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

Agents for maintenance of immunosuppression following lung transplantation

Agents for maintenance of immunosuppression following lung transplantation
Drug* Mechanism of action Suggested dose* Drug monitoring Metabolism/transporter effects with selected drug interactions Common major adverse effects Comments
Glucocorticoids
Methylprednisolone, prednisone

Inhibits humoral and cell-mediated immunity

Binds with DNA sequences (+/– nuclear factor-kB) to inhibit production of inflammatory cytokines

At time of transplant: 500 to 1000 mg methylprednisolone intravenously

Maintenance oral prednisone dose: 0.5 to 1 mg/kg per day initially after transplant with taper to a goal of 5 to 10 mg per day over several months to one year

Protocols may vary by institution
N/A

Cytochrome P450 3A4 substrate

Numerous clinically relevant interactions in transplant patientsΔ

Diabetes

GERD, PUD

Osteoporosis

Skeletal muscle wasting

Hypertension

Hypercholesterolemia

Change in appetite

Weight gain
Do not stop abruptly; may be taken with food to reduce dyspepsia
Calcineurin inhibitors
Cyclosporine Prevents T cell activation and proliferation by inhibiting the production of interleukins and other cytokines

2 to 3 mg/kg per day intravenously at time of transplantation given in a 24 hour infusion or 1 to 1.5 mg/kg in two or more four hour infusions and until patient can tolerate oral intake. Adjust dose to target trough concentration of 250 to 350 ng/mL and two hour post dose (C2) concentration of 900 to 1200 ng/mL.

Maintenance oral dose: 3 to 5 mg/kg twice per day 12 hours apart at a consistent time each day in relation to meals and adjusted according to trough or C2 concentration as above.

Monitor BUN, Cr, magnesium, potassium

Trough levels typically used but levels drawn two hours post dose (C2) are most accurate

Metabolized by cytochrome P450 enzyme system

Cytochrome P450 3A4 substrate/inhibitor, P-glycoprotein substrate/inhibitor

Marked increase in levels of statin drugs when co-administered

Numerous clinically relevant interactions in transplant patientsΔ

Renal dysfunction including acute nephrotoxicity

Hypertension

Hypercholesterolemia

Gingival hyperplasia

Neurotoxicity (tremor, headache, encephalopathy, focal deficits)

Hirsutism

High inter- and intra-individual absorption variability.

Blood concentration monitoring is necessary for any change in formulation (including switch between generics or brands) to determine need for dose alteration. The microemulsion form, which is also known as "modified" (Neoral) is generally better absorbed than the non-modified form (Sandimmune).
Tacrolimus Prevents T cell activation and proliferation by inhibiting the production of interleukins and other cytokines

Initial dose sublingually: 0.04 to 0.05 mg/kg per day in two divided doses (eg, 1 to 2 mg sublingually every 12 hours)[1,2]§; OR

Initial dose intravenously: 0.03 to 0.05 mg/kg per day by continuous infusion over 24 hours; risk of infusion reactions and other toxicities (refer to accompanying text)

Maintenance oral dose: 0.05 mg/kg twice daily taken 12 hours apart at a consistent time each day in relation to meals

Initial and maintenance: Adjust dose to target trough level of 10 to 15 ng/mL for months 1 to 3; 8 to 13 ng/mL for months 4 to 12; and 6 to 8 ng/mL after the first 12 months, although target levels vary based on center practice and clinical circumstances

Monitor glucose, LFTs, BUN, Cr, calcium, magnesium, potassium

Monitor trough levels

Metabolized by cytochrome P450 enzyme system

Cytochrome P450 3A4 substrate, P-glycoprotein substrate

Numerous clinically relevant interactions in transplant patientsΔ

Renal dysfunction

Diabetes

Hypertension (less than cyclosporine)

Hypercholesterolemia

Altered mental status

Headache

Focal neurological deficits

High inter- and intra-individual absorption variability

Blood concentration monitoring is necessary for change in formulation (including switch between generics or brands) and between methods of administration (eg, sublingual, oral, and IV administration) to determine need for dose alteration
Antimetabolites
Mycophenolate mofetil (CellCept)

Nucleotide blocking agent

Inhibits T cell proliferation by blocking nucleotide synthesis

IV equivalent to oral

Starting dose: 1000 to 1500 mg twice daily within 72 hours after transplantation

Oral doses should be taken on an empty stomach or at a consistent time each day in relation to meals if taken with food to improve tolerability

Giving total daily dose in three or four equally divided doses might improve GI tolerability

Monitor WBC

Dose adjusted for leukopenia

Serum concentration monitoring not routinely performed¥
Levels decreased by:¥
  • Antacids
  • Aluminum hydroxide
  • Magnesium (oral)
  • Bile acid sequestrants (eg, cholestyramine)
  • Cyclosporine (but not tacrolimus)
  • Magnesium (oral)
  • Proton pump inhibitors
  • Rifamycins (eg, rifampin)

Levels increased by:¥

  • Acyclovir
  • Probenecid

May inactivate protein bound drugs, especially hormonal contraceptives (use nonhormonal method of contraception)

Nausea, vomiting, diarrhea, abdominal pain

Diarrhea may occur after months of treatment

GI tolerability may improve with more frequent dosing (same total daily dose) or by changing to enteric-coated mycophenolate sodium (below)

Myelosuppression, anemia

Increased risk of CMV disease
Serum concentration monitoring is not routinely performed¥
Mycophenolate sodium, enteric coated (Myfortic)

Nucleotide blocking agent

Inhibits T cell proliferation by blocking nucleotide synthesis

Starting dose: 720 to 1080 mg orally twice daily on an empty stomach

To convert from mycophenolate mofetil 1000 mg every 12 hours, switch to mycophenolate sodium enteric coated 720 mg every 12 hours

Monitor WBC

Dose adjusted for leukopenia

Serum concentration monitoring not routinely performed¥
Levels decreased by:¥
  • Antacids
  • Aluminum hydroxide
  • Bile acid sequestrants (eg, cholestyramine)
  • Cyclosporine (but not tacrolimus)
  • Magnesium (oral)
  • Rifamycins (eg, rifampin)

Levels increased by:¥

  • Acyclovir
  • Probenecid

May inactivate protein bound drugs, especially hormonal contraceptives (use nonhormonal method of contraception)

Nausea, vomiting

Diarrhea

Myelosuppression, anemia

Increased risk of CMV disease
Serum concentration monitoring is not routinely performed¥
Azathioprine

Nucleotide blocking agent

Inhibits T and B cell proliferation by blocking nucleotide synthesis

IV equivalent to oral

Starting dose: 1 to 2 mg/kg daily and adjusted to prevent development of leukopenia

Monitor WBC

Dose adjusted for leukopenia

Monitor LFTs
Levels increased by:
  • Allopurinol (Preferably avoid co-administration. If co-administration required, reduce azathioprine by up to 70%.)

Use with febuxostat is contraindicated

May diminish anticoagulant effects of warfarin

Nausea, vomiting

Diarrhea

Bone marrow suppression

Liver abnormality
Requires TPMT enzyme for metabolism. Individuals with profound initial side effects may be deficient in this enzyme.
mTOR inhibitors
Sirolimus Inhibits T cell proliferation by cell cycle arrest in G1 phase

Sirolimus is initiated at least three months post-transplantation, due to effects on wound healing.

Liquid and tablet form.

Starting dose: 2 mg orally per day. Adjusted to maintain a trough target between 8 and 12 ng/mL when used without calcineurin inhibitor. Adjust to maintain trough level between 4 and 8 ng/mL when used with calcineurin inhibitor.

Monitor CBC, LFTs

Obtain trough level three to four days after initiation and seven days after any change in dose

Metabolized by cytochrome P450 3A enzyme system

Cytochrome P450 3A4 substrate, P-glycoprotein substrate

Cyclosporine dose should be decreased by one-half to two-thirds when co-administered with sirolimus[3]

Numerous clinically relevant interactions in transplant patientsΔ

Delayed wound healing

Fatal airway anastomotic dehiscence if administered early after lung transplantation

Myelosuppression

Hypercholesterolemia

Pulmonary toxicity

LFT abnormalities

Diarrhea

Nausea
Associated with increased incidence of deep venous thrombosis[4]
Everolimus Inhibits T cell proliferation by cell cycle arrest in G1 phase

Everolimus is initiated at least three months post-transplantation, due to effects on wound healing

Starting dose: 1.5 mg orally every 12 hours and adjusted to maintain a trough target between 3 and 12 ng/mL in combination with cyclosporine and a glucocorticoid

In combination with tacrolimus, higher doses of everolimus may be needed to maintain trough levels within target range compared to doses when used with cyclosporine

Monitor CBC, LFTs, BUN, Cr, glucose

Monitor trough levels; steady state levels are reached four to five days after a dose change

Metabolized by cytochrome P450 3A enzyme system

Cytochrome P450 3A4 substrate, P-glycoprotein substrate

Cyclosporine dose should be decreased by one-half to two-thirds when co-administered with everolimus

Numerous clinically relevant interactions in transplant patientsΔ

Delayed wound healing

Bone marrow suppression

Hypercholesterolemia

Pulmonary toxicity

Diarrhea

Nausea
 

GERD: gastroesophageal reflux disease; PUD: peptic ulcer disease; Cr: creatinine; LFT: liver function tests; BUN: blood urea nitrogen; ECG: electrocardiogram; WBC: white blood cell count; CMV: cytomegalovirus; MPA: mycophenolic acid (active metabolite of mycophenolate); TPMT: thiopurine methyltransferase; mTOR: mechanistic target of rapamycin; CBC: complete blood count.

* Initial immunosuppressant doses shown should be adjusted based upon patient-specific factors including organ function and potential drug interactions. Drug therapy should be managed by transplant specialists with expertise in therapeutic drug monitoring and doses should be adjusted based upon measurement of immunosuppressant concentrations. The US Food and Drug Administration has approved tacrolimus for use in lung transplantation, but other medications have not been approved. The doses are suggested based on the experience of large lung transplantation centers. Dosing protocols vary by institution.

¶ Immunosuppressants are subject to numerous drug interactions, particularly with drugs or foods that inhibit or induce cytochrome CYP 450 3A4 and/or P-glycoprotein transporters (P-gp). Drug therapy should be managed by transplant specialists with expertise in therapeutic drug monitoring and doses adjusted based upon measurement of immunosuppressant concentrations, particularly whenever drug therapy is altered. The table is NOT a complete list of all possible interactions. To determine specific drug interactions and suggestions for management, consult a drug interactions database such as Lexicomp drug interactions included with UpToDate.

Δ For additional information, refer to UpToDate table on major drug interactions with immunosuppressants: cyclosporine, tacrolimus, sirolimus, or everolimus.

◊ For additional information, refer to UpToDate topic on pharmacology and side effects of cyclosporine and tacrolimus.

§ Sublingual administration of tacrolimus may be an alternative for lung transplant recipients who are unable to swallow capsules[1,2].

¥ Serum levels of mycophenolate active metabolite (MPA) can be altered by some drug interactions. However, adjustment of mycophenolate dose may not be necessary depending upon the expected effect of the interaction and patient factors (eg, rejection risk); single MPA serum levels are not reliable for determining mycophenolate exposure or predicting efficacy and are not routinely performed (refer to accompanying text).
References:
  1. Tsapepas D, Saal S, Benkert S, et al. Sublingual tacrolimus: a pharmacokinetic evaluation pilot study. Pharmacotherapy 2013; 33:31.
  2. Watkins KD, Boettger RF, Hanger KM, et al. Use of sublingual tacrolimus in lung transplant recipients. J Heart Lung Transplant 2012; 31:127.
  3. Glanville AR, Aboyoun C, Klepetko W, et al. Three-year results of an investigator-driven multicenter, international, randomized open-label de novo trial to prevent BOS after lung transplantation. J Heart Lung Transplant 2015; 34:16.
  4. Ahya VN, McShane PJ, Baz MA, et al. Increased risk of venous thromboembolism with a sirolimus based immunosuppression regimen in lung transplantation. J Heart Lung Transplant 2011; 30:175.

Prepared with data from:

  1. Bhorade SM, Stern E. Immunosuppression for lung transplantation. Proc Am Thorac Soc 2009; 6:47.
  2. Korom S, Boehler A, Weder W. Immunosuppressive therapy in lung transplantation: state of the art. Eur J Cardiothorac Surg 2009; 35:1045.
  3. Floreth T, Bhorade SM. Current trends in immunosuppression for lung transplantation. Semin Respir Crit Care Med 2010; 31:172.
Graphic 60918 Version 14.0

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