine (nonmodified): Infants ≥6 months, Children, and Adolescents: Initial: 10 to 14 mg/kg/day has been used for renal transplants (the manufacturer's labeling includes dosing from initial clinical trials of 15 mg/kg/day [range: 14 to 18 mg/kg/day]); administer 4 to 12 hours prior to organ transplantation. Continue initial dose daily for 1 to 2 weeks; taper by 5% per week to a maintenance dose of 5 to 10 mg/kg/day; some renal transplant recipients may be dosed as low as 3 mg/kg/day to achieve target concentrations.

Note: When using nonmodified formulation, cyclosporine concentrations may increase in liver transplant recipients when the T-tube is closed; may need to decrease dose (Ref).

Cyclosporine (modified): Dose differs based on type of transplant, consult institution protocol: Dosage below is the mean (±SD) of initial doses used in a 1994 survey.

Initial dose:

Kidney: 9 ± 3 mg/kg/day divided every 12 hours

Liver: 8 ± 4 mg/kg/day divided every 12 hours

Heart: 7 ± 3 mg/kg/day divided every 12 hours

Conversion to cyclosporine (modified) from cyclosporine (nonmodified): Start with daily dose previously used and adjust to obtain preconversion cyclosporine trough concentration. Plasma concentrations should be monitored every 4 to 7 days and dose adjusted as necessary, until desired trough concentration is obtained. When transferring patients with previously poor absorption of cyclosporine (nonmodified), monitor trough concentrations at least twice weekly (especially if initial dose exceeds 10 mg/kg daily); high plasma concentrations are likely to occur due to improved bioavailability.

Dosing: Kidney Impairment: Pediatric

There are no pediatric-specific dosage adjustments provided in the manufacturer's labeling; based on experience in adult patients, monitor concentrations and adjust dose for increases in serum creatinine.

Nephrotic syndrome:

eGFR <30 mL/minute/1.73 m2: Reduce dose or discontinue therapy (Ref).

Dosing: Hepatic Impairment: Pediatric

Mild to moderate impairment: There are no dosage adjustments provided in the manufacturer's labeling; monitor blood concentrations.

Severe impairment: There are no dosage adjustments provided in the manufacturer's labeling; however, metabolism is extensively hepatic (exposure is increased). Monitor blood concentrations; may require dose reduction.

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 tacrolimus, cyclosporine may have a lower 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

• Black or Hispanic kidney transplant recipients (Ref)

• Hepatitis C virus infection (Ref)

Drug-induced gingival overgrowth

Cyclosporine is a common cause of drug-induced gingival overgrowth (GO) (ie, gingival hyperplasia). Overgrowth may initially occur on the labial gingiva and in between the teeth; as overgrowth progresses, papillae appear to coalesce and patients may go on to experience swelling, bleeding, and difficulties with chewing and speech (Ref). Cyclosporine-induced GO may be reversible upon discontinuation of therapy (Ref); recurrence is likely when cyclosporine cannot be discontinued (Ref). Tacrolimus is less likely to result in GO as compared to cyclosporine (Ref).

Mechanism: Dose-related (Ref); exact mechanism not clearly established.

Onset: Delayed. Most cases occur within 3 months of treatment initiation; however, cases have been reported as early as 1 month following initiation (Ref)

Risk factors:

• Greater exposure to cyclosporine (ie, higher doses, increased serum concentrations [≥400 ng/mL]) (Ref)

• Concomitant use of other medications known to increase the risk of gingival hyperplasia (eg, calcium channel blockers, phenytoin) (Ref)

• Poor oral hygiene (eg, preexisting gingival inflammation (Ref); preexisting plaque accumulation (Ref)) (Ref)

• Prior history of gingival hyperplasia (Ref)

• The impact of human leukocyte antigen (HLA) types and matching on the risk of gingival overgrowth vary:

- HLA-A24 expression has been associated with an increased risk (Ref)

- HLA-DR1 expression has been associated with a decreased risk (Ref)

- HLA-A mismatching between donor and host following kidney transplant was found to be protective (Ref)

- No association with risk and HLA haplotype was detected in kidney transplant recipients (Ref)

Drug-induced thrombotic microangiopathy

Cyclosporine use may cause drug-induced thrombotic microangiopathy (DITMA) which can result in microangiopathic hemolytic anemia with thrombocytopenia (MAHA), microvascular thrombosis with vessel wall abnormalities (Ref). Patients generally present with kidney impairment and hypertension (Ref); however, other organ systems may also be impacted by cyclosporine-related DITMA (eg, pulmonary, dermatologic, musculoskeletal, hepatic, gastrointestinal) (Ref). Graft failure may or may not occur (Ref). Resolution can be achieved with early detection of the syndrome, cyclosporine discontinuation or dosage reduction, and initiation of syndrome-specific supportive treatment; however, kidney impairment may persist (Ref).

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

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

Risk factors:

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

- Ischemia-reperfusion endothelial injury

- Kidney infections

- Vascular rejection

- Anticardiolipin antibodies

- Malignancies

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

Hepatotoxicity

Hepatotoxicity, characterized by hyperbilirubinemia, mild to moderate transaminase elevations, and biliary tract disease (eg, cholelithiasis) may occur in pediatric and adult patients (Ref). Liver injury, including cholestasis, jaundice, hepatitis, and hepatic failure, has also been reported. Improvement may be seen following dosage reduction (Ref)

Mechanism: Dose-related; related to the pharmacologic action (impaired bile acid flow resulting in intrahepatic cholestasis) (Ref)

Onset: Varied. May occur 2 to 8 weeks post transplant; however, later onset (>1 year post transplant) has been described (Ref)().

Risk factors:

• Greater exposure to cyclosporine (ie, higher doses, increased serum concentrations (Ref))

• Comorbid infection (eg, hepatitis C virus (Ref))

• Concomitant use of other potentially hepatotoxic medications

• Patients receiving total parenteral nutrition (Ref)

Hyperkalemia

Mild to severe hyperkalemia may occur in patients who receive a calcineurin inhibitor (CNI), including cyclosporine; one retrospective trial described less severe as well as a shorter duration of hyperkalemia in patients treated with cyclosporine as compared to patients who received tacrolimus (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 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)

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). Cyclosporine-based regimens may be associated with a higher incidence of hypertension as compared to tacrolimus-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: Dose-dependent (Ref); 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, and endothelin-1) and a decrease of vasodilator mediators (eg, prostaglandins, nitric oxide) (Ref). Cyclosporine 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:

• Increasing dose (Ref)

• Longer duration

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

- Preexisting hypertension

- Elevated body max index

- Males

- Black patients

- Older donor age

- Delayed graft function

- Glucocorticoid use

- Recurrent disease

- Acute rejection

- Post-transplant proteinuria

Infections

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

Viral infections reported with use of cyclosporine include polyomavirus infection (which may result in polyomavirus-associated nephropathy [PVAN]), JC virus infection-associated progressive multifocal leukoencephalopathy, cytomegalovirus disease (CMV), and reactivation of hepatitis B virus or hepatitis C virus (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, including cyclosporine, as compared to mTOR inhibitors (eg, sirolimus or everolimus) (Ref); cyclosporine may be associated with a lower risk of infections as compared to glucocorticoids or other immunosuppressants (eg, cyclophosphamide) when used for the treatment of idiopathic membranous nephropathy (Ref).

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

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 cyclosporine exposure

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

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 lower in transplant recipients who receive cyclosporine as compared to tacrolimus (Ref); however, not all studies have observed this pattern (Ref).

Mechanism: Dose- and time-related; related to the pharmacologic action. Cyclosporine 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)

- Note: 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)

- Note: 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 human leukocyte antigen 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

- Patients with psoriasis with prior use of psoralen plus ultraviolet A photochemotherapy and possibly methotrexate or other immunosuppressants, UVB, coal tar, or radiation.

Nephrotoxicity

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

Acute CNI nephrotoxicity: Considered to be acute, functional, dose-dependent, and generally reversible after dose reduction or discontinuation; characterized by moderate increased serum creatinine concentrations unexplained by other causes for acute kidney injury (AKI) with a concomitant increase in cyclosporine trough concentrations (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 CNI nephrotoxicity: 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, and arteriolar myocytes (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, and endothelin-1) and a decrease of vasodilator mediators (eg, prostaglandins, nitric oxide) (Ref).

Chronic CNI nephrotoxicity : 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 TGF-β and activation of the renin-angiotensin system may also contribute to the tubular-interstitial effects seen in patients with chronic CNI nephrotoxicity (Ref). 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]) (Ref).

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 cyclosporine (Ref)

• Increased local exposure to cyclosporine (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) (Ref)

• Patients receiving cyclosporine for the treatment of steroid-dependent nephrotic syndrome as compared to patients with steroid-sensitive nephrotic syndrome (Ref)

Neurotoxicity

Dose-related neurotoxicity may occur with use of calcineurin inhibitors (CNIs), including cyclosporine. Neurotoxicity may be less common with cyclosporine as compared to tacrolimus (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). Headaches are often described as migrainous, occipital, and frequent (eg, weekly) (Ref).

More severe symptoms of neurotoxicity may occur less frequently and include seizure, visual disturbance (including cortical blindness), 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. Cyclosporine-induced neurotoxicity may be related to the ability of CNIs to increase the permeability of the blood brain barrier via multiple mechanisms; once within the CNS, cyclosporine 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 cyclosporine exposure; including dosing strategies that result in high peak concentrations (eg, 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)

• Concomitant use of high-dose corticosteroids

RPLS: Risk is increased in patients with significant fluid overload, elevated blood pressure, hypomagnesemia, hypocholesterolemia, concomitant use of high-dose corticosteroids, graft-versus-host disease, or impaired kidney function (Ref)

Adverse Reactions

The following adverse drug reactions and incidences are derived from product labeling unless otherwise specified. Adverse reactions reported with systemic use, including rheumatoid arthritis, psoriasis, and transplantation (kidney, liver, and heart). Percentages noted include the highest frequency regardless of indication/dosage. Frequencies may vary for specific conditions or formulation.

Kidney, liver, and heart transplant:

>10%:

Cardiovascular: Hypertension (13% to 53%) (table 1)

Cyclosporine: Adverse Reaction: Hypertension

Drug (Cyclosporine)

Comparator (Azathioprine)

Indication

Number of Patients (Cyclosporine)

Number of Patients (Azathioprine)

53%

N/A

Heart transplant

112

N/A

26%

18%

Kidney transplant

227

228

13%

N/A

Kidney transplant

705

N/A

27%

N/A

Liver transplant

75

N/A

Endocrine & metabolic: Hirsutism (21% to 45%)

Gastrointestinal: Gingival hyperplasia (4% to 16%) (table 2)

Cyclosporine: Adverse Reaction: Gingival Hyperplasia

Drug (Cyclosporine)

Comparator (Azathioprine)

Indication

Number of Patients (Cyclosporine)

Number of Patients (Azathioprine)

5%

N/A

Heart transplant

112

N/A

9%

N/A

Kidney transplant

705

N/A

4%

0%

Kidney transplant

227

228

16%

N/A

Liver transplant

75

N/A

Genitourinary: Urinary tract infection (21%)

Infection: Viral infection (16%) (table 3)

Cyclosporine: Adverse Reaction: Viral Infection

Drug (Cyclosporine)

Comparator (Azathioprine with Steroids)

Indication

Number of Patients (Cyclosporine)

Number of Patients (Azathioprine with Steroids)

16%

18%

Kidney transplant

227

228

Nervous system: Headache (2% to 15%) (table 4)

Cyclosporine: Adverse Reaction: Headache

Drug (Cyclosporine)

Comparator (Azathioprine)

Indication

Number of Patients (Cyclosporine)

Number of Patients (Azathioprine)

15%

N/A

Heart transplant

112

N/A

2%

<1%

Kidney transplant

227

228

2%

N/A

Kidney transplant

705

N/A

4%

N/A

Liver transplant

75

N/A

Neuromuscular & skeletal: Tremor (21% to 55%) (table 5)

Cyclosporine: Adverse Reaction: Tremor

Drug (Cyclosporine)

Comparator (Azathioprine)

Indication

Number of Patients (Cyclosporine)

Number of Patients (Azathioprine)

31%

N/A

Heart transplant

112

N/A

21%

N/A

Kidney transplant

705

N/A

12%

0%

Kidney transplant

227

228

55%

N/A

Liver transplant

75

N/A

Renal: Nephrotoxicity (25% to 38%) (table 6)

Cyclosporine: Adverse Reaction: Nephrotoxicity

Drug (Cyclosporine)

Comparator (Azathioprine)

Indication

Number of Patients (Cyclosporine)

Number of Patients (Azathioprine)

38%

N/A

Heart transplant

112

N/A

32%

6%

Kidney transplant

227

228

25%

N/A

Kidney transplant

705

N/A

37%

N/A

Liver transplant

75

N/A

1% to 10%:

Cardiovascular: Chest pain (≤4%), edema (≤2%), flushing (≤4%)

Dermatologic: Acne vulgaris (1% to 6%), nail disease (brittle fingernails) (≤2%), skin infection (≤7%)

Endocrine & metabolic: Gynecomastia (≤4%), hyperglycemia (≤2%)

Gastrointestinal: Abdominal distress (≤7%), anorexia (≤2%), diarrhea (3% to 8%), gastritis (≤2%), hiccups (≤2%), nausea and vomiting (4% to 10%), peptic ulcer (≤2%)

Hematologic & oncologic: Anemia (≤2%), leukopenia (≤6%), malignant lymphoma (≤6%) (table 7), thrombocytopenia (≤2%)

Cyclosporine: Adverse Reaction: Malignant Lymphoma

Drug (Cyclosporine)

Comparator (Azathioprine)

Indication

Number of Patients (Cyclosporine)

Number of Patients (Azathioprine)

6%

N/A

Heart transplant

112

N/A

1%

N/A

Kidney transplant

705

N/A

<1%

0%

Kidney transplant

227

228

1%

N/A

Liver transplant

75

N/A

Hepatic: Hepatotoxicity (≤7%)

Hypersensitivity: Hypersensitivity reaction (≤2%)

Infection: Abscess (4%), cytomegalovirus disease (5%) (table 8), fungal infection (systemic: 2%) (table 9), localized fungal infection (8%) (table 10), septicemia (5%), wound infection (≤7%)

Cyclosporine: Adverse Reaction: Cytomegalovirus Disease

Drug (Cyclosporine)

Comparator (Azathioprine with Steroids)

Indication

Number of Patients (Cyclosporine)

Number of Patients (Azathioprine with Steroids)

5%

12%

Kidney transplant

227

228

Cyclosporine: Adverse Reaction: Fungal Infection

Drug (Cyclosporine)

Comparator (Azathioprine with Steroids)

Indication

Number of Patients (Cyclosporine)

Number of Patients (Azathioprine with Steroids)

Comments

2%

4%

Kidney transplant

227

228

Systemic fungal infection

Cyclosporine: Adverse Reaction: Localized Fungal Infection

Drug (Cyclosporine)

Comparator (Azathioprine with Steroids)

Indication

Number of Patients (Cyclosporine)

Number of Patients (Azathioprine with Steroids)

8%

10%

Kidney transplant

227

228

Nervous system: Confusion (≤2%), paresthesia (1% to 3%), seizure (1% to 5%) (table 11)

Cyclosporine: Adverse Reaction: Seizure

Drug (Cyclosporine)

Comparator (Azathioprine)

Indication

Number of Patients (Cyclosporine)

Number of Patients (Azathioprine)

4%

N/A

Heart transplant

112

N/A

3%

1%

Kidney transplant

227

228

1%

N/A

Kidney transplant

705

N/A

5%

N/A

Liver transplant

75

N/A

Neuromuscular & skeletal: Myalgia (≤2%)

Ophthalmic: Conjunctivitis (≤2%)

Otic: Hearing loss (≤2%), tinnitus (≤2%)

Respiratory: Pneumonia (6%), sinusitis (≤7%)

Miscellaneous: Fever (≤2%)

<1%:

Cardiovascular: Acute myocardial infarction

Dermatologic: Night sweats, pruritus, trichorrhexis

Endocrine & metabolic: Weight loss

Gastrointestinal: Aphthous stomatitis, constipation, dysphagia, pancreatitis

Genitourinary: Hematuria

Hematologic & oncologic: Upper gastrointestinal hemorrhage

Nervous system: Anxiety, depression, lethargy, tingling sensation

Neuromuscular & skeletal: Arthralgia, asthenia

Ophthalmic: Visual disturbance

Psoriasis or rheumatoid arthritis (RA):

Incidence reported for RA unless otherwise noted.

>10%:

Cardiovascular: Edema (5% to 14%), hypertension (psoriasis, RA: 8% to 28%) (table 12)

Cyclosporine: Adverse Reaction: Hypertension

Drug (Cyclosporine)

Placebo

Indication

Number of Patients (Cyclosporine)

Number of Patients (Placebo)

Comments

28%

N/A

Psoriasis

182

N/A

Neoral

25%

N/A

Psoriasis

185

N/A

Sandimmune

26%

N/A

Rheumatoid arthritis

155

N/A

Sandimmune

25%

N/A

Rheumatoid arthritis

143

N/A

Neoral

8%

2%

Rheumatoid arthritis

269

201

Sandimmune

Dermatologic: Hypertrichosis (psoriasis: 5% to 7%; RA: 15% to 19%, skin rash (psoriasis: 1% to 3%; RA: 12%)

Endocrine & metabolic: Increased nonprotein nitrogen (18% to 19%), increased serum triglycerides (psoriasis: ≤15%)

Gastrointestinal: Abdominal pain (psoriasis: 3% to 6%; RA: 15%), diarrhea (psoriasis: 5% to 6%; RA: 12% to 13%), dyspepsia (psoriasis: 2% to 3%; RA: 8% to 12%), nausea (psoriasis: 6%; RA: 18% to 23%)

Genitourinary: Female genital tract disease (psoriasis: 9% to 12%)

Infection: Infection (psoriasis: 24% to 25%; RA: 1% to 3%)

Nervous system: Headache (psoriasis, RA: 14% to 25%) (table 13), pain (psoriasis: 3% to 4%; RA: 6% to 13%), paresthesia (psoriasis, RA: 5% to 11%)

Cyclosporine: Adverse Reaction: Headache

Drug (Cyclosporine)

Placebo

Indication

Number of Patients (Cyclosporine)

Number of Patients (Placebo)

Comments

16%

N/A

Psoriasis

182

N/A

Neoral

14%

N/A

Psoriasis

185

N/A

Sandimmune

25%

N/A

Rheumatoid arthritis

143

N/A

Neoral

23%

N/A

Rheumatoid arthritis

155

N/A

Sandimmune

17%

9%

Rheumatoid arthritis

269

201

Sandimmune

Neuromuscular & skeletal: Lower limb cramp (2% to 12%), tremor (7% to 13%) (table 14)

Cyclosporine: Adverse Reaction: Tremor

Drug (Cyclosporine)

Placebo

Indication

Number of Patients (Cyclosporine)

Number of Patients (Placebo)

Comments

13%

N/A

Rheumatoid arthritis

143

N/A

Neoral

8%

4%

Rheumatoid arthritis

269

201

Sandimmune

7%

N/A

Rheumatoid arthritis

155

N/A

Sandimmune

Renal: Increased serum creatinine (psoriasis: 16% to 20%; RA: ≥30%: 39% to 48%; RA: ≥50%: 18% to 24%) (table 15), nephrotoxicity (psoriasis: 21%; RA: 10%)

Cyclosporine: Adverse Reaction: Increased Serum Creatinine

Drug (Cyclosporine)

Placebo

Indication

Number of Patients (Cyclosporine)

Number of Patients (Placebo)

Comments

20%

N/A

Psoriasis

182

N/A

Neoral

16%

N/A

Psoriasis

185

N/A

Sandimmune

48%

N/A

Rheumatoid arthritis

143

N/A

Neoral; creatinine elevations ≥30%

43%

13%

Rheumatoid arthritis

269

201

Sandimmune; creatinine elevations ≥30%

39%

N/A

Rheumatoid arthritis

155

N/A

Sandimmune; creatinine elevations ≥30%

24%

3%

Rheumatoid arthritis

269

201

Sandimmune; creatinine elevations ≥50%

18%

N/A

Rheumatoid arthritis

155

N/A

Sandimmune; creatinine elevations ≥50%

18%

N/A

Rheumatoid arthritis

143

N/A

Neoral; creatinine elevations ≥50%

Respiratory: Flu-like symptoms (psoriasis: 8% to 19%; RA: 3%), upper respiratory tract infection (psoriasis, RA: 8% to 14%)

1% to 10%:

Cardiovascular: Abnormal heart sounds (1% to 3%), acute myocardial infarction (1% to 3%), cardiac arrhythmia (2% to 5%), cardiac failure (1% to 3%), chest pain (psoriasis, RA: 1% to 6%), flushing (psoriasis, RA: 1% to 3%), peripheral ischemia (1% to 3%)

Dermatologic: Acne vulgaris (psoriasis: 1% to 3%), cellulitis (1% to 3%), dermatitis (1% to 3%), diaphoresis (1% to 3%), dyschromia (1% to 3%), eczema (1% to 3%), enanthema (1% to 3%), folliculitis (psoriasis, RA: 1% to 3%), hyperkeratosis (psoriasis: 1% to 3%), nail disease (1% to 3%), pruritus (psoriasis, RA: 1% to 3%), urticaria (1% to 3%), xeroderma (psoriasis, RA: 1% to 3%)

Endocrine & metabolic: Decreased libido (1% to 3%), diabetes mellitus (1% to 3%), goiter (1% to 3%), hot flash (psoriasis, RA: 1% to 3%), hyperkalemia (1% to 3%), hyperuricemia (1% to 3%), hypoglycemia (1% to 3%), hypomagnesemia (4% to 6%), increased libido (1% to 3%), increased serum cholesterol (psoriasis: <3%), menstrual disease (3%), weight gain (1% to 3%), weight loss (1% to 3%)

Gastrointestinal: Abdominal distention (psoriasis: 1% to 3%), constipation (psoriasis, RA: 1% to 3%), dysgeusia (1% to 3%), dysphagia (1% to 3%), enlargement of salivary glands (1% to 3%), eructation (1% to 3%), esophagitis (1% to 3%), flatulence (4% to 5%), gastric ulcer (1% to 3%), gastritis (1% to 3%), gastroenteritis (1% to 3%), gingival hemorrhage (psoriasis, RA: 1% to 3%), gingival hyperplasia (psoriasis, RA: 2% to 6%) (table 16), gingivitis (3% to 4%), glossitis (1% to 3%), increased appetite (psoriasis: 1% to 3%), peptic ulcer (1% to 3%), tongue disease (1% to 3%), vomiting (6% to 9%), xerostomia (1% to 3%)

Cyclosporine: Adverse Reaction: Gingival Hyperplasia

Drug (Cyclosporine)

Placebo

Indication

Number of Patients (Cyclosporine)

Number of Patients (Placebo)

Comments

6%

N/A

Psoriasis

185

N/A

Sandimmune

4%

N/A

Psoriasis

182

N/A

Neoral

4%

N/A

Rheumatoid arthritis

155

N/A

Sandimmune

4%

N/A

Rheumatoid arthritis

143

N/A

Neoral

2%

1%

Rheumatoid arthritis

269

201

Sandimmune

Genitourinary: Breast fibroadenosis (1% to 3%), hematuria (1% to 3%), leukorrhea (1%), mastalgia (1% to 3%), nocturia (1% to 3%), urinary frequency (psoriasis, RA: 1% to 4%), urinary incontinence (1% to 3%), urinary tract infection (3%), urinary urgency (1% to 3%), urine abnormality (1% to 3%), uterine hemorrhage (1% to 3%)

Hematologic & oncologic: Abnormal erythrocytes (psoriasis: 1% to 3%), anemia (1% to 3%), bleeding tendency disorder (psoriasis: 1% to 3%), carcinoma (1% to 3%), disorder of hemostatic components of blood (psoriasis: 1% to 3%), leukopenia (1% to 3%), lymphadenopathy (1% to 3%), malignant neoplasm of skin (psoriasis: 1% to 3%), purpuric disease (2% to 4%), qualitative disorder of platelet function (psoriasis: 1% to 3%), rectal hemorrhage (3%)

Hepatic: Hyperbilirubinemia (psoriasis, RA: 1% to 3%)

Hypersensitivity: Angioedema (1% to 3%)

Infection: Abscess (including renal; 1% to 3%), bacterial infection (1% to 3%), candidiasis (1% to 3%), fungal infection (1% to 3%), herpes simplex infection (1% to 3%), herpes zoster infection (1% to 3%), viral infection (1% to 3%)

Nervous system: Anxiety (1% to 3%), confusion (1% to 3%), depression (3% to 6%), dizziness (psoriasis: 1% to 3%; RA: 6% to 8%), drowsiness (1% to 3%), emotional lability (1% to 3%), hypoesthesia (1% to 3%), insomnia (psoriasis, RA: 1% to 4%) (table 17), lack of concentration (1% to 3%), malaise (1% to 3%), migraine (2% to 3%), nervousness (psoriasis, RA: 1% to 3%), neuropathy (1% to 3%), paranoid ideation (1% to 3%), psychiatric disturbance (psoriasis: 4% to 5%), rigors (3%), vertigo (psoriasis, RA: 1% to 3%)

Cyclosporine: Adverse Reaction: Insomnia

Drug (Cyclosporine)

Placebo

Indication

Number of Patients (Cyclosporine)

Number of Patients (Placebo)

Comments

4%

2%

Rheumatoid arthritis

269

201

Sandimmune

3%

N/A

Rheumatoid arthritis

143

N/A

Neoral

1%

N/A

Rheumatoid arthritis

155

N/A

Sandimmune

Neuromuscular & skeletal: Arthralgia (psoriasis, RA: 1% to 6%), arthropathy (4%), asthenia (1% to 3%), bone fracture (1% to 3%), bursitis (1% to 3%), dislocation (1% to 3%), muscle spasm (≤4%), myalgia (1% to 3%), stiffness (1% to 3%), synovial cyst (1% to 3%), tendinopathy (1% to 3%)

Ophthalmic: Cataract (1% to 3%), conjunctivitis (1% to 3%), eye pain (1% to 3%), visual disturbance (psoriasis, RA: 1% to 3%)

Otic: Deafness (1% to 3%), tinnitus (1% to 3%), vestibular disturbance (1% to 3%)

Renal: Increased blood urea nitrogen (1% to 3%), polyuria (1% to 3%), pyelonephritis (1% to 3%)

Respiratory: Abnormal breath sounds (1% to 3%), bronchospasm (psoriasis, RA: ≤5%), cough (psoriasis, RA: ≤5%), dyspnea (psoriasis, RA: ≤5%), epistaxis (1% to 3%), pharyngitis (5%), respiratory tract infection (psoriasis: 1% to 3%), rhinitis (psoriasis, RA: ≤5%), sinusitis (4%), tonsillitis (1% to 3%)

Miscellaneous: Fever (psoriasis: 1% to 3%)

<1%: Hematologic & oncologic: Basal cell carcinoma of skin (psoriasis), malignant lymphoma, squamous cell carcinoma of skin (psoriasis)

Frequency not defined (any indication):

Cardiovascular: Glomerular capillary thrombosis

Hematologic & oncologic: Lymphoproliferative disorder

Renal: Renal tubular necrosis

Postmarketing (any indication):

Dermatologic: Exacerbation of psoriasis

Endocrine & metabolic: Gout (Clive 2000), hyperkalemia (Farouk 2020), hyperuricemia (Clive 2000)

Gastrointestinal: Cholestasis (Gbur 1991)

Hematologic & oncologic: Hemolytic-uremic syndrome (Chiurchiu 2002, Remuzzi 1989), malignant neoplasm (Varga 1991), microangiopathic hemolytic anemia with thrombocytopenia (Ödek 2014), thrombotic microangiopathy (Al Nouri 2014)

Hepatic: Hepatic failure, hepatitis, jaundice

Hypersensitivity: Anaphylaxis (including nonimmune anaphylaxis; possibly associated with Cremophor EL vehicle in injection formulation) (Moeinian 2018)

Immunologic: Graft complications

Infection: Increased susceptibility to infection (including JC virus infection, BK virus, polyomavirus infection, and polyomavirus associated nephropathy) (Bansal 2008)

Nervous system: Central nervous system toxicity, encephalopathy (Yamamoto 2002), fatigue (Mathews 2010), idiopathic intracranial hypertension (Morente 2015), migraine (Maghrabi 1998), progressive multifocal leukoencephalopathy (Aksamit 1995), reversible posterior leukoencephalopathy syndrome (Sood 2003)

Neuromuscular & skeletal: Lower extremity pain (possibly a manifestation of Calcineurin-inhibitor induced pain syndrome) (Lawson 2003), myopathy (Ding 2019)

Ophthalmic: Papilledema (Yu 2019)

Contraindications

Hypersensitivity to cyclosporine or any component of the formulation. IV cyclosporine is contraindicated in hypersensitivity to polyoxyethylated castor oil (Cremophor EL).

Rheumatoid arthritis and psoriasis patients with abnormal renal function, uncontrolled hypertension, or malignancies. Concomitant treatment with PUVA or UVB therapy, methotrexate, other immunosuppressive agents, coal tar, or radiation therapy are also contraindications for use in patients with psoriasis.

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

Canadian labeling: Additional contraindications (not in the US labeling): Concurrent use with bosentan; rheumatoid arthritis and psoriasis patients with primary or secondary immunodeficiency excluding autoimmune disease, uncontrolled infection, or malignancy (excluding non-melanoma skin cancer).

Warnings/Precautions

Disease-related concerns:

• Hepatic impairment: Cyclosporine has extensive hepatic metabolism and exposure is increased in patients with severe hepatic impairment. May require dose reduction.

• Psoriasis: Appropriate use: If receiving other immunosuppressive agents, radiation or UV therapy, concurrent use of cyclosporine is not recommended.

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 withoutsevere disease, hold cyclosporine 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]).

• Transplant recipients: Make dose adjustments based on blood concentrations; dependent on organ transplanted, time after transplant, organ function, and CsA toxicity.

Dosage form specific issues:

• Corn oil: Product may contain corn oil.

• Ethanol: Products may contain ethanol. The alcohol content should be considered in certain patient populations, including patients who are pregnant or breastfeeding, patients with liver disease, seizure disorders, alcohol dependency, or pediatric patients.

• Modified/nonmodified forms: Cyclosporine (modified) refers to the oral solution and capsule dosage formulations of cyclosporine in an aqueous dispersion (previously referred to as “microemulsion”).

• Polyoxyethylated castor oil: Cyclosporine for injection contains the vehicle polyoxyethylated castor oil (Cremophor EL), which is associated with hypersensitivity (anaphylactic) reactions. Due to the risk for anaphylaxis, IV cyclosporine should be reserved for use in patients unable to take an oral formulation.

• Propylene glycol: Some dosage forms may contain propylene glycol; large amounts are potentially toxic and have been associated with hyperosmolality, lactic acidosis, seizures, and respiratory depression; use caution (AAP 1997; Zar 2007).

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).

• Vaccines: Live, attenuated vaccines may be less effective; vaccination should be avoided.

Warnings: Additional Pediatric Considerations

Some dosage forms may contain propylene glycol; in neonates large amounts of propylene glycol delivered orally, intravenously (eg, >3,000 mg/day), or topically have been associated with potentially fatal toxicities which can include metabolic acidosis, seizures, renal failure, and CNS depression; toxicities have also been reported in children and adults including hyperosmolality, lactic acidosis, seizures and respiratory depression; use caution (AAP 1997; Shehab 2009).

Dosage Forms Considerations

Cyclosporine (modified): Gengraf and Neoral

Cyclosporine (non-modified): SandIMMUNE

Cyclosporine injection contains polyoxyethylated castor oil (Cremophor EL)

Dosage Forms: US

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

Capsule, Oral:

Gengraf: 25 mg, 100 mg [contains fd&c blue #2 (indigotine,indigo carmine), polyoxyl/peg-35 castor oil(cremophor el)]

Neoral: 25 mg, 100 mg [contains alcohol, usp]

SandIMMUNE: 25 mg, 100 mg

Generic: 25 mg, 50 mg, 100 mg

Solution, Intravenous:

SandIMMUNE: 50 mg/mL (5 mL) [contains alcohol, usp, polyoxyl/peg-35 castor oil(cremophor el)]

Generic: 50 mg/mL (5 mL)

Solution, Oral:

Gengraf: 100 mg/mL (50 mL) [contains propylene glycol]

Neoral: 100 mg/mL (50 mL) [contains alcohol, usp]

SandIMMUNE: 100 mg/mL (50 mL) [contains alcohol, usp]

Generic: 100 mg/mL (50 mL)

Generic Equivalent Available: US

Yes

Pricing: US

Capsules (cycloSPORINE Modified Oral)

25 mg (per each): $1.38

50 mg (per each): $2.74

100 mg (per each): $5.50

Capsules (cycloSPORINE Oral)

25 mg (per each): $3.85

100 mg (per each): $15.35 - $17.35

Capsules (Gengraf Oral)

25 mg (per each): $2.10

100 mg (per each): $8.40

Capsules (Neoral Oral)

25 mg (per each): $3.24

100 mg (per each): $12.86

Capsules (SandIMMUNE Oral)

25 mg (per each): $5.23

100 mg (per each): $20.68

Solution (cycloSPORINE Intravenous)

50 mg/mL (per mL): $9.39

Solution (cycloSPORINE Modified Oral)

100 mg/mL (per mL): $5.99

Solution (Gengraf Oral)

100 mg/mL (per mL): $9.49

Solution (Neoral Oral)

100 mg/mL (per mL): $14.00

Solution (SandIMMUNE Intravenous)

50 mg/mL (per mL): $16.10

Solution (SandIMMUNE Oral)

100 mg/mL (per mL): $20.05

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:

Neoral: 10 mg, 25 mg [contains alcohol, usp]

Neoral: 50 mg [contains alcohol, usp, fd&c blue #2 (indigo carm) aluminum lake]

Neoral: 100 mg [contains alcohol, usp]

Generic: 25 mg, 50 mg, 100 mg

Solution, Intravenous:

SandIMMUNE IV: 50 mg/mL (1 mL, 5 mL) [contains alcohol, usp, polyoxyl/peg-35 castor oil(cremophor el)]

Solution, Oral:

Neoral: 100 mg/mL (50 mL) [contains alcohol, usp, propylene glycol]

Administration: Adult

Oral solution: Do not administer liquid from plastic or styrofoam cup. May dilute Neoral oral solution with room temperature orange juice or apple juice. May dilute Sandimmune oral solution with milk, chocolate milk, or orange juice. Avoid changing diluents frequently. Mix thoroughly and drink at once. Use syringe provided to measure dose. Mix in a glass container and rinse container with more diluent to ensure total dose is taken. Do not rinse syringe before or after use (may cause dose variation).

Administer this medication consistently with relation to time of day and meals.

Combination therapy with renal or heart transplantation:

Everolimus: May administer cyclosporine at the same time as everolimus.

Sirolimus: The manufacturer recommends administering sirolimus 4 hours after cyclosporine. If given concurrently to facilitate adherence, additional monitoring may be required with initiation or dose adjustment of either agent.

IV: The manufacturer recommends that following dilution, IV admixture be administered over 2 to 6 hours. However, many transplant centers administer as divided doses (2 doses/day) or as a 24-hour continuous infusion.

Anaphylaxis has been reported with IV use; reserve for patients who cannot take oral form. Patients should be under continuous observation for at least the first 30 minutes of the infusion and should be monitored frequently thereafter. Maintain patent airway; other supportive measures and agents for treating anaphylaxis should be present when IV drug is given. To minimize leaching of DEHP, non-PVC sets should be used for administration. Cyclosporine levels should not be drawn from the infusion line in which cyclosporine is infused, as falsely elevated cyclosporine levels may occur if used for therapeutic drug monitoring; once a line is used for a cyclosporine IV infusion, it should be considered contaminated (Ref).

Administration: Pediatric

Oral: Administer consistently with relation to time of day and meals.

Oral solution: Use syringe provided to measure dose. Administer with oral syringe, glass dropper, or glass container (not plastic or styrofoam cup); to improve palatability, may mix Sandimmune oral solution with milk, chocolate milk, or orange juice that is at room temperature. May mix Neoral with orange juice or apple juice that is at room temperature; avoid mixing with grapefruit juice; Neoral mixed with milk can be unpalatable; mix in a glass container, stir well and drink at once; do not allow to stand before drinking; rinse container with more diluent to ensure that the total dose is taken. After use, dry outside of dosing syringe; do not rinse with water or other cleaning agents; if dosage syringe requires cleaning it must be completely dry before resuming use; introduction of water into the product will cause dose variations.

Parenteral: To minimize leaching of DEHP, non-PVC containers and sets should be used for administration. May administer by IV intermittent infusion or continuous infusion; for intermittent infusion, administer over 2 to 6 hours. Anaphylaxis has been reported with IV use. Patients should be continuously monitored for at least the first 30 minutes of the infusion and should be monitored frequently thereafter.

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.

Use: Labeled Indications

Cyclosporine modified:

Transplant rejection prophylaxis: Prophylaxis of organ rejection in kidney, liver, and heart transplants (commonly used in combination with an antiproliferative immunosuppressive agent and corticosteroid).

Psoriasis: Treatment of severe, recalcitrant plaque psoriasis in non-immunocompromised adults unresponsive to or unable to tolerate other systemic therapy

Cyclosporine non-modified:

Transplant rejection (prophylaxis): Prophylaxis of organ rejection in kidney, liver, and heart transplants (commonly used in combination with an antiproliferative agent and a corticosteroid)

Transplant rejection, chronic (treatment): May be used for the treatment of chronic rejection (kidney, liver, and heart) in patients previously treated with other immunosuppressive agents. Note: While approved for the treatment of chronic organ rejection, other therapies are clinically preferred in this setting.

Use: Off-Label: Adult

Aplastic anemia; Focal segmental glomerulosclerosis; Graft-versus-host disease, acute (prevention); Graft-versus-host disease, chronic (treatment); Immune thrombocytopenia, refractory; Lung transplant (prevention of acute rejection); Myasthenia gravis, chronic immunosuppressive therapy; T-cell large granular lymphocytic leukemia, symptomatic; Ulcerative colitis, severe refractory; Uveitis

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

CycloSPORINE may be confused with cycloPHOSphamide, Cyklokapron, cycloSERINE, voclosporin

CycloSPORINE modified (Neoral, Gengraf) may be confused with cycloSPORINE non-modified (SandIMMUNE)

Gengraf may be confused with Prograf

Neoral may be confused with Neurontin, Nizoral

SandIMMUNE may be confused with SandoSTATIN

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.

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; Inhibits BCRP/ABCG2, BSEP/ABCB11, CYP2C9 (weak), CYP3A4 (weak), OATP1B1/1B3 (SLCO1B1/1B3), P-glycoprotein/ABCB1

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.

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

AcetaZOLAMIDE: May increase the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Adalimumab: May decrease the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Afatinib: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Afatinib. Management: If combined, administer the P-gp inhibitor simultaneously with, or after, the dose of afatinib. Monitor closely for signs and symptoms of afatinib toxicity and if the combination is not tolerated, reduce the afatinib dose by 10 mg. Risk D: Consider therapy modification

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

Aliskiren: CycloSPORINE (Systemic) may increase the serum concentration of Aliskiren. Risk X: Avoid combination

Allopurinol: May increase the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Alpelisib: BCRP/ABCG2 Inhibitors may increase the serum concentration of Alpelisib. Management: Avoid coadministration of BCRP/ABCG2 inhibitors and alpelisib due to the potential for increased alpelisib concentrations and toxicities. If coadministration cannot be avoided, closely monitor for increased alpelisib adverse reactions. Risk D: Consider therapy modification

ALPRAZolam: CYP3A4 Inhibitors (Weak) may increase the serum concentration of ALPRAZolam. Risk C: Monitor therapy

Ambrisentan: CycloSPORINE (Systemic) may increase the serum concentration of Ambrisentan. Management: Limit ambrisentan dose to 5 mg daily and monitor for ambrisentan adverse reactions in patients receiving cyclosporine. Risk D: Consider therapy modification

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

Amphotericin B: May enhance the nephrotoxic effect of CycloSPORINE (Systemic). Amphotericin B may increase the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Androgens: May enhance the hepatotoxic effect of CycloSPORINE (Systemic). Androgens may increase the serum concentration of CycloSPORINE (Systemic). Management: Consider avoiding concomitant use of androgens and cyclosporine. If concomitant use is unavoidable, monitor serum cyclosporine concentrations and for signs and symptoms of hepatotoxicity. Cyclosporine dose reductions may be required. Risk D: Consider therapy modification

Antifungal Agents (Azole Derivatives, Systemic): May decrease the metabolism of CycloSPORINE (Systemic). Fluconazole and isavuconazonium considerations are addressed in separate monographs. Management: Consider reducing cyclosporine doses by 50% to 80% during coadministration with ketoconazole, 50% with voriconazole or itraconazole, and 25% with posaconazole. Cyclosporine dose reductions may be required with other azoles. Risk D: Consider therapy modification

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

Armodafinil: May decrease the serum concentration of CycloSPORINE (Systemic). 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

Ascorbic Acid: May decrease the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Asunaprevir: OATP1B1/1B3 (SLCO1B1/1B3) Inhibitors may increase the serum concentration of Asunaprevir. Risk X: Avoid combination

Atogepant: OATP1B1/1B3 (SLCO1B1/1B3) Inhibitors may increase the serum concentration of Atogepant. Management: For episodic migraine, the recommended atogepant dose is 10 mg or 30 mg once daily if given with OATP1B1/1B3 inhibitors. For chronic migraine, the recommended atogepant dose is 30 mg once daily with OATP1B1/1B3 inhibitors. Risk D: Consider therapy modification

Atorvastatin: CycloSPORINE (Systemic) may increase the serum concentration of Atorvastatin. Risk X: Avoid combination

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

Baricitinib: Immunosuppressants (Therapeutic Immunosuppressant Agents) may enhance the immunosuppressive effect of Baricitinib. 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

Berberine-Containing Products: May increase the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Berotralstat: CycloSPORINE (Systemic) may increase the serum concentration of Berotralstat. Berotralstat may increase the serum concentration of CycloSPORINE (Systemic). Management: Decrease the berotralstat dose to 110 mg daily when combined with cyclosporine. Additionally, monitor for increased cyclosporine concentrations and toxicities when these agents are combined. Risk D: Consider therapy modification

Bilastine: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Bilastine. Risk X: Avoid combination

Bosentan: CycloSPORINE (Systemic) may increase the serum concentration of Bosentan. Bosentan may decrease the serum concentration of CycloSPORINE (Systemic). Risk X: Avoid combination

Brincidofovir: OATP1B1/1B3 (SLCO1B1/1B3) Inhibitors may increase the serum concentration of Brincidofovir. Management: Consider alternatives to OATP1B/1B3 inhibitors in patients treated with brincidofovir. If coadministration is required, administer OATP1B1/1B3 inhibitors at least 3 hours after brincidofovir and increase monitoring for brincidofovir adverse reactions. Risk D: Consider therapy modification

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

Calcium Channel Blockers (Nondihydropyridine): May decrease the metabolism of CycloSPORINE (Systemic). CycloSPORINE (Systemic) may decrease the metabolism of Calcium Channel Blockers (Nondihydropyridine). Risk C: Monitor therapy

Caspofungin: CycloSPORINE (Systemic) may enhance the adverse/toxic effect of Caspofungin. CycloSPORINE (Systemic) may increase the serum concentration of Caspofungin. Caspofungin may increase the serum concentration of CycloSPORINE (Systemic). Management: Weigh potential benefits of caspofungin against a possible elevated risk of hepatotoxicity. Monitor liver function and re-evaluate treatment in patients with abnormal values. Mild transaminase elevations may occur relatively commonly. Risk D: Consider therapy modification

Celiprolol: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Celiprolol. 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 (Systemic): May increase the serum concentration of CycloSPORINE (Systemic). Management: Cyclosporine dose reductions will likely be required with initiation of concurrent chloramphenicol. Monitor cyclosporine concentrations and response closely following initiation and/or discontinuation of chloramphenicol. Risk D: Consider therapy modification

Chloroquine: May increase the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Cholic Acid: BSEP/ABCB11 Inhibitors may decrease the excretion of Cholic Acid. Management: Avoid the use of bile salt efflux pump inhibitors with cholic acid. If such a combination cannot be avoided, monitor serum transaminases (eg, AST, ALT) and bilirubin closely. Risk D: Consider therapy modification

Ciprofloxacin (Systemic): May enhance the nephrotoxic effect of CycloSPORINE (Systemic). Risk C: Monitor therapy

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

Clarithromycin: May increase the serum concentration of CycloSPORINE (Systemic). Management: Monitor for increased serum concentrations/toxic effects of cyclosporine if combined with clarithromycin. Cyclosporine dose reductions and/or prolongation of the dosing interval will likely be required. Risk D: Consider therapy modification

Clofazimine: May increase the serum concentration of CYP3A4 Substrates (High risk with Inhibitors). 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: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Colchicine. Colchicine distribution into certain tissues (e.g., brain) may also be increased. Management: This combination is often contraindicated, but combined use may be permitted with dose adjustment and monitoring. Recommendations vary based on brand, indication, use of CYP3A4 inhibitors, and hepatic/renal function. See interaction monograph for details. Risk D: Consider therapy modification

Colesevelam: May decrease the serum concentration of CycloSPORINE (Systemic). Management: Administer cyclosporine at least 4 hours prior to colesevelam. Monitor for decreased cyclosporine concentrations during concomitant colesevelam therapy. Risk D: Consider therapy modification

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

CycloPHOSphamide: May decrease the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

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

CYP3A4 Inducers (Strong): May decrease the serum concentration of CycloSPORINE (Systemic). Management: Monitor closely for reduced cyclosporine concentrations when combined with strong CYP3A4 inducers. Cyclosporine dose increases will likely be required to maintain adequate serum concentrations. Risk D: Consider therapy modification

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

CYP3A4 Inhibitors (Strong): May increase the serum concentration of CycloSPORINE (Systemic). Management: Monitor cyclosporine serum concentrations and clinical cyclosporine closely with concurrent use of any strong CYP3A4 inhibitor. Cyclosporine dose reductions and/or prolongation of the dosing interval will likely be required. Risk D: Consider therapy modification

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

Dabigatran Etexilate: P-glycoprotein/ABCB1 Inhibitors may increase serum concentrations of the active metabolite(s) of Dabigatran Etexilate. Risk C: Monitor therapy

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

Digoxin: CycloSPORINE (Systemic) may increase the serum concentration of Digoxin. Risk C: Monitor therapy

Disulfiram: May enhance the adverse/toxic effect of Products Containing Ethanol. Management: Do not use disulfiram with dosage forms that contain ethanol. Risk X: Avoid combination

Dofetilide: CYP3A4 Inhibitors (Weak) may increase the serum concentration of Dofetilide. Risk C: Monitor therapy

DOXOrubicin (Conventional): P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of DOXOrubicin (Conventional). Risk X: Avoid combination

DOXOrubicin (Liposomal): P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of DOXOrubicin (Liposomal). Risk C: Monitor therapy

DroNABinol: May increase the serum concentration of CycloSPORINE (Systemic). Specifically, dronabinol may displace cyclosporine from its protein-binding sites, leading to an increased concentration of active, unbound drug. Risk C: Monitor therapy

Dronedarone: CycloSPORINE (Systemic) may increase the serum concentration of Dronedarone. Risk X: Avoid combination

Edoxaban: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Edoxaban. 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

Elagolix: OATP1B1/1B3 (SLCO1B1/1B3) Inhibitors may increase the serum concentration of Elagolix. Risk X: Avoid combination

Elagolix, Estradiol, and Norethindrone: OATP1B1/1B3 (SLCO1B1/1B3) Inhibitors may increase the serum concentration of Elagolix, Estradiol, and Norethindrone. Specifically, concentrations of elagolix may be increased. Risk X: Avoid combination

Elbasvir and Grazoprevir: OATP1B1/1B3 (SLCO1B1/1B3) Inhibitors may increase the serum concentration of Elbasvir and Grazoprevir. Risk X: Avoid combination

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

Eltrombopag: CycloSPORINE (Systemic) may decrease the serum concentration of Eltrombopag. Risk C: Monitor therapy

Eluxadoline: OATP1B1/1B3 (SLCO1B1/1B3) Inhibitors may increase the serum concentration of Eluxadoline. Management: Decrease the eluxadoline dose to 75 mg twice daily if combined with OATP1B1/1B3 inhibitors and monitor patients for increased eluxadoline effects/toxicities. Risk D: Consider therapy modification

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

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

Ethinyl Estradiol-Containing Products: May increase the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Etoposide: CycloSPORINE (Systemic) may decrease the metabolism of Etoposide. Management: Consider reducing the dose of etoposide by 50% if the patient is receiving, or has recently received, cyclosporine. Monitor for increased toxic effects of etoposide if cyclosporine is initiated, the dose is increased, or it has been recently discontinued. Risk D: Consider therapy modification

Etoposide Phosphate: CycloSPORINE (Systemic) may increase the serum concentration of Etoposide Phosphate. CycloSPORINE may decrease the metabolism, via CYP isoenzymes, and decrease the p-glycoprotein-mediated elimination of Etoposide Phosphate. Management: Consider empiric etoposide phosphate dose reductions of at least 50% in patients receiving, or who have recently received, cyclosporine. Monitor closely for increased toxicity of etoposide phosphate if cyclosporine is initiated or dose changed. Risk D: Consider therapy modification

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

Everolimus: CycloSPORINE (Systemic) may increase the serum concentration of Everolimus. Management: When everolimus is used as a post-transplant immunosuppressant (Zortress brand), lower cyclosporine doses and lower target serum cyclosporine concentrations must be used. Monitor both everolimus and cyclosporine serum concentrations closely when combined. Risk D: Consider therapy modification

Ezetimibe: May increase the serum concentration of CycloSPORINE (Systemic). CycloSPORINE (Systemic) may increase the serum concentration of Ezetimibe. Risk C: Monitor therapy

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

Fibric Acid Derivatives: CycloSPORINE (Systemic) may enhance the nephrotoxic effect of Fibric Acid Derivatives. Fibric Acid Derivatives may decrease the serum concentration of CycloSPORINE (Systemic). Management: Careful consideration of the risks and benefits should be undertaken prior to use of this combination; extra monitoring of renal function and cyclosporine concentrations will likely be required. Adjustment of cyclosporine dose may be necessary. Risk D: Consider therapy modification

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

Fimasartan: CycloSPORINE (Systemic) may increase the serum concentration of Fimasartan. Risk C: Monitor therapy

Finerenone: CYP3A4 Inhibitors (Weak) may increase the serum concentration of Finerenone. Risk C: Monitor therapy

Flibanserin: CYP3A4 Inhibitors (Weak) may increase the serum concentration of Flibanserin. Risk C: Monitor therapy

Fluconazole: May increase the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Fluvastatin: CycloSPORINE (Systemic) may increase the serum concentration of Fluvastatin. Management: Limit fluvastatin to 20 mg twice daily and avoid use of fluvastatin extended-release tablets in patients who are also receiving cyclosporine and monitor for fluvastatin toxicities (eg, myalgia, myopathy, rhabdomyolysis). Risk D: Consider therapy modification

Foscarnet: May enhance the nephrotoxic effect of CycloSPORINE (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-Valganciclovir: May enhance the nephrotoxic effect of CycloSPORINE (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

Glecaprevir and Pibrentasvir: CycloSPORINE (Systemic) may increase the serum concentration of Glecaprevir and Pibrentasvir. Management: Glecaprevir/pibrentasvir is not recommended for use in patients requiring stable doses of cyclosporine greater than 100 mg per day. If combined with lower doses of cyclosporine, monitor for increased glecaprevir/pibrentasvir toxicities. Risk D: Consider therapy modification

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

GlyBURIDE: CycloSPORINE (Systemic) may diminish the therapeutic effect of GlyBURIDE. GlyBURIDE may increase the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Grape Juice, Purple: May decrease the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Grapefruit Juice: May decrease the metabolism of CycloSPORINE (Systemic). Management: Monitor for altered cyclosporine concentrations/effects if grapefruit intake is increased/decreased. Advise patients to not alter their pattern of grapefruit/grapefruit juice intake without consulting their healthcare provider. Risk X: Avoid combination

Griseofulvin: May decrease the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Hydroxychloroquine: May increase the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Ilaprazole: May increase the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Imipenem: May enhance the neurotoxic effect of CycloSPORINE (Systemic). 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

Interleukin-6 (IL-6) Inhibiting Therapies: May decrease the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Ixabepilone: CYP3A4 Inhibitors (Weak) may increase the serum concentration of Ixabepilone. Risk C: Monitor therapy

Lapatinib: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Lapatinib. Risk C: Monitor therapy

Larotrectinib: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Larotrectinib. Risk C: Monitor therapy

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

Lefamulin: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Lefamulin. Management: Avoid concomitant use of lefamulin tablets with P-glycoprotein/ABCB1 inhibitors. If concomitant use is required, monitor for lefamulin adverse effects. Risk D: Consider therapy modification

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

Lemborexant: CYP3A4 Inhibitors (Weak) may increase the serum concentration of Lemborexant. Management: The maximum recommended dosage of lemborexant is 5 mg, no more than once per night, when coadministered with weak CYP3A4 inhibitors. Risk D: Consider therapy modification

Lercanidipine: May increase the serum concentration of CycloSPORINE (Systemic). CycloSPORINE (Systemic) may increase the serum concentration of Lercanidipine. Risk X: Avoid combination

Letermovir: May increase the serum concentration of CycloSPORINE (Systemic). CycloSPORINE (Systemic) may increase the serum concentration of Letermovir. Management: Decrease the letermovir dose to 240 mg daily when combined with cyclosporine. Additionally, monitor for increased cyclosporine concentrations and toxicities when combined with letermovir. Risk D: Consider therapy modification

Linezolid: CycloSPORINE (Systemic) may increase the serum concentration of Linezolid. Risk C: Monitor therapy

Lomitapide: CYP3A4 Inhibitors (Weak) may increase the serum concentration of Lomitapide. Management: Patients on lomitapide 5 mg/day may continue that dose. Patients taking lomitapide 10 mg/day or more should decrease the lomitapide dose by half. The lomitapide dose may then be titrated up to a max adult dose of 30 mg/day. Risk D: Consider therapy modification

Lonafarnib: CYP3A4 Inhibitors (Weak) may increase the serum concentration of Lonafarnib. Management: Avoid concurrent use of lonafarnib with weak CYP3A4 inhibitors. If concurrent use is unavoidable, reduce the lonafarnib dose to or continue at a dose of 115 mg/square meter. Monitor for evidence of arrhythmia, syncope, palpitations, or similar effects. Risk D: Consider therapy modification

Loop Diuretics: CycloSPORINE (Systemic) may enhance the adverse/toxic effect of Loop Diuretics. Specifically, the risk for hyperuricemia and gout may be increased. Risk C: Monitor therapy

Lovastatin: CycloSPORINE (Systemic) may increase the serum concentration of Lovastatin. Risk X: Avoid combination

Lumateperone: CycloSPORINE (Systemic) may increase the serum concentration of Lumateperone. Risk C: Monitor therapy

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

Melphalan: May enhance the nephrotoxic effect of CycloSPORINE (Systemic). Risk C: Monitor therapy

Melphalan Flufenamide: May enhance the nephrotoxic effect of CycloSPORINE (Systemic). Risk C: Monitor therapy

Methotrexate: CycloSPORINE (Systemic) may increase the serum concentration of Methotrexate. This may result in nausea, vomiting, oral ulcers, hepatotoxicity and/or nephrotoxicity. Methotrexate may increase the serum concentration of CycloSPORINE (Systemic). This may result in nephrotoxicity. Risk C: Monitor therapy

Methotrimeprazine: Products Containing Ethanol may enhance the adverse/toxic effect of Methotrimeprazine. Specifically, a disulfiram-like reaction may occur and CNS depressant effects may be increased. Management: Avoid products containing alcohol in patients treated with methotrimeprazine. Risk X: Avoid combination

MethylPREDNISolone: CycloSPORINE (Systemic) may enhance the neuroexcitatory and/or seizure-potentiating effect of MethylPREDNISolone. MethylPREDNISolone may decrease the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Metoclopramide: May increase the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Midazolam: CYP3A4 Inhibitors (Weak) may increase the serum concentration of Midazolam. Risk C: Monitor therapy

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

MiFEPRIStone: May increase the serum concentration of CycloSPORINE (Systemic). Risk X: Avoid combination

Minoxidil (Systemic): CycloSPORINE (Systemic) may enhance the adverse/toxic effect of Minoxidil (Systemic). Severe hypertrichosis has been reported with this combination. Risk C: Monitor therapy

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

MitoXANTRONE: CycloSPORINE (Systemic) may increase the serum concentration of MitoXANTRONE. Risk C: Monitor therapy

Momelotinib: OATP1B1/1B3 (SLCO1B1/1B3) Inhibitors may increase the serum concentration of Momelotinib. Risk C: Monitor therapy

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

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

Multivitamins/Fluoride (with ADE): May increase the serum concentration of CycloSPORINE (Systemic). Multivitamins/Fluoride (with ADE) may decrease the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Multivitamins/Minerals (with ADEK, Folate, Iron): May increase the serum concentration of CycloSPORINE (Systemic). Multivitamins/Minerals (with ADEK, Folate, Iron) may decrease the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Multivitamins/Minerals (with AE, No Iron): May increase the serum concentration of CycloSPORINE (Systemic). Multivitamins/Minerals (with AE, No Iron) may decrease the serum concentration of CycloSPORINE (Systemic). 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

Mycophenolate: CycloSPORINE (Systemic) may decrease the serum concentration of Mycophenolate. Specifically, cyclosporine may decrease concentrations of the active metabolite mycophenolic acid. Management: Mycophenolate requirements may be greater in patients receiving cyclosporine. Monitor mycophenolate dosing and response to therapy particularly closely when adjusting concurrent cyclosporine (starting, stopping, or changing dose). Risk D: Consider therapy modification

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

Nadolol: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Nadolol. Risk C: Monitor therapy

Naldemedine: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Naldemedine. Risk C: Monitor therapy

Naloxegol: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Naloxegol. Risk C: Monitor therapy

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

Neuromuscular-Blocking Agents: CycloSPORINE (Systemic) may enhance the neuromuscular-blocking effect of Neuromuscular-Blocking Agents. Risk C: Monitor therapy

NiMODipine: CYP3A4 Inhibitors (Weak) may increase the serum concentration of NiMODipine. Risk C: Monitor therapy

Nirmatrelvir and Ritonavir: May increase the serum concentration of CycloSPORINE (Systemic). Management: Consider avoiding this combination if possible and using alternative anti-COVID-19 therapy if appropriate. If coadministration is required, consider reducing cyclosporine dose by 80% and monitoring cyclosporine concentrations 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 CycloSPORINE (Systemic). Nonsteroidal Anti-Inflammatory Agents may increase the serum concentration of CycloSPORINE (Systemic). CycloSPORINE (Systemic) may increase the serum concentration of Nonsteroidal Anti-Inflammatory Agents. Management: Consider alternatives to nonsteroidal anti-inflammatory agents (NSAIDs). Monitor for evidence of nephrotoxicity, as well as increased serum cyclosporine concentrations and systemic effects (eg, hypertension) during concomitant therapy with NSAIDs. Risk D: Consider therapy modification

Nonsteroidal Anti-Inflammatory Agents (Topical): May enhance the adverse/toxic effect of CycloSPORINE (Systemic). Specifically, the nephrotoxicity of cyclosporine (systemic) may be increased. Risk C: Monitor therapy

Norethindrone: May increase the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Norfloxacin: May increase the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Obeticholic Acid: BSEP/ABCB11 Inhibitors may increase serum concentrations of the active metabolite(s) of Obeticholic Acid. Management: Avoid concomitant use of obeticholic acid and bile salt efflux pump (BSEP) inhibitors if possible. If concomitant therapy is necessary, monitor patients for elevated liver transaminases and elevated bilirubin. Risk D: Consider therapy modification

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

Ombitasvir, Paritaprevir, and Ritonavir: May increase the serum concentration of CycloSPORINE (Systemic). Management: Reduce cyclosporine dose by 80% when initiating therapy with ombitasvir/paritaprevir/ritonavir and monitor cyclosporine blood levels closely. Risk D: Consider therapy modification

Ombitasvir, Paritaprevir, Ritonavir, and Dasabuvir: May increase the serum concentration of CycloSPORINE (Systemic). Management: Reduce cyclosporine dose 80% when initiating therapy with ombitasvir/paritaprevir/ritonavir/dasabuvir and monitor cyclosporine blood levels closely. Risk D: Consider therapy modification

Omeprazole: May increase the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Orlistat: May decrease the serum concentration of CycloSPORINE (Systemic). Management: Administer oral cyclosporine 3 hours after orlistat. Monitor for decreased serum concentrations of oral cyclosporine, even with the recommended dose separation. Risk D: Consider therapy modification

Ornidazole: May enhance the adverse/toxic effect of Products Containing Ethanol. Specifically, a disulfiram-like reaction may occur. 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

PAZOPanib: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of PAZOPanib. Risk X: Avoid combination

PAZOPanib: BCRP/ABCG2 Inhibitors may increase the serum concentration of PAZOPanib. Risk X: Avoid combination

P-glycoprotein/ABCB1 Inducers: May decrease the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

P-glycoprotein/ABCB1 Inhibitors: May increase the serum concentration of CycloSPORINE (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

Pimozide: CYP3A4 Inhibitors (Weak) may increase the serum concentration of Pimozide. Risk X: Avoid combination

Pitavastatin: CycloSPORINE (Systemic) may increase the serum concentration of Pitavastatin. Risk X: Avoid combination

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

Potassium-Sparing Diuretics: May enhance the hyperkalemic effect of CycloSPORINE (Systemic). Risk X: Avoid combination

Pralsetinib: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Pralsetinib. Management: If this combo cannot be avoided, decrease pralsetinib dose from 400 mg daily to 300 mg daily; from 300 mg daily to 200 mg daily; and from 200 mg daily to 100 mg daily. Risk D: Consider therapy modification

Pravastatin: CycloSPORINE (Systemic) may increase the serum concentration of Pravastatin. Pravastatin may increase the serum concentration of CycloSPORINE (Systemic). Management: Initiate pravastatin dose at 10 mg daily and limit the maximum pravastatin dose to 20 mg daily in patients who are also receiving cyclosporine. Monitor for pravastatin toxicities (eg, myalgia, myopathy, rhabdomyolysis) if combined. Risk D: Consider therapy modification

PrednisoLONE (Systemic): CycloSPORINE (Systemic) may enhance the neuroexcitatory and/or seizure-potentiating effect of PrednisoLONE (Systemic). PrednisoLONE (Systemic) may decrease the serum concentration of CycloSPORINE (Systemic). CycloSPORINE (Systemic) may increase the serum concentration of PrednisoLONE (Systemic). Risk C: Monitor therapy

PredniSONE: CycloSPORINE (Systemic) may enhance the neuroexcitatory and/or seizure-potentiating effect of PredniSONE. CycloSPORINE (Systemic) may increase serum concentrations of the active metabolite(s) of PredniSONE. PredniSONE may decrease the serum concentration of CycloSPORINE (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

Probucol: May decrease the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Pyrazinamide: May enhance the myopathic (rhabdomyolysis) effect of CycloSPORINE (Systemic). Pyrazinamide may decrease the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Quercetin: May increase the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Quinupristin and Dalfopristin: May increase the serum concentration of CycloSPORINE (Systemic). 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

Ranolazine: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Ranolazine. Risk C: Monitor therapy

Red Yeast Rice: CycloSPORINE (Systemic) may increase the serum concentration of Red Yeast Rice. Risk X: Avoid combination

Relugolix: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Relugolix. Management: Avoid coadministration of relugolix with oral P-gp inhibitors whenever possible. If combined, take relugolix at least 6 hours prior to the P-gp inhibitor and monitor patients more frequently for adverse reactions. Risk D: Consider therapy modification

Relugolix, Estradiol, and Norethindrone: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Relugolix, Estradiol, and Norethindrone. Management: Avoid use of relugolix/estradiol/norethindrone with P-glycoprotein (P-gp) inhibitors. If concomitant use is unavoidable, relugolix/estradiol/norethindrone should be administered at least 6 hours before the P-gp inhibitor. Risk D: Consider therapy modification

Repaglinide: CycloSPORINE (Systemic) may increase the serum concentration of Repaglinide. Management: Limit the daily repaglinide dose to a maximum of 6 mg with concurrent use of cyclosporine, and monitor closely for increased repaglinide effects. Risk D: Consider therapy modification

Repotrectinib: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Repotrectinib. Risk X: Avoid combination

Revefenacin: OATP1B1/1B3 (SLCO1B1/1B3) Inhibitors may increase serum concentrations of the active metabolite(s) of Revefenacin. Risk X: Avoid combination

RifAXIMin: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of RifAXIMin. Risk C: Monitor therapy

Rimegepant: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Rimegepant. Management: Avoid administration of another dose of rimegepant within 48 hours if given concomitantly with a P-glycoprotein (P-gp) inhibitor. Risk D: Consider therapy modification

RisperiDONE: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of RisperiDONE. Risk C: Monitor therapy

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

RomiDEPsin: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of RomiDEPsin. Risk C: Monitor therapy

Rosuvastatin: CycloSPORINE (Systemic) may increase the serum concentration of Rosuvastatin. Management: Limit rosuvastatin to 5 mg daily in patients who are also receiving cyclosporine, and monitor patients for increased rosuvastatin toxicities. Canadian labeling contraindicates concomitant use of rosuvastatin with cyclosporine. Risk D: Consider therapy modification

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

Secnidazole: Products Containing Ethanol may enhance the adverse/toxic effect of Secnidazole. Risk X: Avoid combination

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

Silodosin: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Silodosin. Risk C: Monitor therapy

Simeprevir: May increase the serum concentration of CycloSPORINE (Systemic). CycloSPORINE (Systemic) may increase the serum concentration of Simeprevir. Risk X: Avoid combination

Simvastatin: CycloSPORINE (Systemic) may increase the serum concentration of Simvastatin. Risk X: Avoid combination

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 (Conventional): May enhance the adverse/toxic effect of CycloSPORINE (Systemic). An increased risk of calcineurin inhibitor-induced hemolytic uremic syndrome/thrombotic thrombocytopenic purpura/thrombotic microangiopathy (HUS/TTP/TMA) has been described. CycloSPORINE (Systemic) may increase the serum concentration of Sirolimus (Conventional). This is of specific concern with cyclosporine [MODIFIED]. Management: Administer oral doses of sirolimus 4 hours after doses of cyclosporine. Monitor for toxic effects of sirolimus if used with cyclosporine. Risk D: Consider therapy modification

Sirolimus (Protein Bound): P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Sirolimus (Protein Bound). Risk X: Avoid combination

Somatostatin Analogs: May decrease the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Sparsentan: May increase the serum concentration of CycloSPORINE (Systemic). CycloSPORINE (Systemic) may increase the serum concentration of Sparsentan. Risk X: Avoid combination

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

St John's Wort: May decrease the serum concentration of CycloSPORINE (Systemic). Management: Consider alternatives to St. John's wort (SJW). If the combination cannot be avoided, monitor for decreased cyclosporine concentrations/effects. Monitor for increased cyclosporine concentrations/effects following SJW discontinuation. Risk D: Consider therapy modification

Sulfinpyrazone: May decrease the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Sulfonamide Antibiotics: May enhance the nephrotoxic effect of CycloSPORINE (Systemic). Sulfonamide Antibiotics may decrease the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

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

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

Talazoparib: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Talazoparib. Risk C: Monitor therapy

Talazoparib: BCRP/ABCG2 Inhibitors may increase the serum concentration of Talazoparib. Risk C: Monitor therapy

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: OATP1B1/1B3 (SLCO1B1/1B3) Inhibitors may increase the serum concentration of Taurursodiol. Risk X: Avoid combination

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

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

Tegaserod (Withdrawn from US Market): P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Tegaserod (Withdrawn from US Market). Risk C: Monitor therapy

Temsirolimus: May enhance the adverse/toxic effect of CycloSPORINE (Systemic). An increased risk of calcineurin inhibitor-induced hemolytic uremic syndrome/thrombotic thrombocytopenic purpura/thrombotic microangiopathy (HUS/TTP/TMA) has been described with concomitant sirolimus use. Risk C: Monitor therapy

Teniposide: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Teniposide. Risk C: Monitor therapy

Tenofovir Disoproxil Fumarate: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Tenofovir Disoproxil Fumarate. Risk C: Monitor therapy

Terbinafine (Systemic): May decrease the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

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

Ticagrelor: CycloSPORINE (Systemic) may increase the serum concentration of Ticagrelor. Risk C: Monitor therapy

Ticlopidine: May decrease the serum concentration of CycloSPORINE (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

TOLBUTamide: CYP2C9 Inhibitors (Weak) may increase the serum concentration of TOLBUTamide. Risk C: Monitor therapy

Topotecan: BCRP/ABCG2 Inhibitors may increase the serum concentration of Topotecan. Risk X: Avoid combination

Topotecan: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Topotecan. Risk X: Avoid combination

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

Triazolam: CYP3A4 Inhibitors (Weak) may increase the serum concentration of Triazolam. Risk C: Monitor therapy

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

Ubrogepant: CycloSPORINE (Systemic) may increase the serum concentration of Ubrogepant. Management: Use an initial ubrogepant dose of 50 mg and consider avoiding a second dose for 24 hours when used with cyclosporine. Risk D: Consider therapy modification

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

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

Venetoclax: P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of Venetoclax. Management: Reduce the venetoclax dose by at least 50% in patients requiring concomitant treatment with P-glycoprotein (P-gp) inhibitors. Resume the previous venetoclax dose 2 to 3 days after discontinuation of a P-gp inhibitor. Risk D: Consider therapy modification

VinCRIStine (Liposomal): P-glycoprotein/ABCB1 Inhibitors may increase the serum concentration of VinCRIStine (Liposomal). Risk X: Avoid combination

Vitamin E (Systemic): May increase the serum concentration of CycloSPORINE (Systemic). Vitamin E (Systemic) may decrease the serum concentration of CycloSPORINE (Systemic). Risk C: Monitor therapy

Voxilaprevir: OATP1B1/1B3 (SLCO1B1/1B3) Inhibitors may increase the serum concentration of Voxilaprevir. Risk X: Avoid combination

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

Zavegepant: OATP1B1/1B3 (SLCO1B1/1B3) Inhibitors may increase the serum concentration of Zavegepant. Risk X: Avoid combination

Food Interactions

Grapefruit juice increases cyclosporine serum concentrations. Management: Avoid grapefruit juice with concomitant oral cyclosporine use.

Reproductive Considerations

Cyclosporine is an acceptable immunosuppressant for use in kidney (EBPG 2002; KDIGO 2009; López 2014), liver (AASLD [Lucey 2013]), or heart (ISHLT [Velleca 2022]) transplant recipients planning a pregnancy. Conception may be considered for patients on a stable/low maintenance dose for ≥1 year following transplant (AASLD [Lucey 2013]; EBPG 2002; ISHLT [Velleca 2022]; López 2014).

Cyclosporine is considered acceptable for use in patients with rheumatic and musculoskeletal diseases who are planning to become pregnant and are not able to use alternative therapies; however, blood pressure monitoring is recommended. Conception should be planned during a period of quiescent/low disease activity (ACR [Sammaritano 2020]). Patients treated with cyclosporine for lupus nephritis should continue treatment while planning to become pregnant; conception may be considered after 6 months of inactive disease (EULAR/ERA-EDTA [Bertsias 2012]).

Data related to paternal use of cyclosporine are limited. However, available data have not shown cyclosporine adversely impacts male fertility or increases the risk of adverse pregnancy outcomes when used prior to conception (Mouyis 2019). Potential effects on fertility are associated with higher doses; conception can be attempted once lower serum levels can be maintained and the allograft is functioning (Georgiou 2016). Erectile dysfunction was found to occur more frequently with cyclosporine than other immunosuppressants in a study of renal transplant recipients; consider screening patients with risk factors prior to use (Zakhem 2019). Cyclosporine is considered acceptable for use in patients with rheumatic and musculoskeletal diseases who are planning to father a child (ACR [Sammaritano 2020])

Pregnancy Considerations

Cyclosporine crosses the placenta (Claris 1993).

In a study of 15 pregnant patients, maternal concentrations did not correlate with those found in the umbilical cord (n=14). Cyclosporine was detected in the serum of one newborn for several days after birth (Claris 1993).

Cyclosporine is not associated with specific teratogenic effects, but maternal use may be associated with an increased risk of intrauterine growth restriction, small for gestational age babies, maternal hypertension, and preeclampsia (EBPG 2002). Premature births and low birth weight were consistently observed in pregnant transplant recipients (additional pregnancy complications also present). In utero exposure to cyclosporine has not been found to influence renal function or blood pressure in children followed up to 7 years of age (limited data).

Some formulations may contain alcohol; the alcohol content should be taken into consideration prior to prescribing to patients who are pregnant.

Cyclosporine levels decline during pregnancy (KDIGO 2009) and increased monitoring is recommended (AASLD [Lucey 2013]; EBPG 2002; ISHLT [Velleca 2022]; López 2014).

Cyclosporine is an acceptable immunosuppressant for use in patients who become pregnant following a kidney (EBPG 2002; López 2014) or heart transplant (ISHLT [Velleca 2022]). Cyclosporine may also be used when needed in pregnant patients following a liver transplant (AASLD [Lucey 2013]).

For other indications, cyclosporine is not a preferred agent. Cyclosporine is considered acceptable for the treatment of myasthenia gravis in pregnant patients who are not controlled with or unable to tolerate corticosteroids (Sanders 2016). If therapy is needed for psoriasis, other agents are preferred; however, if cyclosporine is used, limit to shortest duration and lowest possible dose (Kaushik 2019). Cyclosporine can be used during pregnancy for refractory cases of lupus nephritis (EULAR/ERA-EDTA [Berstias 2012]) and other rheumatic and musculoskeletal diseases in patients who are not able to use alternative therapies; however, close monitoring of blood pressure is recommended (ACR [Sammaritano 2020]). Cyclosporine may be useful for the treatment of immune thrombocytopenia in pregnant patients who are refractory to preferred agents (Provan 2019). Use of cyclosporine for inflammatory bowel disease is limited to salvage therapy in patients who are pregnant (AGA [Mahadevan 2019]).

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

Breastfeeding Considerations

Cyclosporine is present in breast milk.

Concentrations of cyclosporine in milk vary widely (ACR [Sammaritano 2020]).

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 mother.

Recommendations for breastfeeding in patients taking cyclosporine following a kidney transplant differ; generally breastfeeding may be considered with maternal use of maintenance doses (Constantinescu 2014; EBPG 2002; KDIGO 2009; López 2014). Cyclosporine is considered compatible for use in patients with inflammatory bowel disease who wish to breastfeed (AGA [Mahadevan 2019]). Cyclosporine may be continued or initiated in patients with rheumatic and musculoskeletal diseases who are breastfeeding. Infants should be closely monitored. Infant drug levels should be measured if adverse events such as recurrent infections occur (ACR [Sammaritano 2020]). Some formulations may contain alcohol which may be present in breast milk; the alcohol content should be taken into consideration prior to prescribing to a breastfeeding mother.

Dietary Considerations

Avoid grapefruit juice with oral cyclosporine use.

Monitoring Parameters

Monitor cyclosporine plasma concentrations, renal function (serum creatinine and BUN; especially with concomitant use of other nephrotoxic drugs), and BP periodically and following the addition, modification, or deletion of other medications. Monitor for hypersensitivity reactions (IV cyclosporine). Monitor for signs/symptoms of hepatotoxicity, secondary malignancy, diabetes mellitus, infection, progressive multifocal leukoencephalopathy (eg, hemiparesis, apathy, confusion, cognitive deficiencies, ataxia). Monitor for progressive cognitive or motor deficits; magnetic resonance imaging may be required for diagnosis of posterior reversible encephalopathy syndrome (PRES).

Nephrotic syndrome (Canadian labeling): Baseline BP (2 readings within 2 weeks), fasting serum creatinine (at least 3 levels within 2 weeks), creatinine clearance, urinalysis, CBC, liver function, serum uric acid, serum potassium, and malignancy screening (eg, skin, mouth, lymph nodes). Biweekly monitoring of BP for initial 3 months and then monthly thereafter, frequent monitoring of renal function and periodic cyclosporine trough levels are recommended during therapy. Consider renal biopsy in patients with steroid-dependent minimal change nephropathy who have been maintained on therapy >1 year.

Transplant recipients: Cyclosporine trough levels, serum electrolytes, renal function, hepatic function, blood pressure, lipid profile, blood sugar, and HbA1c

Psoriasis therapy: Baseline BP, serum creatinine (2 levels each), BUN, CBC, serum magnesium, potassium, uric acid, lipid profile. Every other week monitoring of BP, CBC, serum creatinine, and levels of BUN, uric acid, potassium, lipids, and magnesium during the first 3 months of treatment for psoriasis. Monthly monitoring is recommended after this initial period. (Note: The Canadian labeling recommends bimonthly monitoring of serum creatinine after the initial period if serum creatinine remains stable and cyclosporine dose is ≤2.5 mg/kg daily, and monthly monitoring for higher doses). Also evaluate any atypical skin lesions prior to therapy. Increase the frequency of BP monitoring after each alteration in dosage of cyclosporine. Consider test for tuberculosis (TB) infection (latent TB) (AAD/NPF [Menter 2020]).

Reference Range

Reference ranges are method dependent and specimen dependent; use the same analytical method consistently; carefully consider the laboratory method used for cyclosporine concentration to determine target level required for specific indication.

Method-dependent and specimen-dependent: Trough levels should be obtained:

Oral: 12 hours after dose or immediately prior to next dose (chronic usage).

IV: 12 hours after dose or immediately prior to next dose.

Therapeutic trough range in solid organ transplant (kidney, liver, heart): Not absolutely defined, dependent on organ transplanted, time after transplant, organ function, concurrent immunosuppression, center-specific protocol, and CsA toxicity:

General range of 100 to 400 ng/mL (SI: 83.2 to 332.8 nmol/L); refer to institutional protocol for specific therapeutic ranges.

Toxic level: Not well defined, nephrotoxicity may occur at any level.

Recommended cyclosporine therapeutic ranges when administered in combination with an mTOR inhibitor (eg, everolimus) for heart transplant (ISHLT [Velleca 2022 ]).

3 to 6 months post-transplant: 75 to 200 ng/mL (SI: 62.4 to 166.4 nmol/L).

>6 months post-transplant: 50 to 100 ng/mL (SI: 41.6 to 83.2 nmol/L).

Recommended cyclosporine therapeutic ranges when administered in combination with everolimus for renal transplant (Zortress product labeling 2018):

Month 1 post-transplant: 100 to 200 ng/mL (SI: 83.2 to 166.4 nmol/L).

Months 2 and 3 post-transplant: 75 to 150 ng/mL (SI: 62.4 to 124.8 nmol/L).

Months 4 and 5 post-transplant: 50 to 100 ng/mL (SI: 41.6 to 83.2 nmol/L).

Months 6 to 12 post-transplant: 25 to 50 ng/mL (SI: 20.8 to 41.6 nmol/L).

Alternative monitoring (renal transplant): Two-hour post cyclosporine dose level (obtaining a level 2 hours after administration correlates well with drug exposure throughout the 12-hour dosing interval [Schiff 2007]).

IL-2 induction therapy (2-hour post cyclosporine dose level):

Month 1 and 2: >1,500 ng/mL (SI: >1,248 nmol/L).

Month 3: 1,200 to 1,400 ng/mL (SI: 998.4 to 1,164.8 nmol/L).

Month 4 to 12: 600 to 1,000 ng/mL (SI: 499.2 to 832 nmol/L).

Month >12: ~800 ng/mL (SI: ~665.6 nmol/L).

Antithymocyte globulin (rabbit) induction therapy (2-hour post cyclosporine dose level):

Month 1 to 3: 1,000 to 1,200 ng/mL (SI: 832 to 998.4 nmol/L).

Month 4 to 12: 600 to 1,000 ng/mL (SI: 499.2 to 832 nmol/L).

Month >12: ~800 ng/mL (SI: ~665.6 nmol/L).

Lung transplant (off-label use): Target cyclosporine levels were maintained between 250 and 350 ng/mL (SI: 208 to 291.2 nmol/L) during the first month, and around 200 ng/mL (SI: 166.4 nmol/L) thereafter, depending on renal function (Zuckermann 2003).

Nephrotic syndrome (off-label use): Target trough concentration (pediatrics): 80 to 100 ng/mL (SI: 66.6 to 83.2 nmol/L) (Gellermann 2013). Target trough levels may vary based on protocol; point in therapy, and/or treatment goals.

Mechanism of Action

Inhibition of production and release of interleukin II and inhibits interleukin II-induced activation of resting T-lymphocytes.

Pharmacokinetics (Adult Data Unless Noted)

Absorption: Oral:

Cyclosporine (non-modified): Erratic and incomplete; dependent on presence of food, bile acids, and GI motility; larger oral doses are needed in pediatrics due to shorter bowel length and limited intestinal absorption

Cyclosporine (modified): Erratic and incomplete; increased absorption, up to 30% when compared to cyclosporine (non-modified); less dependent on food, bile acids, or GI motility when compared to cyclosporine (non-modified)

Distribution: Widely in tissues and body fluids including the liver, pancreas, and lungs

Vdss: 4-6 L/kg in renal, liver, and marrow transplant recipients (slightly lower values in cardiac transplant recipients; children <10 years have higher values); ESRD: 3.49 L/kg

Protein binding: 90% to 98% to lipoproteins

Metabolism: Extensively hepatic via CYP3A4; forms at least 25 metabolites; extensive first-pass effect following oral administration

Bioavailability: Oral:

Cyclosporine (non-modified): Dependent on patient population and transplant type (<10% in adult liver transplant recipients and as high as 89% in renal transplant recipients); bioavailability of Sandimmune capsules and oral solution are equivalent; bioavailability of oral solution is ~30% of the IV solution

Children: 28% (range: 17% to 42%); gut dysfunction common in BMT patients and oral bioavailability is further reduced

Cyclosporine (modified): Bioavailability of Neoral capsules and oral solution are equivalent:

Children: 43% (range: 30% to 68%)

Adults: 23% greater than with cyclosporine (non-modified) in renal transplant recipients; 50% greater in liver transplant recipients.

Half-life elimination: Oral: May be prolonged in patients with hepatic impairment and shorter in pediatric patients due to the higher metabolism rate

Cyclosporine (non-modified): Biphasic: Alpha: 1.4 hours; Terminal: 19 hours (range: 10-27 hours)

Cyclosporine (modified): Biphasic: Terminal: 8.4 hours (range: 5-18 hours)

Time to peak, serum: Oral:

Cyclosporine (non-modified): 2-6 hours; some patients have a second peak at 5-6 hours

Cyclosporine (modified): Renal transplant: 1.5-2 hours

Excretion: Primarily feces; urine (6%, 0.1% as unchanged drug and metabolites); clearance is more rapid in pediatric patients than in adults

Pharmacokinetics: Additional Considerations (Adult Data Unless Noted)

Altered kidney function: In a limited number of patients with end-stage renal disease (ESRD) (creatinine clearance <5 mL/minute), cyclosporine 3.5 mg/kg IV over 4 hours administered at the end of a hemodialysis session resulted in a mean volume of distribution of 3.49 L/kg and systemic clearance of 0.369 L/h/kg. This systemic clearance was approximately two-thirds the mean systemic clearance (0.56 L/h/kg) of cyclosporine in controls with normal renal function. In a small number of liver transplant recipients, the mean clearance on and off hemodialysis was 463 mL/min and 398 mL/min, respectively. Less than 1% of the dose was recovered in the dialysate.

Hepatic function impairment: Severe hepatic impairment may result in significantly reduced clearance and increased cyclosporine exposures.

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

  • (AE) United Arab Emirates: Restasis | Sandimmun neoral | Sigmasporin | Sigmasporin microral;
  • (AR) Argentina: Gengraf | Sandimmun | Sandimmun neoral | Sigmasporin microral;
  • (AT) Austria: Cicloralhexal | Ikervis | Neoimmun | Sandimmun | Sandimmun n. | Sandimmun neoral | Vanquoral | Verkazia;
  • (AU) Australia: Apo ciclosporin | Cicloral | Cyclosporin sandoz | Cysporin | Ikervis | Neoral | Sandimmun | Sandimmun conc;
  • (BD) Bangladesh: Neoral;
  • (BE) Belgium: Ciqorin | Neoral sandimmun | Sandimmun;
  • (BG) Bulgaria: Ciclosporin alkaloid | Equoral | Ikervis | Sandimmun neoral;
  • (BR) Brazil: Ciclosporina | Ciclosporina sandoz | Gengraf | Restasis | Sandimmun | Sandimmun neoral | Sigmasporin microral;
  • (CH) Switzerland: Ciclosol | Ciqorin | Sandimmun | Sandimmun neoral | Verkazia;
  • (CL) Chile: Gengraf | Restasis | Sandimmun | Sigmasporin microral;
  • (CN) China: Ciclosporin a | Cyclosporin | Gengraf | Implanta | Lizhu huanming | Neocyspin | Sandimmun | Sandimmun neoral | Tian ke | Tianke | Yin pu lan ta;
  • (CO) Colombia: Biosporin | Cicladvan | Ciclosporina | Citabiclos | Genosporin | Inhiblock | Restasis | Sandimmun | Sandimmun neoral;
  • (CZ) Czech Republic: Ciclosporin Mylan | Consupren | Cyclaid | Equoral | Ikervis | Sandimmun | Sandimmun neoral;
  • (DE) Germany: Cicloral | Ciclosporin 1a-pharma | Ciclosporin dura | Ciclosporin pro | Ciqorin | Deximune | Ikervis | Immunosporin | Neoral | Sandimmun | Sandimmun neoral | Sandimmun optoral;
  • (DK) Denmark: Sandimmun;
  • (DO) Dominican Republic: Restasis | Sandimmun neoral;
  • (EC) Ecuador: Citabiclos | Restasis | Sandimmun;
  • (EE) Estonia: Ciclo hexal | Equoral | Ikervis | Sandimmun | Sandimmun neoral;
  • (EG) Egypt: Abrammune | Arpimune me | Cyclosporine | Restasis | Sandimmun neoral;
  • (ES) Spain: Ciclosporina Mylan | Ciqorin | Ikervis | Sandimmun | Sandimmun neoral;
  • (ET) Ethiopia: Cyclosporine | Sandimmun neoral;
  • (FI) Finland: Ciqorin | Ikervis | Sandimmun;
  • (FR) France: Neoral | Sandimmun | Verkazia;
  • (GB) United Kingdom: Capimune | Capsorin | Cyclosporin | Ikervis | Neoral | Sandimmun | Vanquoral | Verkazia;
  • (GR) Greece: Imunofar | Sandimmun | Sandimmun neoral | Sporilen;
  • (HK) Hong Kong: Gengraf | Ikervis | Restasis | Sandimmun | Sandimmun neoral;
  • (HR) Croatia: Ciklosporin alkaloid | Equoral | Sandimmun neoral;
  • (HU) Hungary: Ciqorin | Ikervis | Sandimmun | Sandimmun neoral;
  • (ID) Indonesia: Ikervis | Sandimmun | Sandimmun neoral;
  • (IE) Ireland: Ikervis | Neoral | Sandimmun;
  • (IL) Israel: Deximune | Sandimmun | Sandimmun neoral;
  • (IN) India: Arpimune me | Arpimune o | Consiral | Cyclonox | Cyclophil me | Grafotas | Graftin | Iminoral | Imudrops | Imusporin | Panimum bioral | Panimun | Panimun bioral | Pasilow | Psoranext | Psorid | Psortude | Restasis | Sandimmun | Sandimmun neoral | Sorigo | Zymmune;
  • (IT) Italy: Ciqorin | Ikervis | Restasis | Sandimmun | Sandimmun neoral | Verkazia;
  • (JO) Jordan: Clasgen | Restasis | Sandimmun | Sandimmun neoral;
  • (JP) Japan: Amadra | Ciclosporin sandoz | Cicporal | Neomerck | Neoral | Papilock Mini | Sandimmun;
  • (KE) Kenya: Arpimune me | Panimun bioral | Sandimmun neoral;
  • (KR) Korea, Republic of: A lux | Anaporin | Bearporine | Biclosporin | Biporin | Cipol | Cipol n | Clacier | Cycrine | Cyporin | Cysis | Dryfree | Ensporin | Eyetasis | Gengraf | Husporin | Ikervis | Implanta | Lestine | Ocuporine | Optiporine | Resporin | Retas | S porin | Sandimmun | Sandimmun neoral | Sporine s | T sporin;
  • (KW) Kuwait: Restasis | Sandimmun neoral;
  • (LB) Lebanon: Consupren | Sandimmun | Sandimmun neoral | Sigmasporin;
  • (LT) Lithuania: Ciclo hexal | Depores | Equoral | Ikervis | Sandimmun | Sandimmun neoral | Sandimmun optoral;
  • (LU) Luxembourg: Ciqorin | Neoral sandimmun | Sandimmun;
  • (LV) Latvia: Ciqorin | Equoral | Ikervis | Sandimmun | Sandimmun neoral;
  • (MX) Mexico: Ciclosporina | Emicrox | Equoral | Neolem | Restasis | Sandimmun | Sandimmun neoral | Supremunn;
  • (MY) Malaysia: Gengraf | Ikervis | Restasis | Sandimmun | Sandimmun neoral;
  • (NG) Nigeria: Sandiummum neoral;
  • (NL) Netherlands: Ciclosporine | Ciclosporine pch | Ciqorin | Ikervis | Neoral | Sandimmune;
  • (NO) Norway: Ciqorin | Ikervis | Restasis | Sandimmun | Sandimmun neoral;
  • (NZ) New Zealand: Restasis | Sandimmun;
  • (PE) Peru: Clacier | Sandimmun | Sandimmun neoral | Sigmaimmun;
  • (PH) Philippines: Ikervis | Sandimmun;
  • (PK) Pakistan: Bioral | Cipol n | Consupren | Gramune | Imun heoral | Neocyspin | Sandimmun | Sandimmun neoral | Tianke;
  • (PL) Poland: Cyclaid | Equoral | Gengraf | Ikervis | Sandimmun | Sandimmun neoral;
  • (PR) Puerto Rico: Cequa | Cyclosporine | Gengraf | Neoral | Restasis | Sandimmune;
  • (PT) Portugal: Ciclosporina | Ikervis | Sandimmun | Sandimmun neoral;
  • (PY) Paraguay: Ciclosporina bioethic pharma | Ciclosporina prosalud | Cyrin | Imusporin | Neocyspin | Panimun bioral | Sandimmun | Sandimmun neoral | Sigmasporin microral | Transporina | Zaven me;
  • (QA) Qatar: Sandimmun | Sandimmun Neoral | Sigmasporin Microral;
  • (RO) Romania: Ciqorin | Equoral | Ikervis;
  • (RU) Russian Federation: Consupren | Cyclopren | Cycloral | Ecoral | Equoral | Imusporin | Orgasporin | Panimun bioral | Restasis | Sandimmun | Sandimmun neoral | Vero cyclosporin;
  • (SA) Saudi Arabia: Clasgen | Gengraf | Restasis | Sandimmun | Sandimmun neoral;
  • (SE) Sweden: Ciklosporin 2care4 | Ciklosporin ebb | Ciklosporin Ivax | Ciklosporin medartuum | Ciklosporin orifarm | Ciqorin | Equoral | Ikervis | Sandimmun | Sandimmun neoral;
  • (SG) Singapore: Deximune | Equoral | Gengraf | Ikervis | Restasis | Sandimmun | Sandimmun neoral;
  • (SI) Slovenia: Ciklosporin alkaloid | Ciqorin | Ikervis | Restasis | Sandimmun | Sandimmun neoral;
  • (SK) Slovakia: Ciclosporin Mylan | Consupren | Cyclaid | Equoral | Ikervis | Sandimmun | Sandimmun neoral;
  • (TH) Thailand: Cipol n | Consupren | Consupren s | Gengraf | Ikervis | Kasporin | Sandimmun | Sandimmun neoral;
  • (TN) Tunisia: Equoral | Immupry | Sandimmum | Sandimmum neoral;
  • (TR) Turkey: Depores | Depores x | Drosporin | Epostin | Gengraf | Panosporin | Restasis | Sandimmun | Sandimmun neoral | Sigmasporin microral | Tearon;
  • (TW) Taiwan: Ikervis | Immurin | Sandimmun | Sandimmun neoral;
  • (UA) Ukraine: Equoral | Restasis | Sandimmun | Sandimmun neoral;
  • (UG) Uganda: Neoral;
  • (UY) Uruguay: Sandimmun | Sandimmun neoral | Sigmasporin microral;
  • (VE) Venezuela, Bolivarian Republic of: Sandimmun neoral;
  • (VN) Viet Nam: Tandorex;
  • (ZA) South Africa: Ciclohexal | Restasis | Sandimmun | Sandimmun neoral
  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. 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]
  3. Actis GC, Debernardi-Venon W, Lagget M, et al. Hepatotoxicity of intravenous cyclosporin A in patients with acute ulcerative colitis on total parenteral nutrition. Liver. 1995;15(6):320-323. doi:10.1111/j.1600-0676.1995.tb00692.x [PubMed 8609812]
  4. Aksamit AJ Jr, de Groen PC. Cyclosporine-related leukoencephalopathy and PML in a liver transplant recipient. Transplantation. 1995;60(8):874-876. [PubMed 7482750]
  5. 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]
  6. al-Uzri A, Conley SB, Orlandi P, So S, Salvatierra O Jr. Aggressive early cyclosporine therapy is desirable in pediatric renal transplantation. Transplant Proc. 1994;26(1):88-90. [PubMed 8109036]
  7. Alvarez F, Atkison PR, Grant DR, et al. NOF-11: a one-year pediatric randomized double-blind comparison of neoral versus sandimmune in orthotopic liver transplantation. Transplantation. 2000;69(1):87-92. doi:10.1097/00007890-200001150-00016 [PubMed 10653385]
  8. Alvarez F, Ciocca M, Cañero-Velasco C, et al. Short-term cyclosporine induces a remission of autoimmune hepatitis in children. J Hepatol. 1999;30(2):222-227. doi:10.1016/s0168-8278(99)80065-8 [PubMed 10068099]
  9. Andrews DJ, Cramb R. Cyclosporin: revisions in monitoring guidelines and review of current analytical methods. Ann Clin Biochem. 2002;39(Pt 5):424-435. doi:10.1258/000456302320314430 [PubMed 12227848]
  10. 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]
  11. Aragon E, Chan YH, Ng KH, Lau YW, Tan PH, Yap HK. Good outcomes with mycophenolate-cyclosporine-based induction protocol in children with severe proliferative lupus nephritis. Lupus. 2010;19(8):965-973. doi:10.1177/0961203310366855 [PubMed 20581019]
  12. Aronoff GR, Bennett WM, Berns JS, et al, Drug Prescribing in Renal Failure: Dosing Guidelines for Adults and Children, 5th ed. Philadelphia, PA: American College of Physicians; 2007;116.
  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. Baca V, Catalán T, Villasís-Keever M, Ramón G, Morales AM, Rodríguez-Leyva F. Effect of low-dose cyclosporine A in the treatment of refractory proteinuria in childhood-onset lupus nephritis. Lupus. 2006;15(8):490-495. doi:10.1191/0961203306lu2312oa [PubMed 16942000]
  15. Bagg A, Dunphy CH. Immunosuppressive and immunomodulatory therapy-associated lymphoproliferative disorders. Semin Diagn Pathol. 2013;30(2):102-112. doi:10.1053/j.semdp.2012.08.005 [PubMed 23541274]
  16. Bansal S, Lucia MS, Wiseman A. A case of polyomavirus-associated nephropathy presenting late after transplantation. Nat Clin Pract Nephrol. 2008;4(5):283-287. doi:10.1038/ncpneph0784 [PubMed 18334968]
  17. 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]
  18. Battiwalla M, Melenhorst J, Saunthararajah Y, et al. HLA-DR4 predicts haematological response to cyclosporine in T-large granular lymphocyte lymphoproliferative disorders. Br J Haematol. 2003;123(3):449-453. doi:10.1046/j.1365-2141.2003.04613.x [PubMed 14617004]
  19. Bechstein WO. Neurotoxicity of calcineurin inhibitors: impact and clinical management. Transpl Int. 2000;13(5):313-326. doi:10.1007/s001470050708 [PubMed 11052266]
  20. Benfield MR, Tejani A, Harmon WE, McDonald R, Stablein DM, McIntosh M, Rose S; CCTPT Study Group. A randomized multicenter trial of OKT3 mAbs induction compared with intravenous cyclosporine in pediatric renal transplantation. Pediatr Transplant. 2005;9(3):282-292. doi:10.1111/j.1399-3046.2005.00296.x [PubMed 15910382]
  21. 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]
  22. Bertsias GK, Tektonidou M, Amoura Z, et al. Joint European League Against Rheumatism and European Renal Association-European Dialysis and Transplant Association (EULAR/ERA-EDTA) recommendations for the management of adult and paediatric lupus nephritis. Ann Rheum Dis. 2012;71(11):1771-1782. doi:10.1136/annrheumdis-2012-201940 [PubMed 22851469]
  23. Bird SJ. Chronic immunotherapy for myasthenia gravis. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed February 28, 2022.
  24. Boni R, Dummer R. Abscessed inflammation as a serious complication of low dose cyclosporin A in atopic dermatitis. Eur J Dermatol. 1995;5:268-269.
  25. Boratyńska M, Radwan-Oczko M, Falkiewicz K, Klinger M, Szyber P. Gingival overgrowth in kidney transplant recipients treated with cyclosporine and its relationship with chronic graft nephropathy. Transplant Proc. 2003;35(6):2238-2240. doi:10.1016/s0041-1345(03)00800-5 [PubMed 14529900]
  26. Braun N, Schmutzler F, Lange C, et al. Immunosuppressive treatment for focal segmental glomerulosclerosis in adults. Cochrane Database Syst Rev. 2008;2008(3):CD003233. doi:10.1002/14651858.CD003233.pub2 [PubMed 18646090]
  27. Burckart G, Starzl T, Williams L, et al. Cyclosporine monitoring and pharmacokinetics in pediatric liver transplant patients. Transplant Proc. 1985;17(1):1172-1175. [PubMed 20640180]
  28. Burckart GJ, Venkataramanan R, Ptachcinski RJ, et al. Cyclosporine absorption following orthotopic liver transplantation. J Clin Pharmacol.1986;26(8):647-651. doi:10.1002/j.1552-4604.1986.tb02966.x [PubMed 3540030]
  29. Cabiddu G, Spotti D, Gernone G, et al. A best-practice position statement on pregnancy after kidney transplantation: focusing on the unsolved questions. The Kidney and Pregnancy Study Group of the Italian Society of Nephrology [published correction appears in J Nephrol. 2018 Jul 6]. J Nephrol. 2018;31(5):665-681. doi:10.1007/s40620-018-0499-x [PubMed 29949013]
  30. Caforio AL, Gambino A, Tona F, et al. Sulfinpyrazone reduces cyclosporine levels: a new drug interaction in heart transplant recipients. J Heart Lung Transplant. 2000;19(12):1205-1208. doi:10.1016/s1053-2498(00)00216-3 [PubMed 11124491]
  31. 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]
  32. 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]
  33. Cantarovich M. New approaches to individualizing cyclosporine dose. Transplantation. 2004;77(3):357-358. doi:10.1097/00007890-200402150-00008 [PubMed 14983877]
  34. Cantarovich M, Barkun JS, Tchervenkov JI, Besner JG, Aspeslet L, Metrakos P. Comparison of neoral dose monitoring with cyclosporine through levels versus 2-hr postdose levels in stable liver transplant patients. Transplantation. 1998;66(12):1621-1627. doi:10.1097/00007890-199812270-00009 [PubMed 9884249]
  35. Cattran DC, Appel GB, Hebert LA, et al. A randomized trial of cyclosporine in patients with steroid-resistant focal segmental glomerulosclerosis. North America Nephrotic Syndrome Study Group. Kidney Int. 1999;56(6):2220-2226. doi:10.1046/j.1523-1755.1999.00778.x [PubMed 10594798]
  36. Cebeci I, Kantarci A, Firatli E, et al. Evaluation of the frequency of HLA determinants in patients with gingival overgrowth induced by cyclosporine-A. J Clin Periodontol. 1996;23(8):737-742. doi:10.1111/j.1600-051x.1996.tb00603.x [PubMed 8877659]
  37. 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]
  38. Chao NJ, Schmidt GM, Niland JC, et al. Cyclosporine, methotrexate, and prednisone compared with cyclosporine and prednisone for prophylaxis of acute graft-versus-host disease. N Engl J Med. 1993;329(17):1225-1230. doi:10.1056/NEJM199310213291703 [PubMed 8413388]
  39. Chao NJ, Snyder DS, Jain M, et al. Equivalence of 2 effective graft-versus-host disease prophylaxis regimens: results of a prospective double-blind randomized trial. Biol Blood Marrow Transplant. 2000;6(3):254-261. doi:10.1016/s1083-8791(00)70007-3 [PubMed 10871150]
  40. Chiurchiu C, Ruggenenti P, Remuzzi G. Thrombotic microangiopathy in renal transplantation. Ann Transplant. 2002;7(1):28-33. [PubMed 12221901]
  41. 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]
  42. Choudhary DR, Naithani R, Mahapatra M, Kumar R, Mishra P, Saxena R. Efficacy of cyclosporine as a single agent therapy in chronic idiopathic thrombocytopenic purpura. Haematologica. 2008;93(10):e61-e62. doi:10.3324/haematol.13481 [PubMed 18827257]
  43. Choudhry S, Bagga A, Hari P, Sharma S, Kalaivani M, Dinda A. Efficacy and safety of tacrolimus versus cyclosporine in children with steroid-resistant nephrotic syndrome: a randomized controlled trial. Am J Kidney Dis. 2009;53(5):760-769. doi:10.1053/j.ajkd.2008.11.033 [PubMed 19268410]
  44. Chung EK, Yee J, Kim JY, Gwak HS. Low cyclosporine concentrations in children and time to acute graft versus host disease. BMC Pediatr. 2020;20(1):206. doi:10.1186/s12887-020-02125-6 [PubMed 32393210]
  45. Ciafaloni E, Nikhar NK, Massey JM, Sanders DB. Retrospective analysis of the use of cyclosporine in myasthenia gravis. Neurology. 2000;55(3):448-450. doi:10.1212/wnl.55.3.448 [PubMed 10932288]
  46. Clardy CW, Schroeder TJ, Myre SA, et al. Clinical variability of cyclosporine pharmacokinetics in adult and pediatric patients after renal, cardiac, hepatic, and bone-marrow transplants. Clin Chem. 1988;34(10):2012-2015. [PubMed 3048779]
  47. Claris O, Picaud JC, Brazier JL, Salle BL. Pharmacokinetics of cyclosporin A in 16 newborn infants of renal or cardiac transplant mothers. Dev Pharmacol Ther. 1993;20(3-4):180-185. doi:10.1159/000457560 [PubMed 7828451]
  48. Clive DM. Renal transplant-associated hyperuricemia and gout. J Am Soc Nephrol. 2000;11(5):974-979. doi:10.1681/ASN.V115974 [PubMed 10770978]
  49. 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]
  50. 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]
  51. 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]
  52. Cuarterolo ML, Ciocca M, López S, Araujo M, Álvarez F. Autoimmune hepatitis in children: prednisone plus azathioprine versus cyclosporine: a randomized trial. J Pediatr Gastroenterol Nutr. 2020;71(3):376-380. doi:10.1097/MPG.0000000000002776 [PubMed 32520828]
  53. Cuarterolo M, Ciocca M, Velasco CC, et al. Follow-up of children with autoimmune hepatitis treated with cyclosporine. J Pediatr Gastroenterol Nutr. 2006;43(5):635-639. doi:10.1097/01.mpg.0000235975.75120.38 [PubMed 17130741]
  54. 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]
  55. 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]
  56. Davies MG, Bowers PW. Alopecia areata arising in patients receiving cyclosporin immunosuppression. Br J Dermatol. 1995;132(5):835-836. doi:10.1111/j.1365-2133.1995.tb00742.x [PubMed 7772501]
  57. 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]
  58. Debray D, Maggiore G, Girardet JP, Mallet E, Bernard O. Efficacy of cyclosporin A in children with type 2 autoimmune hepatitis. J Pediatr. 1999;135(1):111-114. doi:10.1016/s0022-3476(99)70339-2 [PubMed 10393616]
  59. Dehghani SM, Taghavi SA, Geramizadeh B, et al. Hepatitis B recurrence after liver transplantation: a single center experiences and review the literature. Hepat Mon. 2013;13(1):e6609. doi:10.5812/hepatmon.6609 [PubMed 23483668]
  60. de Saussure P, Soravia C, Morel P, Hadengue A. Low-dose oral microemulsion ciclosporin for severe, refractory ulcerative colitis. Aliment Pharmacol Ther. 2005;22(3):203-208. doi: 10.1111/j.1365-2036.2005.02552.x. [PubMed 16091057]
  61. 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]
  62. Díaz-Llopis M, Gallego-Pinazo R, García-Delpech S, Salom-Alonso D. General principles for the treatment of non-infectious uveitis. Inflamm Allergy Drug Targets. 2009;8(4):260-265. doi:10.2174/187152809789352203 [PubMed 19754409]
  63. Ducharme MP, Warbasse LH, Edwards DJ. Disposition of intravenous and oral cyclosporine after administration with grapefruit juice. Clin Pharmacol Ther. 1995;57(5):485-491. doi:10.1016/0009-9236(95)90032-2 [PubMed 7768070]
  64. Dunn CJ, Wagstaff AJ, Perry CM, Plosker GL, Goa KL. Cyclosporin: an updated review of the pharmacokinetic properties, clinical efficacy and tolerability of a microemulsion-based formulation (neoral)1 in organ transplantation. Drugs. 2001;61(13):1957-2016. doi:10.2165/00003495-200161130-00006 [PubMed 11708766]
  65. 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):50-55. [PubMed 12091650]
  66. 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]
  67. Ellis D, Jaffe R, Green M, Janosky JJ, Lombardozzi-Lane S, Shapiro R, Scantlebury V, Vivas C, Jordan ML. 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]
  68. Emilia G, Morselli M, Luppi M, et al. Long-term salvage therapy with cyclosporin A in refractory idiopathic thrombocytopenic purpura. Blood. 2002;99(4):1482-1485. doi:10.1182/blood.v99.4.1482 [PubMed 11830504]
  69. Erdman J, Wolfram J, Nimke D, et al. Lung transplant outcomes in adults in the United States: retrospective cohort study using real-world evidence from the SRTR. Transplantation. 2022;106(6):1233-1242. doi:10.1097/TP.0000000000004011 [PubMed 34974456]
  70. 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.
  71. 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]
  72. 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]
  73. Feuerstein JD, Isaacs KL, Schneider Y, Siddique SM, Falck-Ytter Y, Singh S; AGA Institute Clinical Guidelines Committee. AGA clinical practice guidelines on the management of moderate to severe ulcerative colitis. Gastroenterology. 2020;158(5):1450-1461. doi:10.1053/j.gastro.2020.01.006 [PubMed 31945371]
  74. Follath F, Wenk M, Vozeh S, et al. Intravenous cyclosporine kinetics in renal failure. Clin Pharmacol Ther. 1983;34(5):638-643. doi:10.1038/clpt.1983.226 [PubMed 6627824]
  75. Fraenkel L, Bathon JM, England BR, et al. 2021 American College of Rheumatology guideline for the treatment of rheumatoid arthritis. Arthritis Care Res (Hoboken). 2021;73(7):924-939. doi:10.1002/acr.24596 [PubMed 34101387]
  76. Franulović OZ, Rajacić N, Lesar T, Kuna AT, Morić BV. Cyclosporine induced biochemical remission in childhood autoimmune hepatitis. Coll Antropol. 2012;36(3):973-979. [PubMed 23213960]
  77. Fu LW, Yang LY, Chen WP, Lin CY. Clinical efficacy of cyclosporin a neoral in the treatment of paediatric lupus nephritis with heavy proteinuria. Br J Rheumatol. 1998;37(2):217-221. doi:10.1093/rheumatology/37.2.217 [PubMed 9569080]
  78. 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]
  79. Fujinaga S, Shimizu T. Chronic cyclosporine-induced nephrotoxicity in children with steroid-resistant nephrotic syndrome. Pediatr Nephrol. 2013;28(10):2065-2066. doi:10.1007/s00467-013-2535-3 [PubMed 23780471]
  80. Funch DP, Ko HH, Travasso J, et al. Posttransplant lymphoproliferative disorder among renal transplant patients in relation to the use of mycophenolate mofetil. Transplantation. 2005;80(9):1174-11780. doi:10.1097/01.tp.0000169035.10572.c6 [PubMed 16314782]
  81. Garbin LM, Tonani M, Salvador M, et al. Cyclosporine level: difference between blood samples collected through peripheral and central venous access. J Clin Nurs. 2013;22(3-4):395-404. doi:10.1111/j.1365-2702.2012.04187.x [PubMed 22805438]
  82. Gauthier A, Bleyzac N, Garnier N, et al. Goal-oriented monitoring of cyclosporine is effective for graft-versus-host disease prevention after hematopoietic stem cell transplantation in sickle cell disease and thalassemia major. Biol Blood Marrow Transplant. 2020;26(12):2285-2291. doi:10.1016/j.bbmt.2020.01.016 [PubMed 32007639]
  83. Gellermann J, Weber L, Pape L, et al. Mycophenolate mofetil versus cyclosporin A in children with frequently relapsing nephrotic syndrome. J Am Soc Nephrol. 2013;24(10):1689-1697. doi:10.1681/ASN.2012121200 [PubMed 23813218]
  84. Gengraf (cyclosporine) capsules [prescribing information]. North Chicago, IL: AbbVie Inc; February 2021.
  85. Gengraf (cyclosporine) oral solution [prescribing information]. North Chicago, IL: AbbVie Inc; February 2021.
  86. George JN, Nester CM. Syndromes of thrombotic microangiopathy. N Engl J Med. 2014;371(7):654-666. doi:10.1056/NEJMra1312353 [PubMed 25119611]
  87. Georgiou GK, Dounousi E, Harissis HV. Calcineurin inhibitors and male fertility after renal transplantation - a review. Andrologia. 2016;48(5):483-490. doi:10.1111/and.12477 [PubMed 26341518]
  88. Gipson DS, Massengill SF, Yao L, et al. Management of childhood onset nephrotic syndrome. Pediatrics. 2009;124(2):747-757. doi:10.1542/peds.2008-1559 [PubMed 19651590]
  89. Gipson DS, Trachtman H, Kaskel FJ, et al. Clinical trial of focal segmental glomerulosclerosis in children and young adults. Kidney Int. 2011;80(8):868-878. doi:10.1038/ki.2011.195 [PubMed 21734640]
  90. 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]
  91. Green M. Introduction: Infections in solid organ transplantation. Am J Transplant. 2013;13(suppl 4):3-8. doi:10.1111/ajt.12093 [PubMed 23464993]
  92. Griffith BP, Bando K, Hardesty RL, et al. A prospective randomized trial of FK506 versus cyclosporine after human pulmonary transplantation. Transplantation. 1994;57(6):848-851. doi:10.1097/00007890-199403270-00013 [PubMed 7512292]
  93. Grimer M; 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):S98-S105. doi:10.1111/j.1440-1797.2006.00735.x [PubMed 17316288]
  94. Gulbis B, Adler M, Ooms HA, Desmet JM, Leclerc JL, Primo G. Liver-function studies in heart-transplant recipients treated with cyclosporin A. Clin Chem. 1988;34(9):1772-1774. [PubMed 2901299]
  95. Hahn BH, McMahon MA, Wilkinson A, et al; American College of Rheumatology. American College of Rheumatology guidelines for screening, treatment, and management of lupus nephritis. Arthritis Care Res (Hoboken). 2012;64(6):797-808. doi:10.1002/acr.21664 [PubMed 22556106]
  96. Hardinger KL, Koch MJ, Brennan DC. Current and future immunosuppressive strategies in renal transplantation. Pharmacotherapy. 2004;24(9):1159-76. doi:10.1592/phco.24.13.1159.38094 [PubMed 15460177]
  97. Hefti AF, Eshenaur AE, Hassell TM, Stone C. Gingival overgrowth in cyclosporine A treated multiple sclerosis patients. J Periodontol. 1994;65(8):744-749. doi:10.1902/jop.1994.65.8.744 [PubMed 7965549]
  98. Higgins EM, Hughes JR, Snowden S, Pembroke AC. Cyclosporin-induced periungual granulation tissue. Br J Dermatol. 1995;132(5):829-830. doi:10.1111/j.1365-2133.1995.tb00737.x [PubMed 7772496]
  99. 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]
  100. Hodson EM, Sinha A, Cooper TE. Interventions for focal segmental glomerulosclerosis in adults. Cochrane Database Syst Rev. 2022;2(2):CD003233. doi:10.1002/14651858.CD003233.pub3 [PubMed 35224732]
  101. Hollander AA, van Rooij J, Lentjes GW, et al. The effect of grapefruit juice on cyclosporine and prednisone metabolism in transplant patients. Clin Pharmacol Ther. 1995;57(3):318-324. doi:10.1016/0009-9236(95)90157-4 [PubMed 7697949]
  102. Holt DW, Mueller EA, Kovarik JM, van Bree JB, Richard F, Kutz K. Sandimmun neoral pharmacokinetics: impact of the new oral formulation. Transplant Proc. 1995;27(1):1434-1437. [PubMed 7878935]
  103. Honcharik N, Anthone S. Activated charcoal in acute cyclosporin overdose. Lancet. 1985;1(8436):1051. doi:10.1016/s0140-6736(85)91660-5 [PubMed 2859506]
  104. Hood KA. Drug-induced gingival hyperplasia in transplant recipients. Prog Transplant. 2002;12(1):17-21; quiz 22-3. doi:10.7182/prtr.12.1.k0605089820vt807 [PubMed 11993065]
  105. 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]
  106. Horina JH, Wirnsberger GH, Kenner L, Holzer H, Krejs GJ. Increased susceptibility for CsA-induced hepatotoxicity in kidney graft recipients with chronic viral hepatitis C. Transplantation. 1993;56(5):1091-1094. doi:10.1097/00007890-199311000-00008 [PubMed 8249106]
  107. 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]
  108. Horton RC, Bonser RS. Interaction between cyclosporin and fluoxetine. BMJ. 1995;311(7002):422. doi:10.1136/bmj.311.7002.422 [PubMed 7640589]
  109. Hošková L, Málek I, Kopkan L, Kautzner J. Pathophysiological mechanisms of calcineurin inhibitor-induced nephrotoxicity and arterial hypertension. Physiol Res. 2017;66(2):167-180. doi:10.33549/physiolres.933332 [PubMed 27982677]
  110. Hughes RL. Cyclosporine-related central nervous system toxicity in cardiac transplantation. N Engl J Med. 1990;323(6):420-421. doi:10.1056/NEJM199008093230616 [PubMed 2370897]
  111. "Inactive" ingredients in pharmaceutical products: update (subject review). American Academy of Pediatrics Committee on Drugs. Pediatrics. 1997;99(2):268-278. [PubMed 9024461]
  112. Inamoto Y, Flowers ME, Appelbaum FR, et al. A retrospective comparison of tacrolimus versus cyclosporine with methotrexate for immunosuppression after allogeneic hematopoietic cell transplantation with mobilized blood cells. Biol Blood Marrow Transplant. 2011;17(7):1088-1092. doi:10.1016/j.bbmt.2011.01.017 [PubMed 21421070]
  113. Isnard Bagnis C, Tezenas du Montcel S, Beaufils H, et al. Long-term renal effects of low-dose cyclosporine in uveitis-treated patients: follow-up study. J Am Soc Nephrol. 2002;13(12):2962-2968. doi:10.1097/01.asn.0000034945.61533.26 [PubMed 12444215]
  114. Kabeer MH, Filo RS, Milgrom ML, et al. Central pontine myelinolysis following orthotopic liver transplant: association with cyclosporine toxicity. Postgrad Med J. 1995;71(834):239-241. doi:10.1136/pgmj.71.834.239 [PubMed 7784287]
  115. Kagawa Y, Sawada J, Yamada S, et al. Relationship between development of nephrotoxicity and blood concentration of cyclosporine A in bone-marrow transplanted recipients who received the continuous intravenous infusion. Biol Pharm Bull. 2003;26(8):1115-1119. doi:10.1248/bpb.26.1115 [PubMed 12913261]
  116. Kahan BD, Conley S, Portman R, et al. Parent-to-child transplantation with cyclosporine immunosuppression. J Pediatr.1987;111(6 Pt 2):1012-1016. doi:10.1016/s0022-3476(87)80047-1 [PubMed 3316572]
  117. 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]
  118. Kaufman DB, Kaplan B, Kanwar YS, Abecassis M, Stuart FP. The successful use of tacrolimus (FK506) in a pancreas/kidney transplant recipient with recurrent cyclosporine-associated hemolytic uremic syndrome. Transplantation. 1995;59(12):1737-1739. doi:10.1097/00007890-199506270-00017 [PubMed 7541579]
  119. Kaushik SB, Lebwohl MG. Psoriasis: which therapy for which patient: focus on special populations and chronic infections. J Am Acad Dermatol. 2019;80(1):43-53. doi:10.1016/j.jaad.2018.06.056 [PubMed 30017706]
  120. Kawasaki Y, Hosoya M, Takano K, et al. Efficacy of cyclosporine therapy for systemic lupus erythematosus in childhood. Pediatr Int. 2008;50(2):257-259. doi:10.1111/j.1442-200X.2008.02548.x [PubMed 18353074]
  121. Kengne-Wafo S, Massella L, Diomedi-Camassei F, et al. Risk factors for cyclosporin A nephrotoxicity in children with steroid-dependant nephrotic syndrome. Clin J Am Soc Nephrol. 2009;4(9):1409-1416. doi:10.2215/CJN.01520209 [PubMed 19628686]
  122. 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]
  123. 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]
  124. Kidney Disease: Improving Global Outcomes (KDIGO) Glomerulonephritis Work Group. KDIGO Clinical Practice Guideline for Glomerulonephritis. Kidney Int. Suppl. 2012;139-274.
  125. 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):S1-155. doi:10.1111/j.1600-6143.2009.02834.x [PubMed 19845597]
  126. Kino KJ, Wittkowsky AK. Influence of bile acid replacement on cyclosporine absorption in a patient with jejunoileal bypass. Pharmacotherapy. 1995;15(3):350-352. [PubMed 7667169]
  127. Klintmalm GB, Iwatsuki S, Starzl TE. Cyclosporin A hepatotoxicity in 66 renal allograft recipients. Transplantation. 1981;32(6):488-489. doi:10.1097/00007890-198112000-00007 [PubMed 7041349]
  128. Koc S, Leisenring W, Flowers ME, et al. Therapy for chronic graft-versus-host disease: a randomized trial comparing cyclosporine plus prednisone versus prednisone alone. Blood. 2002;100(1):48-51. doi:10.1182/blood.v100.1.48 [PubMed 12070007]
  129. Kowdley KV, Keeffe EB. Hepatotoxicity of transplant immunosuppressive agents. Gastroenterol Clin North Am. 1995;24(4):991-1001. [PubMed 8749908]
  130. 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]
  131. 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]
  132. 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]
  133. Lamy T, Loughran TP Jr. How I treat LGL leukemia. Blood. 2011;117(10):2764-7274. doi:10.1182/blood-2010-07-296962 [PubMed 21190991]
  134. Lanino E, Rondelli R, Locatelli F, et al. Early (day -7) versus conventional (day -1) inception of cyclosporine-A for graft-versus-host disease prophylaxis after unrelated donor hematopoietic stem cell transplantation in children. Long-term results of an AIEOP prospective, randomized study. Biol Blood Marrow Transplant. 2009;15(6):741-748. doi:10.1016/j.bbmt.2009.03.004 [PubMed 19450759]
  135. Lapointe M, Baillie GM, Bhaskar SS, et al. Cyclosporine-induced hemolytic uremic syndrome and hemorrhagic colitis following renal transplantation. Clin Transplant. 1999;13(6):526-530. doi:10.1034/j.1399-0012.1999.130614.x [PubMed 10617244]
  136. Lavrnic D, Vujic A, Rakocevic-Stojanovic V, et al. Cyclosporine in the treatment of myasthenia gravis. Acta Neurol Scand. 2005;111(4):247-252. doi:10.1111/j.1600-0404.2005.00378.x [PubMed 15740576]
  137. Lawitschka A, Lucchini G, Strahm B, et al. Pediatric acute graft-versus-host disease prophylaxis and treatment: surveyed real-life approach reveals dissimilarities compared to published recommendations. Transpl Int. 2020;33(7):762-772. doi:10.1111/tri.13601 [PubMed 32133691]
  138. Lee A, Bridges LR, Lloyd M, Barker R, Wren DR, Galtrey CM. 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]
  139. Lichtenstein GR, Hanauer SB, Sandborn WJ; Practice Parameters Committee of American College of Gastroenterology. Management of Crohn's disease in adults. Am J Gastroenterol. 2009;104(2):465-483; quiz 464, 484. doi:10.1038/ajg.2008.168 [PubMed 19174807]
  140. Lichtiger S, Present DH, Kornbluth A, et al. Cyclosporine in severe ulcerative colitis refractory to steroid therapy. N Engl J Med. 1994;330(26):1841-1845. doi: 10.1056/NEJM199406303302601. [PubMed 8196726]
  141. Lima RB, Benini V, Sens YA. Gingival overgrowth in renal transplant recipients: a study concerning prevalence, severity, periodontal, and predisposing factors. Transplant Proc. 2008;40(5):1425142-8. doi:10.1016/j.transproceed.2008.01.071 [PubMed 18589122]
  142. Lin CY, Lee SF. Comparison of pharmacokinetics between CsA capsules and Sandimmun Neoral in pediatric patients. Transplant Proc. 1994;26(5):2973-2974. [PubMed 7940938]
  143. 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]
  144. 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]
  145. Locatelli F, Zecca M, Rondelli R, et al. Graft versus host disease prophylaxis with low-dose cyclosporine-A reduces the risk of relapse in children with acute leukemia given HLA-identical sibling bone marrow transplantation: results of a randomized trial. Blood. 2000;95(5):1572-1579. [PubMed 10688810]
  146. 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]
  147. 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]
  148. 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]
  149. Mack CL, Adams D, Assis DN, et al. Diagnosis and management of autoimmune hepatitis in adults and children: 2019 practice guidance and guidelines from the American Association for the Study of Liver Diseases. Hepatology. 2020;72(2):671-722. doi:10.1002/hep.31065 [PubMed 31863477]
  150. Magnasco A, Rossi A, Catarsi P, et al. Cyclosporin and organ specific toxicity: clinical aspects, pharmacogenetics and perspectives. Curr Clin Pharmacol. 2008;3(3):166-173. doi:10.2174/157488408785747674 [PubMed 18781903]
  151. 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]
  152. 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]
  153. 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]
  154. Marsh J, Schrezenmeier H, Marin P, et al. Prospective randomized multicenter study comparing cyclosporin alone versus the combination of antithymocyte globulin and cyclosporin for treatment of patients with nonsevere aplastic anemia: a report from the European Blood and Marrow Transplant (EBMT) Severe Aplastic Anaemia Working Party. Blood. 1999;93(7):2191-2195. [PubMed 10090926]
  155. Martin P, Bleyzac N, Souillet G, et al. Clinical and pharmacological risk factors for acute graft-versus-host disease after paediatric bone marrow transplantation from matched-sibling or unrelated donors. Bone Marrow Transplant. 2003;32(9):881-887. doi:10.1038/sj.bmt.1704239 [PubMed 14561988]
  156. Mathews D, Mathews J, Jones NP. Low-dose cyclosporine treatment for sight-threatening uveitis: efficacy, toxicity, and tolerance. Indian J Ophthalmol. 2010;58(1):55-58. doi:10.4103/0301-4738.58472 [PubMed 20029146]
  157. Mathur H, Moretti AJ, Flaitz CM. Regression of cyclosporia-induced gingival overgrowth upon interruption of drug therapy. Gen Dent. 2003;51(2):159-162. [PubMed 15055689]
  158. McCrindle BW, Rowley AH, Newburger JW, et al. Diagnosis, treatment, and long-term management of Kawasaki disease: a scientific statement for health professionals from the American Heart Association. Circulation. 2017;135(17):e927-e999. doi:10.1161/CIR.0000000000000484 [PubMed 28356445]
  159. McDiarmid SV, Busuttil RW, Ascher NL, et al. FK506 (tacrolimus) compared with cyclosporine for primary immunosuppression after pediatric liver transplantation. Results from the U.S. Multicenter Trial. Transplantation. 1995;59(4):530-536. [PubMed 7533345]
  160. McIntyre HD, Menzies B, Rigby R, Perry-Keene DA, Hawley CM, Hardie IR. Long-term bone loss after renal transplantation: comparison of immunosuppressive regimens. Clin Transplant. 1995;9(1):20-24. [PubMed 7742578]
  161. 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]
  162. Memon M, deMagalhaes-Silverman M, Bloom EJ, et al. Reversible cyclosporine-induced cortical blindness in allogeneic bone marrow transplant recipients. Bone Marrow Transplant. 1995;15(2):283-286. [PubMed 7773219]
  163. Méndez A, Monforte V, Berastegui C, et al. High intra-individual variability of cyclosporine pharmacokinetics in lung transplant recipients without cystic fibrosis. Clin Transplant. 2014;28(6):743-748. doi:10.1111/ctr.12371 [PubMed 24708188]
  164. Menter A, Gelfand JM, Connor C, et al. Joint American Academy of Dermatology-National Psoriasis Foundation guidelines of care for the management of psoriasis with systemic nonbiologic therapies. J Am Acad Dermatol. 2020;82(6):1445-1486. doi:10.1016/j.jaad.2020.02.044 [PubMed 32119894]
  165. Mieli-Vergani G, Vergani D, Baumann U, et al. Diagnosis and management of pediatric autoimmune liver disease: ESPGHAN Hepatology Committee position statement. J Pediatr Gastroenterol Nutr. 2018;66(2):345-360. doi:10.1097/MPG.0000000000001801 [PubMed 29356770]
  166. Mihatsch MJ, Kyo M, Morozumi K, Yamaguchi Y, Nickeleit V, Ryffel B. The side-effects of ciclosporine-A and tacrolimus. Clin Nephrol. 1998;49(6):356-363. [PubMed 9696431]
  167. 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]
  168. Montero N, Pascual J. Immunosuppression and post-transplant hyperglycemia. Curr Diabetes Rev. 2015;11(3):144-154. doi:10.2174/1573399811666150331160846 [PubMed 25824238]
  169. Moreso F, Serón D, Morales JM, et al. Incidence of leukopenia and cytomegalovirus disease in kidney transplants treated with mycophenolate mofetil combined with low cyclosporine and steroid doses. Clin Transplant. 1998;12(3):198-205. [PubMed 9642510]
  170. Morgan JD, Swarbrick MJ, Edwards CM, Donnelly PK. Cyclosporin, nifedipine and gingival hyperplasia: a randomized controlled study. Transpl Int. 1994;7(suppl 1):S320-S321. doi:10.1111/j.1432-2277.1994.tb01380.x [PubMed 11271240]
  171. Mouyis M, Flint JD, Giles IP. Safety of anti-rheumatic drugs in men trying to conceive: a systematic review and analysis of published evidence [published online August 17, 2018]. Semin Arthritis Rheum. doi:10.1016/j.semarthrit.2018.07.011. [PubMed 30220537]
  172. Munar MY, Doyle IC, Meyer MM. Cyclosporine and vancomycin disposition during continuous venovenous hemodiafiltration. Ann Pharmacother. 1995;29(4):374-377. doi:10.1177/106002809502900406 [PubMed 7633014]
  173. Murphy CC, Greiner K, Plskova J, et al. Cyclosporine vs tacrolimus therapy for posterior and intermediate uveitis. Arch Ophthalmol. 2005;123(5):634-641. doi:10.1001/archopht.123.5.634 [PubMed 15883282]
  174. 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]
  175. 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]
  176. Nash RA, Antin JH, Karanes C, et al. Phase 3 study comparing methotrexate and tacrolimus with methotrexate and cyclosporine for prophylaxis of acute graft-versus-host disease after marrow transplantation from unrelated donors. Blood. 2000;96(6):2062-2068. [PubMed 10979948]
  177. Nastasio S, Sciveres M, Matarazzo L, et al. Long-term follow-up of children and young adults with autoimmune hepatitis treated with cyclosporine. Dig Liver Dis. 2019;51(5):712-718. doi:10.1016/j.dld.2018.10.018 [PubMed 30502231]
  178. Nemecek BD, Hammond DA, eds. Demystifying Drug Dosing in Renal Dysfunction. American Society of Health-System Pharmacists; 2019.
  179. Neoral (cyclosporine) [prescribing information]. East Hanover, NJ: Novartis; September 2023.
  180. Neoral (cyclosporine) [prescribing information]. East Hanover, NJ: Novartis; March 2015.
  181. Neoral (cyclosporine) [product monograph]. Montreal, Quebec, Canada: Novartis Pharmaceuticals Canada Inc; February 2023.
  182. Neunert C, Terrell DR, Arnold DM, et al. American Society of Hematology 2019 guidelines for immune thrombocytopenia. Blood Adv. 2019;3(23):3829-3866. doi:10.1182/bloodadvances.2019000966 [PubMed 31794604]
  183. Niese D. A double-blind randomized study of Sandimmun Neoral versus Sandimmun in new renal transplant recipients: results after 12 months. The International Sandimmun Neoral Study Group. Transplant Proc. 1995;27(2):1849-1856. [PubMed 7725531]
  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. Osuji N, Matutes E, Tjonnfjord G, et al. T-cell large granular lymphocyte leukemia: a report on the treatment of 29 patients and a review of the literature. Cancer. 2006;107(3):570-578. doi:10.1002/cncr.22032 [PubMed 16795070]
  186. Ozdal PC, Ortaç S, Taskintuna I, Firat E. Long-term therapy with low dose cyclosporin A in ocular Behçet's disease. Doc Ophthalmol. 2002;105(3):301-312. doi:10.1023/a:1021227019915 [PubMed 12539855]
  187. Page RL 2nd, Ruscin JM, Fish D, Lapointe M. Possible interaction between intravenous azithromycin and oral cyclosporine. Pharmacotherapy. 2001;21(11):1436-1443. doi:10.1592/phco.21.17.1436.34434 [PubMed 11714218]
  188. Parhar KS, Gibson PS, Coffin CS. Pregnancy following liver transplantation: review of outcomes and recommendations for management. Can J Gastroenterol. 2012;26(9):621-626. doi:10.1155/2012/137129 [PubMed 22993734]
  189. Passfall J, Schuller I, Keller F. Pharmacokinetics of cyclosporin during administration of danazol. Nephrol Dial Transplant. 1994;9(12):1807-1878. [PubMed 7708271]
  190. Peffault de Latour R, Kulasekararaj A, Iacobelli S, et al; Severe Aplastic Anemia Working Party of the European Society for Blood and Marrow Transplantation. Eltrombopag added to immunosuppression in severe aplastic anemia. N Engl J Med. 2022;386(1):11-23. doi:10.1056/NEJMoa2109965 [PubMed 34986284]
  191. 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;31(5):CD008817. doi:10.1002/14651858.CD008817.pub2 [PubMed 23728681]
  192. 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):977-183. doi:10.1097/00007890-199704150-00013 [PubMed 9112351]
  193. Plank C, Kalb V, Hinkes B, et al. Cyclosporin A is superior to cyclophosphamide in children with steroid-resistant nephrotic syndrome-a randomized controlled multicentre trial by the Arbeitsgemeinschaft für Pädiatrische Nephrologie. Pediatr Nephrol. 2008;23(9):1483-1493. doi:10.1007/s00467-008-0794-1 [PubMed 18481113]
  194. Pollard S, Nashan B, Johnston A, et al; CONSENT: Consensus on Substitution in European Transplantation. A pharmacokinetic and clinical review of the potential clinical impact of using different formulations of cyclosporin A. Berlin, Germany, November 19, 2001. Clin Ther. 2003;25(6):1654-1669. doi:10.1016/s0149-2918(03)80161-3 [PubMed 12860490]
  195. Porrini E, Delgado P, Alvarez A, et al. The combined effect of pre-transplant triglyceride levels and the type of calcineurin inhibitor in predicting the risk of new onset diabetes after renal transplantation. Nephrol Dial Transplant. 2008;23(4):1436-1441. doi:10.1093/ndt/gfm762 [PubMed 18029372]
  196. 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]
  197. Provan D, Arnold DM, Bussel JB, et al. Updated international consensus report on the investigation and management of primary immune thrombocytopenia. Blood Adv. 2019;3(22):3780-3817. doi:10.1182/bloodadvances.2019000812 [PubMed 31770441]
  198. Provan D, Stasi R, Newland AC, et al. International consensus report on the investigation and management of primary immune thrombocytopenia. Blood. 2010;115(2):168-186. doi:10.1182/blood-2009-06-225565 [PubMed 19846889]
  199. Ptachcinski RJ, Venkataramanan R, Burckart GJ. Clinical pharmacokinetics of cyclosporin. Clin Pharmacokinet. 1986;11(2):107-132. doi:10.2165/00003088-198611020-00002 [PubMed 3514043]
  200. Ratanatharathorn V, Nash RA, Przepiorka D, et al. Phase III study comparing methotrexate and tacrolimus (prograf, FK506) with methotrexate and cyclosporine for graft-versus-host disease prophylaxis after HLA-identical sibling bone marrow transplantation. Blood. 1998;92(7):2303-2314. [PubMed 9746768]
  201. Reali L, Zuliani E, Gabutti L, Schönholzer C, Marone C. Poor oral hygiene enhances gingival overgrowth caused by calcineurin inhibitors. J Clin Pharm Ther. 2009;34(3):255-260. doi:10.1111/j.1365-2710.2008.01000.x [PubMed 19646074]
  202. Refer to manufacturer's labeling.
  203. Remuzzi G, Bertani T. Renal vascular and thrombotic effects of cyclosporine. Am J Kidney Dis. 1989;13(4):261-272. doi:10.1016/s0272-6386(89)80032-0 [PubMed 2650537]
  204. Rodrigo E, Ruiz JC, Angeles de Cos M, et al. Correlation of C0 and C2 levels with cyclosporine side effects in kidney transplantation. Transplant Proc. 2009;41(6):2328-2331. doi:10.1016/j.transproceed.2009.06.155 [PubMed 19715910]
  205. Ross M, Schmidt GM, Niland JC, et al. Cyclosporine, methotrexate, and prednisone compared with cyclosporine and prednisone for prevention of acute graft-vs.-host disease: effect on chronic graft-vs.-host disease and long-term survival. Biol Blood Marrow Transplant. 1999;5(5):285-291. doi:10.1016/s1083-8791(99)70003-0 [PubMed 10534058]
  206. Rovin BH, Adler SG, Barratt J, et al. Executive summary of the KDIGO 2021 guideline for the management of glomerular diseases. Kidney Int. 2021;100(4):753-779. doi:10.1016/j.kint.2021.05.015 [PubMed 34556300]
  207. Rubin DT, Ananthakrishnan AN, Siegel CA, Sauer BG, Long MD. ACG clinical guideline: ulcerative colitis in adults. Am J Gastroenterol. 2019;114(3):384-413. doi:10.14309/ajg.0000000000000152. [PubMed 30840605]
  208. Ruutu T, Gratwohl A, de Witte T, et al. Prophylaxis and treatment of GVHD: EBMT-ELN working group recommendations for a standardized practice. Bone Marrow Transplant. 2014;49(2):168-173. doi:10.1038/bmt.2013.107 [PubMed 23892326]
  209. Sairanen J, Forsell T, Ruutu M. Long-term outcome of patients with interstitial cystitis treated with low dose cyclosporine A. J Urol. 2004;171(6 pt 1):2138-2141. doi:10.1097/01.ju.0000125139.91203.7a [PubMed 15126772]
  210. Sairanen J, Hotakainen K, Tammela TL, Stenman UH, Ruutu M. Urinary epidermal growth factor and interleukin-6 levels in patients with painful bladder syndrome/interstitial cystitis treated with cyclosporine or pentosan polysulfate sodium. Urology. 2008;71(4):630-633. doi:10.1016/j.urology.2007.11.055 [PubMed 18387391]
  211. 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 [PubMed 32090480]
  212. 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]
  213. 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]
  214. Sandimmune (cyclosporine) capsules, injection, oral solution [prescribing information]. East Hanover, NJ: Novartis Pharmaceuticals; September 2023.
  215. Sandimmune (cyclosporine) [prescribing information]. East Hanover, NJ: Novartis; March 2015.
  216. Sandimmune I.V. (cyclosporine) [product monograph]. Montreal, Quebec, Canada: Novartis Pharmaceuticals Canada Inc; February 2023.
  217. Scalzini A, Barni C, Stellini R, Sueri L. Fatal invasive aspergillosis during cyclosporine and steroids treatment for Crohn's disease. Dig Dis Sci. 1995;40(3):528. doi:10.1007/BF02064361 [PubMed 7895537]
  218. Scheinberg P, Nunez O, Weinstein B, et al. Horse versus rabbit antithymocyte globulin in acquired aplastic anemia. N Engl J Med. 2011;365(5):430-438. doi:10.1056/NEJMoa1103975 [PubMed 21812672]
  219. Schiff J, Cole E, Cantarovich M. Therapeutic monitoring of calcineurin inhibitors for the nephrologist. Clin J Am Soc Nephrol. 2007;2(2):374-384. doi:10.2215/CJN.03791106 [PubMed 17699437]
  220. Schneider BJ, Naidoo J, Santomasso BD, et al. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: ASCO guideline update. J Clin Oncol. 2021;39(36):4073-4126. doi:10.1200/JCO.21.01440 [PubMed 34724392]
  221. Sciveres M, Caprai S, Palla G, Ughi C, Maggiore G. Effectiveness and safety of ciclosporin as therapy for autoimmune diseases of the liver in children and adolescents. Aliment Pharmacol Ther. 2004;19(2):209-217. doi:10.1046/j.1365-2036.2003.01754.x [PubMed 14723612]
  222. Seyfinejad B, Jouyban A. Overview of therapeutic drug monitoring of immunosuppressive drugs: analytical and clinical practices. J Pharm Biomed Anal. 2021;205:114315. doi:10.1016/j.jpba.2021.114315 [PubMed 34399192]
  223. Seymour RA, Jacobs DJ. Cyclosporin and the gingival tissues. J Clin Periodontol. 1992;19(1):1-11. doi:10.1111/j.1600-051x.1992.tb01140.x [PubMed 1732303]
  224. 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]
  225. Sharma RK, Kumar P, Rai P, et al. Cyclosporine neurotoxicity in a renal-transplant recipient. Nephron. 1995;70(2):269. doi:10.1159/000188601 [PubMed 7566320]
  226. Shehab N, Lewis CL, Streetman DD, Donn SM. Exposure to the pharmaceutical excipients benzyl alcohol and propylene glycol among critically ill neonates. Pediatr Crit Care Med. 2009;10(2):256-259. doi:10.1097/PCC.0b013e31819a383c [PubMed 19188870]
  227. 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]
  228. 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]
  229. Sood A, Midha V, Sood N. Reversible posterior leukoencephalopathy due to oral cyclosporine in severe ulcerative colitis. Indian J Gastroenterol. 2003;22(6):233-234. [PubMed 15030042]
  230. Storb R, Deeg HJ, Farewell V, et al. Marrow transplantation for severe aplastic anemia: methotrexate alone compared with a combination of methotrexate and cyclosporine for prevention of acute graft-versus-host disease. Blood. 1986a;68(1):119-125. [PubMed 3521761]
  231. Storb R, Deeg HJ, Whitehead J, et al. Methotrexate and cyclosporine compared with cyclosporine alone for prophylaxis of acute graft versus host disease after marrow transplantation for leukemia. N Engl J Med. 1986b;314(12):729-735. doi:10.1056/NEJM198603203141201 [PubMed 3513012]
  232. 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]
  233. Suzuki H, Terai M, Hamada H, et al. Cyclosporin A treatment for Kawasaki disease refractory to initial and additional intravenous immunoglobulin. Pediatr Infect Dis J. 2011;30(10):871-876. doi:10.1097/INF.0b013e318220c3cf [PubMed 21587094]
  234. Suzuki K, Tanaka H, Tsugawa K, Ito E. Effective treatment with cyclosporine A of a child with systemic lupus erythematosus resistant to cyclophosphamide pulse therapy. Tohoku J Exp Med. 2006;208(4):355-359. doi:10.1620/tjem.208.355 [PubMed 16565599]
  235. 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]
  236. Taesch S, Niese D, Mueller EA. Sandimmun neoral, a new oral formulation of cyclosporin A with improved pharmacokinetic characteristics: safety and tolerability in renal transplant patients. Transplant Proc. 1994;26(6):3147-3149. [PubMed 7998097]
  237. ten Tije AJ, Verweij J, Loos WJ, et al. Pharmacological effects of formulation vehicles : implications for cancer chemotherapy. Clin Pharmacokinet. 2003;42(7):665-685. doi:10.2165/00003088-200342070-00005 [PubMed 12844327]
  238. Thomas DW, Newcombe RG, Osborne GR. Risk factors in the development of cyclosporine-induced gingival overgrowth. Transplantation. 2000;69(4):522-5226. doi:10.1097/00007890-200002270-00010 [PubMed 10708105]
  239. Tindall RS, Phillips JT, Rollins JA, Wells L, Hall K. A clinical therapeutic trial of cyclosporine in myasthenia gravis. Ann N Y Acad Sci. 1993;681:539-551. doi:10.1111/j.1749-6632.1993.tb22937.x [PubMed 8357194]
  240. 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]
  241. 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]
  242. Tremoulet AH, Pancoast P, Franco A, et al. Calcineurin inhibitor treatment of intravenous immunoglobulin-resistant Kawasaki disease. J Pediatr. 2012;161(3):506-512. doi:10.1016/j.jpeds.2012.02.048 [PubMed 22484354]
  243. Tugwell P, Pincus T, Yocum D, et al. Combination therapy with cyclosporine and methotrexate in severe rheumatoid arthritis. The Methotrexate-Cyclosporine Combination Study Group. N Engl J Med. 1995;333(3):137-141. doi:10.1056/NEJM199507203330301 [PubMed 7791814]
  244. Tzakis AG, Reyes J, Todo S, et al. FK 506 versus cyclosporine in pediatric liver transplantation. Transplant Proc. 1991;23(6):3010-3015. [PubMed 1721343]
  245. 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. Updated September 2016. Accessed October 5, 2016. https://www.cdc.gov/niosh/docs/2016-161/default.html.
  246. US Multicenter FK506 Liver Study Group. A comparison of tacrolimus (FK 506) and cyclosporine for immunosuppression in liver transplantation. N Engl J Med. 1994;331(17):1110-1115. doi:10.1056/NEJM199410273311702 [PubMed 7523946]
  247. Valapour M, Lehr CJ, Skeans MA, et al. OPTN/SRTR 2020 annual data report: lung. Am J Transplant. 2022;22(suppl 2):438-518. doi:10.1111/ajt.16991 [PubMed 35266615]
  248. Van Assche G, D'Haens G, Noman M, et al. Randomized, double-blind comparison of 4 mg/kg versus 2 mg/kg intravenous cyclosporine in severe ulcerative colitis. Gastroenterology. 2003;125(4):1025-1031. doi:10.1016/s0016-5085(03)01214-9. [PubMed 14517785]
  249. Van den Hof WF, Ruiz-Aracama A, Van Summeren A, et al. Integrating multiple omics to unravel mechanisms of Cyclosporin A induced hepatotoxicity in vitro. Toxicol In Vitro. 2015;29(3):489-501. doi:10.1016/j.tiv.2014.12.016 [PubMed 25562108]
  250. Varga E, Lennon MA, Mair LH. Pre-transplant gingival hyperplasia predicts severe cyclosporin-induced gingival overgrowth in renal transplant patients. J Clin Periodontol. 1998;25(3):225-230. doi:10.1111/j.1600-051x.1998.tb02432.x [PubMed 9543193]
  251. 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]
  252. 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]
  253. Venkataramanan R, Ptachcinski RJ, Burckart GJ, Yang SL, Starzl TE, Van Theil DH. The clearance of cyclosporine by hemodialysis. J Clin Pharmacol. 1984;24(11-12):528-531. doi:10.1002/j.1552-4604.1984.tb02763.x [PubMed 6392355]
  254. Videla C, Vega J, Borja H. Hepatotoxicity associated with cyclosporine monitoring using C2 recommendations in adults renal recipients receiving ketoconazole. Transplant Proc. 2005;37(3):1574-1576. doi:10.1016/j.transproceed.2004.09.011 [PubMed 15866677]
  255. 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]
  256. Vincenti F, Mendez R, Curtis J, et al. A multicenter, prospective study of C2-monitored cyclosporine microemulsion in a U.S. population of de novo renal transplant recipients. Transplantation. 2005;80(7):910-916. doi:10.1097/01.tp.0000173802.70980.50 [PubMed 16249738]
  257. V'lckova-Laskoska MT. Cyclosporin A-induced gingival hyperplasia in psoriasis: review of the literature and case reports. Acta Dermatovenerol Croat. 2005;13(2):108-113. [PubMed 16324424]
  258. Weber A, Fein F, Koch S, et al. Treatment of ulcerative colitis refractory to steroid therapy by oral microemulsion cyclosporine (Neoral). Inflamm Bowel Dis. 2006;12(12):1131-1135. doi: 10.1097/01.mib.0000235096.78736.8e. [PubMed 17119387]
  259. Weinstein S, Lipsitz EC, Addonizio L, Stolar CJ. Cholelithiasis in pediatric cardiac transplant patients on cyclosporine. J Pediatr Surg. 1995;30(1):61-64. doi:10.1016/0022-3468(95)90611-8 [PubMed 7722832]
  260. 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]
  261. Weir MR, Fink JC. Risk for posttransplant Diabetes mellitus with current immunosuppressive medications. Am J Kidney Dis. 1999;34(1):1-13. doi:10.1016/s0272-6386(99)70101-0 [PubMed 10401009]
  262. Wells G, Tugwell P. Cyclosporin A in rheumatoid arthritis: overview of efficacy. Br J Rheumatol. 1993;32(suppl 1):51-56. [PubMed 8448639]
  263. Wolff D, Schleuning M, von Harsdorf S, et al. Consensus conference on clinical practice in chronic GVHD: second-line treatment of chronic graft-versus-host disease. Biol Blood Marrow Transplant. 2011;17(1):1-17. doi:10.1016/j.bbmt.2010.05.011 [PubMed 20685255]
  264. Wu Q, Wang X, Nepovimova E, Wang Y, Yang H, Kuca K. Mechanism of cyclosporine A nephrotoxicity: Oxidative stress, autophagy, and signalings. Food Chem Toxicol. 2018;118:889-907. doi:10.1016/j.fct.2018.06.054 [PubMed 29960018]
  265. Yee GC. Recent advances in cyclosporine pharmacokinetics. Pharmacotherapy. 1991;11(5):130S-134S. [PubMed 1745619]
  266. Zaghetto JM, Yamamoto MM, Souza MB, et al. Chlorambucil and cyclosporine A in Brazilian patients with Behçet's disease uveitis: a retrospective study. Arq Bras Oftalmol. 2010;73(1):40-46. doi:10.1590/s0004-27492010000100007 [PubMed 20464112]
  267. Zakhem GA, Goldberg JE, Motosko CC, Cohen BE, Ho RS. Sexual dysfunction in men taking systemic dermatologic medication: a systematic review. J Am Acad Dermatol. 2019;81(1):163-172. doi:10.1016/j.jaad.2019.03.043 [PubMed 30905792]
  268. Zar T, Graeber C, Perazella MA. Recognition, treatment, and prevention of propylene glycol toxicity. Semin Dial. 2007;20(3):217-219. doi:10.1111/j.1525-139X.2007.00280.x [PubMed 17555487]
  269. 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]
  270. Zortress (everolimus) [prescribing information]. East Hanover, NJ: Novartis Pharmaceutical Co; August 2018.
  271. 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]
  272. Zver S, Zupan IP, Cernelc P. Cyclosporin A as an immunosuppressive treatment modality for patients with refractory autoimmune thrombocytopenic purpura after splenectomy failure. Int J Hematol. 2006;83(3):238-242. doi:10.1532/IJH97.05149 [PubMed 16720554]
Topic 9094 Version 534.0

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