VA Class:AN100
Carmustine, a nitrosourea-derivative alkylating agent, is an antineoplastic agent.
Carmustine is used as an adjunct to radiation therapy following surgery for malignant glioma, such as glioblastoma multiforme and anaplastic astrocytoma.120,125 The addition of carmustine to radiation therapy has not been shown to increase survival time, but a trend toward a higher long-term survival rate (e.g., at 18 months) has been observed.126,127 Carmustine also has been used as adjuvant or salvage therapy for oligodendroglioma, a uniquely chemosensitive form of glioma.120,127,128
The benefit of adjuvant chemotherapy for the treatment of malignant gliomas has not been established.125,129 Although individual studies do not show a definitive benefit, analysis of pooled data130 from the reported results of several published randomized studies suggests that the use of adjuvant chemotherapy prolongs survival in adults with malignant gliomas. However, in a large randomized trial, the addition of lomustine, another nitrosourea, in combination with procarbazine and vincristine to radiation therapy did not prolong median survival or increase the survival rate in patients with high-grade astrocytoma.131
Compared with IV administration, intra-arterial administration of carmustine is associated with excessive toxicity, including blindness and fatal encephalopathy, and inferior survival.132
Adjunctive Therapy at Primary Surgery
The intracranial carmustine wafer implant is used as an adjunct to surgery and radiation therapy for newly diagnosed high-grade malignant glioma.124,135
This indication is based mainly on the results of a randomized, double-blind, placebo-controlled trial of 240 patients with newly diagnosed malignant glioma, including glioblastoma multiforme (207 patients), anaplastic oligoastrocytoma (11 patients), anaplastic oligodendroglioma (11 patients), and anaplastic astrocytoma (2 patients), who were undergoing initial craniotomy for tumor resection.124,152 Treatment regimens consisted of surgery with implantation of either carmustine wafers or placebo wafers (typically 6-8 wafers), and most patients (77.5% in the carmustine wafer group and 81.7% in the placebo wafer group) received a standard course of radiation therapy (55-60 Gy) typically initiated at 3 weeks following surgery.124 Systemic chemotherapy was administered to 17 patients (14%) receiving the carmustine wafer and 12 patients (10%) receiving the placebo wafer; all 6 patients with anaplastic oligodendroglioma received chemotherapy within 30 days of surgical implantation of the carmustine wafer.124
At a minimum follow-up of 3 years, patients receiving the carmustine wafer implant had prolonged median survival (13.9 versus 11.6 months; hazard ratio: 0.73) compared with those receiving the placebo wafer implant.124,152 Subgroup analysis of patients with glioblastoma multiforme did not show any difference in survival for patients receiving the carmustine wafer implant versus the placebo wafer implant.124,152 Although intracranial hypertension (9 versus 2%) and CSF leak (5 versus 0.8%) occurred more frequently in patients receiving the carmustine wafer, the incidence of other adverse neurologic effects or any adverse effect occurring in at least 5% of patients did not differ between those receiving the carmustine wafer or the placebo wafer.124,152
Adjunctive Therapy at Surgery for Recurrent Disease
The intracranial carmustine wafer implant is used as an adjunct to surgery for the palliative treatment of recurrent glioblastoma multiforme in patients for whom surgical resection is indicated.120,124,135,137
This indication is based principally on a randomized, double-blind, placebo-controlled clinical trial of 222 patients undergoing surgical resection for gliomas that recurred following initial surgery and radiation therapy.124,133 Treatment regimens consisted of surgery with implantation of either carmustine wafers or placebo wafers (typically 7-8 wafers).124,133 Chemotherapy was withheld at least 4 weeks (6 weeks for nitrosoureas) prior to and 2 weeks after surgery.124 Among all patients with recurrent malignant glioma, patients receiving carmustine wafer implants had a similar median survival (32 versus 24 weeks) and a similar 6-month survival rate (60 versus 47%) as patients receiving placebo wafer implants.124 No survival benefit was evident in patients who had pathologic diagnoses other than glioblastoma multiforme at the time of surgery.124,133 Among those with glioblastoma multiforme, patients receiving carmustine wafer implants had a longer median survival (28 versus 20 weeks) and an increased 6-month survival rate after surgery (56 versus 36%) compared with those receiving placebo wafer implants.124,133
Safety and efficacy of the carmustine wafer compared with conventional chemotherapy with IV carmustine for the treatment of brain tumors has not been evaluated in randomized trials to date.134
Carmustine-containing regimens are used as an alternative for the palliative treatment of multiple myeloma.105,120,138
Analysis of pooled data139 from randomized studies showed no difference in survival for patients receiving combination chemotherapy versus melphalan and prednisone, a regimen of choice120,138 for multiple myeloma. In a randomized trial, no difference in survival was observed in patients receiving salvage therapy with VBMCP (vincristine, carmustine, melphalan, cyclophosphamide, and prednisone) or VAD (vincristine, doxorubicin, dexamethasone) for relapsed or refractory multiple myeloma following initial treatment with cyclophosphamide and prednisone.140
Although carmustine is labeled for use in combination with other agents as secondary therapy for the treatment of refractory or relapsed Hodgkin's disease,105 combination regimens containing other agents currently are preferred as conventional chemotherapy for this cancer.120,141 The role of carmustine as a component of high-dose chemotherapy used as a preparatory regimen for bone marrow transplantation or peripheral blood stem cell transplantation in the treatment of refractory or relapsed Hodgkin's lymphoma is being investigated.141,142,143
Although carmustine is labeled for use in combination with other agents as secondary therapy for the treatment of refractory or relapsed non-Hodgkin's lymphomas,105 combination regimens containing other agents currently are preferred as conventional chemotherapy for these cancers.120,144 The role of carmustine as a component of high-dose chemotherapy used as a preparatory regimen for bone marrow transplantation in the treatment of refractory or relapsed non-Hodgkin's lymphomas is being investigated.144,145
Although carmustine has been used alone114,116 or in combination therapy114,116,119 for the palliative treatment of metastatic melanoma, low response rates and substantial toxicity have contributed to declining use of this agent.117,122 Response rates reported for carmustine appear to be lower than those reported for dacarbazine as a single agent, but randomized comparisons have not been done.114,116,122 Carmustine has been used in various combination regimens (e.g., carmustine, cisplatin, dacarbazine, and tamoxifen) for the treatment of metastatic melanoma.119 Although early studies reported high response rates for combination chemotherapy regimens for the treatment of metastatic melanoma, evidence from large, randomized trials has not established the superiority of combination regimens compared with dacarbazine alone.114,118,121 Dacarbazine monotherapy currently is a systemic treatment of choice for metastatic melanoma (see Uses: Melanoma in Dacarbazine 10:00 for an overview of therapy for melanoma).115,116,118,120,121,122 Although the nitrosoureas, such as carmustine, are lipid-soluble and can cross the blood-brain barrier, these agents appear to have no appreciable antitumor effect on cerebral metastases.116
Carmustine is used topically for the palliative treatment of cutaneous T-cell lymphoma (mycosis fungoides).120,123,149,150
Reconstitution and Administration
Carmustine is administered by IV infusion and intracranially as wafer implants.
Carmustine is administered by IV infusion. The drug is reconstituted by dissolving the contents of the vial labeled as containing 100 mg of carmustine with 3 mL of sterile dehydrated (absolute) alcohol, followed by addition of 27 mL of sterile water for injection. 105 The resultant solution contains 3.3 mg of carmustine per mL of 10% alcohol. This solution may be further diluted with 5% dextrose injection, and administered by IV infusion over a period of 1-2 hours; administration of carmustine over shorter periods of time may produce intense pain and burning at the injection site and along the vein. (See Cautions: Other Adverse Effects.) Carmustine also has been administered by intra-arterial (into the carotid artery) route; however, such administration has been associated with ocular toxicity.105
The manufacturer recommends that protective gloves be used during handling of carmustine powder and preparation of solutions of the drug, since accidental exposure of the skin to the drug has resulted in transient burning and hyperpigmentation of the affected areas.105 If the lyophilized material or solutions of the drug come in contact with the skin or mucous membrane, the affected area should be washed immediately and thoroughly with soap and water.105
Diluted solutions of carmustine should be inspected visually for particulate matter and discoloration prior to administration whenever solution and container permit.105
Carmustine wafers are implanted intracranially in the resection cavity following surgical resection of brain tumor.124
The manufacturer recommends that double surgical gloves be worn when handling intracranial carmustine wafers since exposure to carmustine can result in severe burning and hyperpigmentation of the skin.124 A surgical instrument dedicated to the handling of carmustine wafers should be used to implant the wafers.124 Any carmustine wafer or wafer remnant should be handled as a potentially cytotoxic material.124 The outside surface of the outer foil pouch containing the carmustine wafer is not sterile;124 the inner pouch is sterile, and the wafer should be placed into a designated sterile field following removal from the pouch.124
The usual IV dosage of carmustine used as a single agent in previously untreated patients is 150-200 mg/m2 administered every 6 weeks; this may be administered as a single dose or divided into daily injections such as 75-100 mg/m2 on 2 successive days.105 If carmustine is administered in combination with other myelosuppressive drugs or to patients with compromised bone marrow function, dosage must be reduced accordingly.105 Subsequent dosage must be determined by the clinical and hematologic response and tolerance of the patient in order to obtain optimum therapeutic results with minimum adverse effects. Clinicians should consult published protocols for the dosage of carmustine and other chemotherapeutic agents and the method and sequence of administration. Because of the delayed and cumulative myelosuppressive effects, carmustine usually is given at intervals of at least 6 weeks.105 However, repeat courses of carmustine should not be administered until leukocyte and platelet counts have returned to acceptable levels (usually 4000/ mm3 and 100,000/ mm3, respectively).105 In addition, an adequate number of neutrophils should be present on the peripheral blood smear.105 The manufacturer suggests that dosage subsequent to the initial course of therapy may be adjusted according to the schedule in the table that follows; however, some clinicians believe the manufacturer's recommendations could result in overdosage, and advocate dosage reductions of 25% when platelet nadirs are 50,000-74,999/ mm3, 50% for nadirs of 25,000-49,999/ mm3, and 75% for nadirs less than 25,000/ mm3.
Nadir After Prior Dose (cells/ mm3) | Percentage of Prior Dose to be Given | |
---|---|---|
Leukocytes | Platelets | |
>4000 | >100,000 | 100% |
3000-3999 | 75,000-99,999 | 100% |
2000-2999 | 25,000-74,999 | 70% |
<2000 | <25,000 | 50% |
Topical Use for Cutaneous T-cell Lymphoma
In the treatment of cutaneous T-cell lymphoma (mycosis fungoides), carmustine has been applied topically as an alcoholic solution or ointment; such dosage forms are not commercially available in the US.149,150 The drug is usually applied topically once daily.150 The usual topical dosage is 10 mg daily for 7-14 weeks (maximum: 17 weeks).149 If response is inadequate, a second course of topical therapy is administered after a rest interval of 6 weeks, using 20 mg daily for 4-8 weeks, as tolerated.149 Clinicians should consult specialized references for specific information and details on the preparation and use of topical carmustine for cutaneous T-cell lymphoma.149,150
The carmustine wafer is implanted at the initial surgery for newly diagnosed high-grade malignant glioma.124 The carmustine wafer also is implanted during surgery for recurrent glioblastoma multiforme.124
In clinical trials of patients undergoing surgery and intracranial implantation of carmustine wafers for recurrent malignant glioma, chemotherapy was withheld for at least 4 weeks (6 weeks for nitrosoureas) prior to and 2 weeks after surgery;124 external beam radiation therapy was administered no sooner than 3 weeks after surgery.151
Following the resection of either newly diagnosed high-grade malignant glioma or recurrent glioblastoma multiforme and confirmation of tumor pathology, when hemostasis is obtained, up to 8 carmustine wafers are placed intracranially to cover as much of the resection cavity as possible.124 The manufacturer recommends the implantation of 8 wafers (each wafer containing 7.7 mg of carmustine with 8 wafers resulting in a total dose of 61.6 mg carmustine).124 If the size and shape of the cavity will not allow placement of 8 wafers, the maximum possible number of wafers should be used.124 Because there is no clinical experience with higher doses, no more than 8 wafers should be placed intracranially per surgical procedure.124
The wafers should be placed intracranially to cover as much of the resection cavity as possible, and slight overlapping of the wafers is permissible.124 Wafers broken in half may be used, but wafers broken in more than 2 pieces should be discarded.124 Oxidized regenerated cellulose (Surgicel®) may be placed over the wafers to secure them against the surface of the resection cavity.124 Following placement of the wafers, the resection cavity should be irrigated and the dura closed in a watertight fashion to minimize the risk of CSF leak.124
Information on the adverse effects associated with use of the intracranial carmustine wafer is derived mainly from data for 240 patients with newly diagnosed malignant glioma or 222 patients with recurrent malignant glioma receiving either carmustine-wafer or placebo-wafer implants in 2 randomized trials.124 Unless otherwise specified, adverse effects occurred at a similar rate in patients receiving carmustine-wafer or placebo-wafer implants.124
Pulmonary toxicity, including acute or delayed onset of pulmonary fibrosis causing death, has occurred in patients receiving systemic carmustine therapy.105 Pulmonary toxicity characterized by pulmonary infiltrates and/or fibrosis occurring 9 days to 43 months following treatment has been reported in patients receiving carmustine or related nitrosoureas.105 Most reported cases of pulmonary toxicity have occurred in patients receiving prolonged carmustine therapy with total doses exceeding 1400 mg/m2; however, pulmonary fibrosis has occurred with lower total doses.105 Other risk factors include prior history of pulmonary disease and duration of carmustine therapy.105 Pulmonary toxicity occasionally has been rapidly progressive and/or fatal.105
In a study of 17 children (aged 1-16 years) receiving carmustine in cumulative doses ranging from 770-1800 mg/m2 combined with cranial radiation therapy for intracranial tumors, 8 children (47%) died of delayed pulmonary fibrosis, including all of those who received initial treatment at less than 5 years of age (5 children).105 Onset of pulmonary fibrosis has been observed up to 17 years following carmustine therapy.105 Clinical findings include pulmonary hypoplasia with upper zone contraction on chest radiographs, and an unusual pattern of upper zone fibrosis on thoracic CT scans; no abnormal findings were observed on gallium scans.105 Late onset of reduction in pulmonary function was observed in all long-term survivors in the study.105 Carmustine-induced pulmonary fibrosis may be slowly progressive and cause death.105
Pulmonary embolism occurred in 8% of patients receiving the intracranial carmustine wafer implant at initial surgery for malignant glioma.124 Pneumonia was reported in 8%, and dyspnea in 3%, of patients.124
Pulmonary embolism was reported in 4-9% of patients with recurrent glioma receiving the intracranial carmustine wafer implant in a randomized trial.151 Aspiration pneumonia has been reported in 1% of patients receiving the carmustine wafer implant in clinical studies to date.124
A serious and frequent adverse effect associated with systemic administration of carmustine is delayed hematologic toxicity, which is cumulative and usually occurs 4-6 weeks after administration of the drug.105 Thrombocytopenia is generally the most severe hematologic effect, appearing and subsiding earlier than other hematologic toxicities; however, both leukopenia and thrombocytopenia may be dose-limiting toxicities.105 Thrombocytopenia usually is evident at approximately 4 weeks and persists for about 1-2 weeks, and leukopenia usually is evident at approximately 5-6 weeks and persists for 1-2 weeks.105 Following repeated doses of carmustine, however, cumulative myelosuppression manifested as more depressed indices or as more prolonged suppression may occur.105 Anemia also occurs, but generally is less frequent and less severe than other hematologic toxicities.105
Anemia was reported in 4-9% of patients with recurrent glioma receiving the intracranial carmustine wafer implant in a randomized trial.151 Thrombocytopenia and leukocytosis each has been reported in 1% of patients receiving the carmustine wafer implant in clinical studies to date.124
Acute leukemia and bone marrow dysplasia have been reported in patients receiving long-term nitrosourea therapy,105,107,108 including some who received carmustine.107,108
Among patients receiving the intracranial carmustine wafer implant at initial surgery for malignant glioma, the most common adverse nervous system effects were hemiplegia (41%), seizures (33%), headache (28%), confusion (23%), brain edema (23%), and aphasia (18%).124 Depression was reported in 16% of patients, and somnolence and speech disorder each occurred in 11% of patients.124 Intracranial hypertension occurred more frequently in patients receiving the carmustine wafer implant (9%) than in those receiving the placebo wafer implant (2%).124,152 Other adverse nervous system effects included amnesia in 9%; personality disorder in 8%; anxiety, facial paralysis, or neuropathy, in 7%; ataxia, hypesthesia, paresthesia, or abnormal thinking, in 6%; and abnormal gait, dizziness, grand mal seizure, hallucinations, insomnia, or tremor, in 5% of patients.124 Brain abscess or meningitis was reported in 5% of patients receiving the carmustine wafer and 6% of patients receiving the placebo wafer.124 Coma occurred in 4%, incoordination in 3%, and hypokinesia in 2%, of patients receiving the carmustine wafer implant.124
In randomized trials of patients with newly diagnosed or recurrent malignant glioma, adverse nervous system effects which may have been related to treatment with the intracranial carmustine wafer implant included seizures and brain edema.124 Among patients with newly diagnosed malignant glioma, seizures or grand mal seizures respectively occurred in about 33 or 5% of patients receiving the carmustine wafer and 38 or 4% of those receiving the placebo wafer.124 Within the first 5 days following wafer implantation, the incidence of seizures was 2.5% in the carmustine wafer-treated group and 4.2% in the placebo wafer-treated group.124 The time of onset of the first postoperative seizure did not differ between the groups.124 Among patients with recurrent disease, the incidence of postoperative seizures was 19% in both the placebo wafer- and carmustine wafer-treated groups; however, the first new or worsened seizure occurred sooner after craniotomy and occurred more frequently during the first 5 postoperative days in patients treated with the carmustine implant.124 The median time to onset of the first new or worsened seizure was 3.5 days in patients receiving carmustine wafers compared with 61 days in those receiving placebo wafers.124 About 54% of patients receiving carmustine wafers experienced the first new or worsened seizure within the first 5 postoperative days compared with 9% of those receiving placebo wafers.124
Among patients with newly diagnosed malignant glioma, brain edema occurred in 22.5% of patients receiving the carmustine wafer and 19.2% of patients receiving the placebo wafer in a randomized trial.124 Brain edema was reported in 4% of patients with recurrent malignant glioma treated with intracranial carmustine wafer implants in a randomized trial.124 Cases of intracerebral mass effect unresponsive to corticosteroids, including one case leading to brain herniation, have been reported in patients treated with carmustine wafers.124 Reoperation, sometimes involving removal of carmustine wafers or wafer remnants, has been necessary in some patients developing brain edema with mass effect due to tumor recurrence, intracranial infection, or necrosis.124 Formation of tumor bed cyst that was unresponsive to treatment with high-dose corticosteroids and required reoperation for drainage also has been reported following implantation of carmustine wafers.136
Pain occurred in 13% of patients receiving the intracranial carmustine wafer implant for newly diagnosed malignant glioma.124 Pain was reported in a greater number (7 versus 1%) of patients receiving intracranial carmustine wafer implants for recurrent malignant glioma in a randomized trial compared with those receiving placebo wafer implants.124
Common adverse nervous system effects occurring in patients undergoing intracranial implantation of the carmustine wafer for recurrent malignant glioma in a randomized trial include convulsion or hemiplegia in 19%, headache in 15%, somnolence in 14%, confusion in 10%, aphasia in 9%, stupor in 6%, intracranial hypertension in 4%, and meningitis or abscess in 4%.124 Other adverse nervous system effects reported in less than 4% but at least 1% of patients receiving the carmustine wafer implant for recurrent malignant glioma in clinical studies to date include hydrocephalus, depression, abnormal thinking, ataxia, dizziness, insomnia, monoplegia, coma, amnesia, diplopia, and paranoid reaction.124 Cerebral hemorrhage and cerebral infarct each was reported in less than 1% of patients receiving the carmustine wafer implant.124 In a small randomized study of patients receiving carmustine-wafer or placebo-wafer implants for the initial treatment of high-grade glioma, hemiparesis occurred in 38% of patients receiving the carmustine wafer.134
Headache also has been reported in patients receiving IV carmustine.105 Other effects reported in conjunction with systemic administration of carmustine but not definitely attributable to the drug include dizziness, loss of equilibrium, and ataxia.
In a randomized trial, encephalopathy, sometimes fatal, was reported in patients receiving intra-arterial carmustine.132
Healing abnormalities, including wound dehiscence; delayed wound healing; subdural, subgaleal, or wound effusions; and CSF leaks, were reported in 16% of patients receiving the intracranial carmustine wafer implant versus 12% of patients receiving the placebo wafer at initial surgery for malignant glioma.124 CSF leaks occurred in 5% of patients receiving the carmustine wafer versus 0.8% of those receiving the placebo wafer.124 Healing abnormalities occurred in 14% of patients receiving the intracranial carmustine wafer implant compared with 5% of those receiving the placebo wafer implant for recurrent malignant glioma in a randomized trial.124
Local burning pain at the site of injection or along the course of the vein and venospasm occur commonly in patients receiving IV carmustine, but true thrombosis and thrombophlebitis occur rarely.38,105 Rapid IV infusion of carmustine may produce intensive flushing of the skin and suffusion of the conjunctiva; these effects occur within 2 hours and may persist for 4 hours after administration of carmustine.105
Infection occurred in 18% of patients receiving the carmustine wafer and 20% of those receiving the placebo wafer for newly diagnosed malignant glioma in a randomized trial.124 Intracranial infection (abscess or meningitis) was reported in 5% of patients receiving the carmustine wafer and 6% of patients receiving the placebo wafer for newly diagnosed malignant glioma in a randomized trial.124 Intracranial infection (meningitis or abscess) occurred in 4% of patients receiving the intracranial carmustine wafer implant compared with 1% of those receiving the placebo wafer implant for recurrent malignant glioma in a randomized trial.124 Among patients with recurrent disease receiving the carmustine wafer implant, there were 2 cases of bacterial meningitis, one case of chemical meningitis, and one case of meningitis of unspecified type; a brain abscess was reported in a patient receiving the placebo wafer.151 Deep wound infection (i.e., infection of subgaleal space, bone, meninges, or neural parenchyma) was reported in 6% of patients receiving the carmustine-wafer or placebo-wafer implant for recurrent disease.151
Pneumonia occurred in 8% of patients receiving the carmustine wafer for newly diagnosed malignant glioma.124 Pneumonia and oral candidiasis (moniliasis) were reported in 4-9% of patients with recurrent glioma receiving the intracranial carmustine wafer implant in a randomized trial; these adverse effects were reported at a similar rate in patients receiving the placebo wafer implant.151 Sepsis has been reported in 1% of patients receiving the carmustine wafer implant in clinical studies to date.124
Nausea and vomiting occur frequently after IV administration of carmustine.105 These effects are dose related and generally occur within a few minutes to 2 hours and persist up to 6 hours after administration of the drug.105 Prior administration of antiemetics may diminish or prevent these effects.105 Other less frequently reported adverse effects of carmustine include diarrhea, esophagitis, anorexia, and dysphagia.
The most common adverse GI effects reported in patients receiving the intracranial carmustine wafer implant at initial surgery for malignant glioma were nausea (22%), vomiting (21%), and constipation (19%); abdominal pain occurred in 8%, and diarrhea occurred in 5%, of patients receiving the carmustine wafer.124 Nausea and vomiting occurred in 8% of patients with recurrent glioma receiving the intracranial carmustine wafer implant in a randomized trial.124 Other adverse GI effects reported in 1-2% of patients receiving the carmustine wafer implant in clinical studies to date include diarrhea, constipation, dysphagia, GI hemorrhage, and fecal incontinence.124
Hepatotoxicity, reported in up to 26% of patients who receive carmustine, is generally mild and reversible, usually occurring when high doses of the drug are administered. Hepatotoxicity may be manifested by increases in serum transaminase, alkaline phosphatase, and bilirubin concentrations;105 jaundice, portal system encephalopathy, and at least 1 death have also been reported.
Abnormal results of liver function tests were reported in 1% of patients receiving the intracranial carmustine wafer implant at initial surgery for malignant glioma.124
A decrease in kidney size, progressive azotemia, and renal failure have occurred in patients who received large cumulative doses after prolonged therapy with carmustine and related nitrosoureas; renal damage has also occurred occasionally in patients receiving lower total doses.105
Hyponatremia was reported in 4-9% of patients with recurrent glioma receiving the intracranial carmustine wafer implant in a randomized trial.151 Hypokalemia has been reported in 1% of patients receiving the carmustine wafer implant in clinical studies to date.124
The manufacturer reports that ocular toxicity, including neuroretinitis, has occurred in patients receiving carmustine therapy, particularly when the drug was injected intra-arterially into the carotid artery.105 In a randomized trial, severe ocular toxicity, including progression of visual loss to blindness, was reported in patients receiving intra-arterial carmustine.132
Among patients receiving the intracranial carmustine wafer implant at initial surgery for malignant glioma, adverse ocular effects included conjunctival edema (7%), abnormal vision (6%), visual field defect (5%), unspecified eye disorder (3%), and diplopia (1%).124
Visual field defect and ocular pain have been reported in 2 and 1%, respectively, of patients receiving the intracranial carmustine wafer implant in clinical studies to date.124 Conjunctivitis was reported at a similar rate in patients with recurrent glioma receiving either carmustine-wafer or placebo-wafer implants in a randomized trial.133
Chest pain,105 hypotension,105 and tachycardia105 have been reported in patients receiving IV carmustine.
Among patients receiving the intracranial carmustine wafer implant at initial surgery for malignant glioma, deep thrombophlebitis occurred in 10%, peripheral edema in 9%, and hemorrhage in 7% of patients.124 Chest pain occurred in 5% of patients receiving the carmustine wafer implant and none of the patients receiving the placebo wafer implant.124
Deep thrombophlebitis was reported in 4-9% of patients with recurrent glioma receiving the intracranial carmustine wafer implant in a randomized trial.151 Hypertension, edema, chest pain, and hypotension have been reported in 3, 2, 1, and 1%, respectively, of patients receiving the carmustine wafer implant in clinical studies to date.124
Among patients receiving the intracranial carmustine wafer implant at initial surgery for malignant glioma, urinary tract infection and urinary incontinence each occurred in 8% of patients.124
Urinary tract infection was reported in about 20% of patients with recurrent glioma receiving either the intracranial carmustine-wafer or placebo-wafer implant in a randomized trial.124 Urinary incontinence has been reported in 2% of patients receiving the carmustine wafer implant in clinical studies to date.124
Endocrine and Metabolic Effects
Diabetes mellitus occurred in 5%, and Cushing's syndrome occurred in 3%, of patients receiving the intracranial carmustine wafer implant at initial surgery for malignant glioma.124 Hyperglycemia has been reported in 3% of patients receiving the intracranial carmustine wafer implant in clinical studies to date.124
Accidental contact of the skin with reconstituted solutions of carmustine has produced transient hyperpigmentation of the affected area.105 Adverse dermatologic effects resulting from topical application of carmustine for the treatment of mycosis fungoides include moderate to severe dermatitis, tiny petechiae simulating hemosiderosis, hyperpigmentation, and telangiectasia.
Rash has been reported in 12% of patients with newly diagnosed malignant glioma and 5% of patients with recurrent glioma receiving the intracranial carmustine wafer implant in randomized trials.124 Alopecia occurred in 10% of patients receiving the intracranial carmustine wafer implant at initial surgery for malignant glioma.124
Back pain was reported in 7%, and myasthenia in 4%, of patients receiving the intracranial carmustine wafer at initial surgery for malignant glioma.124 Neck pain and back pain have been reported in 2 and 1%, respectively, of patients receiving the intracranial carmustine wafer implant in clinical studies to date.124
The most common adverse effect reported in patients receiving the intracranial carmustine wafer implant at initial surgery for malignant glioma in a randomized trial was aggravation reaction, which occurred in 82% of patients.124 As defined by COSTART, aggravation reaction refers to events involving progression of tumor or disease or general deterioration in condition, such as performance status or neurologic status.124 Asthenia or fever occurred in 22 or 18%, respectively, of patients receiving the carmustine wafer at initial surgery.124 Facial edema was reported in 6% of patients receiving the carmustine wafer.124 Other adverse effects occurring in patients receiving the intracranial carmustine wafer at initial surgery for malignant glioma include accidental injury (5%) and allergic reaction (2%).124
Fever has been reported in 12% of patients with recurrent glioma receiving the intracranial carmustine wafer implant in a randomized trial.124 Accidental injury and allergic reaction each has been reported in 1% of patients receiving the carmustine wafer implant in clinical studies to date.124 Allergic reaction also has been reported in patients receiving IV carmustine.105
Precautions and Contraindications
Carmustine is a highly toxic drug with a low therapeutic index, and a therapeutic response is not likely to occur without some evidence of toxicity. The drug must be used only under constant supervision by clinicians experienced in cancer chemotherapy.
Patients who receive myelosuppressive drugs experience an increased frequency of infections as well as possible hemorrhagic complications. Because these complications have caused fatalities, the patient should be instructed to notify the clinician if fever, sore throat, or unusual bleeding or bruising occurs. The patient's hematologic status must be carefully monitored, and blood counts performed weekly during and for at least 6 weeks after carmustine therapy. Carmustine should be administered with caution to patients with depressed platelet, leukocyte, or erythrocyte counts.
Pulmonary toxicity associated with carmustine therapy appears to be dose related, and patients receiving cumulative doses exceeding 1400 mg/m2 are at substantially higher risk than patients receiving lower cumulative doses of the drug. Pulmonary function tests should be performed prior to initiation of and frequently during carmustine therapy. Patients with a baseline forced vital capacity (FVC) or pulmonary diffusion capacity for carbon monoxide (DLCO) that is less than 70% of the predicted value are particularly at risk for carmustine-induced pulmonary toxicity.105 In addition, delayed onset of pulmonary fibrosis, frequently fatal, has occurred up to 17 years following carmustine therapy in children and adolescents.105 (See Cautions: Pulmonary Effects.)
Because carmustine may cause hepatic dysfunction, liver function should be monitored periodically in patients receiving the drug.105 The manufacturer recommends that renal function tests also be monitored periodically during carmustine therapy.105
Carmustine is contraindicated in patients who have demonstrated previous hypersensitivity to the drug.105
Patients undergoing craniotomy for malignant glioma and intracranial implantation of carmustine wafers should be monitored closely for potential complications of craniotomy, including seizures, intracranial infections, abnormal wound healing, and brain edema.124 Reoperation, sometimes involving removal of carmustine wafers or wafer remnants, may be necessary in patients developing brain edema with mass effect due to tumor recurrence, intracranial infection, or necrosis.124 Communication between the surgical resection cavity and the ventricular system could result in migration of carmustine wafers into the ventricular system, leading to obstructive hydrocephalus; any such communication larger than the diameter of a carmustine wafer should be closed prior to implantation of carmustine wafers.124
Imaging studies of the head (e.g., CT scan, MRI) may reveal enhancement in the brain tissue surrounding the resection cavity following intracranial implantation of carmustine wafers; this enhancement may represent edema and inflammation caused by carmustine wafer or tumor progression.124
The use of carmustine wafers is contraindicated in patients who have demonstrated a previous hypersensitivity to carmustine or any component of the implants.124
Safety and efficacy of carmustine in children have not been established.105 Fatal pulmonary fibrosis with delayed onset up to 17 years following treatment has been reported in children receiving IV carmustine therapy for brain tumors.105 Because of the extremely high risk of pulmonary toxicity frequently causing death, particularly in children younger than 5 years of age, the risks and benefits of carmustine therapy must be weighed carefully.105 (See Cautions: Pulmonary Effects.)
Safety and efficacy of intracranial carmustine wafer implants in children have not been established.105
Safety and efficacy of carmustine in geriatric patients have not been studied specifically to date.105,148 While clinical experience has not revealed age-related differences in response, drug dosage generally should be titrated carefully in geriatric patients, usually initiating therapy at the low end of the dosage range.105,148 The greater frequency of decreased hepatic, renal, and/or cardiac function and of concomitant disease and drug therapy observed in the elderly also should be considered.105,148
Because geriatric patients may have decreased renal function and because patients with renal impairment may be at increased risk of carmustine-induced toxicity, drug dosage should be titrated carefully and renal function should be monitored closely in patients in this age group.105,148
Mutagenicity and Carcinogenicity
In vitro tests have shown carmustine to be mutagenic, and both in vitro and in vivo tests have shown the drug to be clastogenic.124 Carmustine has been shown to be carcinogenic in rats and mice, and has been associated with a marked increase in the incidence of tumors, particularly subcutaneous and lung neoplasms, when administered to these animals in dosages approximately equivalent to or less than the usual human dosage.105,124 Nitrosourea therapy, such as carmustine therapy, has carcinogenic potential in humans, and the manufacturer states that acute leukemia and bone marrow dysplasia have been reported in patients receiving long-term therapy with nitrosourea derivatives.105
No mutagenicity or carcinogenicity studies have been performed with the carmustine wafer.124
Pregnancy, Fertility, and Lactation
Carmustine may cause fetal harm when administered to pregnant women, but potential benefits from use of the drug may be acceptable in certain conditions despite possible risks to the fetus.105,113 Carmustine has been shown to be teratogenic in rats and embryotoxic in rats and rabbits receiving dosages approximating the usual human dosage.105,124 There are no adequate and controlled studies to date using carmustine or carmustine wafer implants in pregnant women,105,124 and the drug should be used during pregnancy only in life-threatening situations or severe disease for which safer drugs cannot be used or are ineffective.113 Women of childbearing potential should be advised to avoid becoming pregnant during carmustine therapy.105 When the drug is administered during pregnancy or if the patient becomes pregnant while receiving the drug, the patient should be informed of the potential hazard to the fetus.105,124
Carmustine has been shown to affect fertility in male rats receiving the drug at dosages somewhat higher than the usual human dosage.105
It is not known whether carmustine is distributed in milk.105 Because of the potential for serious adverse reactions to carmustine in nursing infants, nursing should be discontinued during systemic carmustine therapy.105
It is not known whether components of the carmustine wafer, including carmustine, carboxyphenoxypropane, or sebacic acid, are distributed in milk.124 Because of the potential for serious adverse reactions to carmustine in nursing infants, nursing should be discontinued in patients receiving carmustine wafer implants.124
Interactions of carmustine wafers with other drugs have not been formally studied.124 In clinical trials of patients undergoing surgery and intracranial implantation of carmustine wafers for recurrent malignant glioma, chemotherapy was withheld for at least 4 weeks (6 weeks for nitrosoureas) prior to and 2 weeks after surgery.124 The short-term or long-term toxicity of carmustine wafers used in conjunction with systemic chemotherapy has not been fully evaluated.124
Cimetidine may potentiate the myelosuppressive effects (e.g., neutropenia, agranulocytosis) of myelosuppressive drugs (e.g., alkylating agents, antimetabolites) or therapies (e.g., radiation). Concomitant cimetidine therapy has been reported to potentiate the neutropenic and thrombocytopenic effect of carmustine alone or combined with radiation therapy.
Qualitative and quantitative changes in tear films leading to damage of the corneal and conjunctival epithelium have been reported in patients receiving high doses of carmustine and mitomycin.146
In patients receiving carmustine and phenytoin, serum concentrations of phenytoin may be decreased.147 In patients receiving carmustine therapy, serum concentrations of phenytoin should be monitored carefully and dosage adjustments made as necessary.147
In clinical trials of patients undergoing surgery and intracranial implantation of carmustine wafers for recurrent malignant glioma, external beam radiation therapy was initiated no sooner than 3 weeks after surgery.151 In a randomized trial of patients receiving either carmustine wafers or placebo wafers at initial surgery for malignant glioma, most patients received a standard course of radiation therapy (55-60 Gy) initiated at 3 weeks following surgery.124 No short-term or long-term toxicity is known to result from the use of radiation therapy in conjunction with intracranial carmustine wafers.124
There are no known antidotes for overdosage of IV carmustine.105
There is no clinical experience with intracranial implantation of more than 8 carmustine wafers per surgical procedure.124
Although carmustine is believed to act by alkylation of DNA and RNA,105,124 the mechanism of action has not been completely elucidated and other effects such as carbamoylation105 and modification of cellular proteins may be involved. The overall result is thought to be the inhibition of both DNA and RNA synthesis. The manufacturer reports that cross-resistance between carmustine and lomustine has occurred.
The cytotoxic effect of the carmustine wafer is dependent on release of the drug to the tumor cavity in sufficient amounts to achieve concentrations that result in tumoricidal activity.124
The absorption of the copolymer contained in carmustine wafers has not been evaluated in humans.124 Plasma concentrations of carmustine following intracranial implantation of the wafers have not been determined in humans, but in rabbits undergoing surgical implantation of wafers containing 3.85% carmustine, no detectable levels of carmustine were observed in plasma.124
Following IV infusion of carmustine, the steady-state volume of distribution averaged 3.25 L/kg.124 Because of their high lipid solubility, carmustine and/or its metabolites readily cross the blood-brain barrier.105 Substantial CSF concentrations occur almost immediately after IV administration of carmustine, and CSF concentrations of radioactivity have been variously reported to range from 15-70% of concurrent plasma concentrations. Carmustine metabolites are distributed into milk, but in concentrations less than those in maternal plasma.
The carmustine implant is designed to deliver carmustine directly into the surgical cavity created upon resection of a brain tumor.124 The concentration of carmustine achieved in human brain tissue with intracranial implantation of the carmustine wafers has not been determined.124 The distribution of the copolymer contained in carmustine wafers has not been evaluated in humans.124 In rabbits undergoing surgical implantation of wafers containing 3.85% carmustine, no detectable concentrations of carmustine were observed in CSF.124
After IV administration, carmustine is rapidly cleared from the plasma with no intact drug detectable after 15 minutes.105 In studies using 14C-labeled carmustine, prolonged concentrations of radioactivity were present in the plasma and tissue,105 probably as a result of protein binding and/or enterohepatic circulation of metabolites, and may be responsible for the delayed hematologic toxicity of carmustine. (See Cautions.) Following IV infusion of carmustine, the terminal plasma elimination half-life and clearance averaged 22 minutes and 56 mL/minute per kg, respectively.124
When the carmustine wafer is exposed to the aqueous environment of the resection cavity, hydrolysis of the anhydride bonds in the copolymer occurs, resulting in the release of carmustine and two monomers, carboxyphenoxypropane, and sebacic acid.124 The carmustine contained in the wafer diffuses into the surrounding brain tissue.124 The metabolism and excretion of the copolymer contained in carmustine wafers has not been evaluated in humans.124 Animal studies have shown that more than 70% of the copolymer degrades within 3 weeks following implantation of carmustine wafers into brain tissue;124 following hydrolysis of the copolymer, carboxyphenoxypropane is eliminated renally, while sebacic acid (an endogenous fatty acid) is metabolized in the liver and expired as carbon dioxide.124 In humans, wafer remnants have been observed on brain imaging scans or located during subsequent surgical procedures up to 8 months following intracranial implantation.124 Wafer remnants retrieved from 2 patients approximately 2-3 months after implantation were analyzed and found to consist mostly of water and monomeric components with minimal detectable amounts of carmustine.124
Carmustine is rapidly metabolized.105 The antineoplastic and toxic effects of carmustine are thought to be caused by active metabolites.105 One metabolite, an alkylating moiety thought to be chloroethyl carbonium ion, leads to the formation of DNA cross-links.124 The metabolites are slowly excreted in urine. Following IV administration of 14C-labeled carmustine, about 30% of the radioactivity is excreted within 24 hours38 and about 60-70% of the radioactivity is excreted within 96 hours.105,124 Some enterohepatic circulation is believed to occur. Less than 1% of the radioactivity is excreted in feces. About 6-10% of the radioactivity is excreted as respiratory carbon dioxide, and the fate of the remaining 20-30% of a dose has not been determined.105,124
Carmustine, a nitrosourea derivative, is generally considered to be an alkylating agent. The drug occurs as lyophilized yellow flakes or congealed mass.105 Carmustine is slightly soluble in water, freely soluble in alcohol, and highly soluble in lipids.
Carmustine also is commercially available as a component of a biodegradable polymer implant.124 The intracranial carmustine implant is a sterile, off-white to pale yellow wafer measuring approximately 1.45 cm in diameter and 1 mm in thickness that contains 7.7 mg of carmustine and 192.3 mg of polifeprosan 20.124 Carmustine is homogeneously distributed in the matrix of polifeprosan 20, a biodegradable polyanhydride copolymer consisting of poly[bis( p -carboxyphenoxy)propane and sebacic acid in a 20:80 molar ratio that is used to control the local delivery of carmustine.124
Carmustine lyophilized material decomposes to an oily liquid at temperatures of 30.5°C or warmer; the liquefied material appears as an oily film on vials of the drug. Vials of the lyophilized material that may not have been refrigerated adequately should be inspected to determine whether any decomposition has occurred by holding them up to a bright light.105 If the drug appears as dry flakes or as a dry, congealed mass, the vial of carmustine is suitable for use and should be refrigerated immediately if it is to be stored for later reconstitution.105 If an oily film is present inside the vial, the drug should be discarded.105 Carmustine powder must be refrigerated at 2-8°C.105 Both the powder and its solutions must be protected from light.105 When stored according to manufacturer recommendations, the contents of unopened vials of the drug remain stable for up to 3 years.105
Following reconstitution of the lyophilized material with dehydrated (absolute) alcohol and sterile water for injection as directed by the manufacturer, solutions of carmustine are clear and colorless to yellow in color.105 These solutions are stable in glass containers for 8 hours at 25°C when protected from light.101,105 Reconstituted carmustine solutions that are further diluted with 5% dextrose injection to a final concentration of 0.2 mg/mL in glass containers are stable for 8 hours when stored at 25°C and protected from light;105 however, carmustine powder contains no preservatives and the possibility of microbial contamination of reconstituted solutions must be considered. Some data indicate that solutions of carmustine in 5% dextrose injection in PVC containers are rapidly degraded; admixture of the drug in PVC containers should therefore be avoided.102 The manufacturer recommends that only glass containers be used for the administration of solutions of the drug.105 Carmustine is rapidly degraded in aqueous solutions at pH greater than 6,103 and it is recommended that the drug not be admixed with nor administered through a common tubing or site with solutions containing sodium bicarbonate.104
Carmustine wafers must be stored at a temperature of -20°C or less.124 The aluminum foil laminate pouches containing each carmustine wafer should not be opened until immediately prior to the implantation procedure.124 Unopened foil pouches may be kept at ambient room temperature for up to 6 hours at a time.124
Additional Information
For further information on the handling of antineoplastic agents, see the ASHP Guidelines on Handling Hazardous Drugs at [Web].
Excipients in commercially available drug preparations may have clinically important effects in some individuals; consult specific product labeling for details.
Please refer to the ASHP Drug Shortages Resource Center for information on shortages of one or more of these preparations.
Routes | Dosage Forms | Strengths | Brand Names | Manufacturer |
---|---|---|---|---|
Parenteral | For injection, for IV infusion | 100 mg | ||
Local | Implants | 7.7 mg (of carmustine per wafer) | Gliadel® Wafer |
Only references cited for selected revisions after 1984 are available electronically.
38. Kleinman LM et al. Investigational drug information. Drug Intell Clin Pharm . 1976; 10:48-9.
100. Zackheim HS, Epstein EH Jr, McNutt NS et al. Topical carmustine (BCNU) for mycosis fungoides and related disorders. J Am Acad Dermatol . 1983; 9:363-74. [PubMed 6630597]
101. Lauper RD (Bristol-Myers Oncology, Syracuse, NY): Personal communication; 1985 Nov.
102. Benvenuto JA, Anderson RW, Kerkov K et al. Stability and compatibility of antitumor agents in glass and plastic containers. Am J Hosp Pharm . 1981; 38: 1914-8.
103. Laskar PA, Ayres JW. Degradation of carmustine in aqueous media. J Pharm Sci . 1977; 66:1073-6. [PubMed 19612]
104. Colvin M, Hartner J, Summerfield M. Stability of carmustine in the presence of sodium bicarbonate. Am J Hosp Pharm . 1980; 37:677-8. [PubMed 7386477]
105. Bristol-Myers Squibb. BiCNU® (carmustine for injection) prescribing information. Princeton, NJ; 2003 Oct.
106. Patten GA, Billi JE, Rotman HH. Rapidly progressive, fatal pulmonary fibrosis induced by carmustine. JAMA . 1980; 244:687-8. [PubMed 7392168]
107. Johnson R, Douglas J, Corey L et al. Carmustine and myelodysplasia. Ann Intern Med . 1985; 103(6 Part 1):964-5.
108. Greene MH, Boice JD Jr, Strike TA. Carmustine as a cause of acute nonlymphocytic leukemia. N Engl J Med . 1985; 313:579. [PubMed 3894973]
109. Durant JR, Norgard MJ, Murad TM et al. Pulmonary toxicity associated with bischloroethylnitrosourea (BCNU). Ann Intern Med . 1979; 90:191-4. [PubMed 443651]
110. Weiss RB, Muggia FM. Pulmonary effects of carmustine (bischloroethylnitrosourea, BCNU). Ann Intern Med . 1979; 91:131-2. [PubMed 464441]
111. Sweet DL. Pulmonary effects of carmustine (bischloroethylnitrosourea, BCNU). Ann Intern Med . 1979; 91:132. [PubMed 464443]
112. Durant JR. Pulmonary effects of carmustine (bischloroethylnitrosourea, BCNU). Ann Intern Med . 1979; 91:132-3.
113. Food and Drug Administration. Prescription drug advertising; content and format for labeling of human prescription drugs. Fed Regist . 1979; 44:37434-67.
114. Melanoma. From: CancerNet/PDQ. Physician data query (database). Bethesda, MD: National Cancer Institute; 2001 Jun.
115. Duran Garcia E, Santolaya R, Requena T. Treatment of malignant melanoma. Ann Pharmacother . 1999; 33:730-8. [PubMed 10410188]
116. Cohen GL, Falkson CI. Current treatment options for malignant melanoma. Drugs . 1998; 55:791-9. [PubMed 9617594]
117. Rosenberg SA, Yang JC, Schwartzentruber DJ et al. Prospective randomized trial of the treatment of patients with metastatic melanoma using chemotherapy with cisplatin, dacarbazine, and tamoxifen alone or in combination with interleukin-2 and interferon alfa-2b. J Clin Oncol . 1999; 17:968-75. [PubMed 10071291]
118. Falkson CI, Ibrahim J, Kirkwood JM et al. Phase III trial of dacarbazine versus dacarbazine with interferon alpha-2b versus dacarbazine with tamoxifen versus dacarbazine with interferon alpha-2b and tamoxifen in patients with metastatic malignant melanoma: an Eastern Cooperative Oncology Group study. J Clin Oncol . 1998; 16:1743-51. [PubMed 9586887]
119. Margolin KA, Liu PY, Flaherty LE et al. Phase II study of carmustine, dacarbazine, cisplatin, and tamoxifen in advanced melanoma: a Southwest Oncology Group study. J Clin Oncol . 1998; 16:664-9. [PubMed 9469356]
120. Anon. Drugs of choice for cancer. Treat Guidel Med Lett . 2003; 1:41-52. [PubMed 15529105]
121. Chapman PB, Einhorn LH, Meyers ML et al. Phase III multicenter randomized trial of the Dartmouth regimen versus dacarbazine in patients with metastatic melanoma. J Clin Oncol . 1999; 17:2745-51. [PubMed 10561349]
122. Reviewers' comments (personal observations) on melanoma.
123. Mycosis fungoides and the Sezary syndrome. From: CancerNet/PDQ. Physician data query (database). Bethesda, MD: National Cancer Institute; 2001 Sep.
124. Guilford Pharmaceuticals Inc. Gliadel® wafer (polifeprosan 20 with carmustine implant) prescribing information. Baltimore, MD; 2003 Feb.
125. DeAngelis LM. Brain tumors. N Engl J Med . 2001; 344:114-23. [PubMed 11150363]
126. Walker MD, Alexander E Jr, Hunt WE et al. Evaluation of BCNU and/or radiotherapy in the treatment of anaplastic gliomas. A cooperative clinical trial. J Neurosurg . 1978; 49:333-43. [PubMed 355604]
127. Walker MD, Green SB, Byar DP et al. Randomized comparisons of radiotherapy and nitrosoureas for the treatment of malignant glioma after surgery. N Engl J Med . 1980; 303:1323-9. [PubMed 7001230]
128. Cairncross JG, Macdonald DR. Successful chemotherapy for recurrent malignant oligodendroglioma. Ann Neurol . 1988; 23:360-4. [PubMed 3382171]
129. Galanis E, Buckner J. Chemotherapy for high-grade gliomas. Br J Cancer . 2000; 82:1371-80. [PubMedCentral][PubMed 10780513]
130. Fine HA, Dear KB, Loeffler JS et al. Meta-analysis of radiation therapy with and without adjuvant chemotherapy for malignant gliomas in adults. Cancer . 1993; 71:2585-97. [PubMed 8453582]
131. Medical Research Council. Randomized trial of procarbazine, lomustine, and vincristine in the adjuvant treatment of high-grade astrocytoma: a Medical Research Council trial. J Clin Oncol . 2001; 19:509-18. [PubMed 11208845]
132. Shapiro WR. Chemotherapy of malignant gliomas: studies of the BTCG. Rev Neurol (Paris) . 1992; 148:428-34. [PubMed 1448662]
133. Brem H, Piantadosi S, Burger PC et al. Placebo-controlled trial of safety and efficacy of intraoperative controlled delivery by biodegradable polymers of chemotherapy for recurrent gliomas. Lancet . 1995; 345:1008-12. [PubMed 7723496]
134. Valtonen S, Timonen U, Toivanen P et al. Interstitial chemotherapy with carmustine-loaded polymers for high-grade gliomas: a randomized double-blind study. Neurosurgery . 1997; 41:44-8; discussion 48-9. [PubMed 9218294]
135. Adult brain tumors. From PDQ. Physician data query (database). Bethesda, MD: National Cancer Institute; 2005 Mar 24.
136. Engelhard HH. Tumor bed cyst formation after BCNU wafer implantation: report of two cases. Surg Neurol . 2000; 53:220-4. [PubMed 10773252]
137. Anon. Gliadel wafers for treatment of brain tumors. Med Lett Drugs Ther . 1998; 40:92. [PubMed 9760950]
138. Multiple myeloma and other plasma cell neoplasms. From: CancerNet/PDQ. Physician data query (database). Bethesda, MD: National Cancer Institute; 2001 Oct.
139. Myeloma Trialists' Collaborative Group. Combination chemotherapy versus melphalan plus prednisone as treatment for multiple myeloma: an overview of 6,633 patients from 27 randomized trials. J Clin Oncol . 1998; 16:3832-42. [PubMed 9850028]
140. Mineur P, Menard JF, Le Loet X et al. VAD or VMBCP in multiple myeloma refractory to or relapsing after cyclophosphamide-prednisone therapy (protocol MY 85). Br J Haematol. 1998;103:512-7. [PubMed 9827927]
141. Adult Hodgkin's disease. From: CancerNet/PDQ. Physician data query (database). Bethesda, MD: National Cancer Institute; 2001 Sep.
142. Chopra R, McMillan AK, Linch DC et al. The place of high-dose BEAM therapy and autologous bone marrow transplantation in poor-risk Hodgkin's disease. A single-center eight-year study of 155 patients. Blood . 1993; 81:1137-45. [PubMed 8443375]
143. Nademanee A, O'apos;Donnell MR, Snyder DS et al. High-dose chemotherapy with or without total body irradiation followed by autologous bone marrow and/or peripheral blood stem cell transplantation for patients with relapsed and refractory Hodgkin's disease: results in 85 patients with analysis of prognostic factors. Blood . 1995; 85:1381-90. [PubMed 7858268]
144. Adult non-Hodgkin's lymphoma. From: CancerNet/PDQ. Physician data query (database). Bethesda, MD: National Cancer Institute; 2001 Oct.
145. Mills W, Chopra R, McMillan A et al. BEAM chemotherapy and autologous bone marrow transplantation for patients with relapsed or refractory non-Hodgkin's lymphoma. J Clin Oncol . 1995; 13:588-95. [PubMed 7884420]
146. Cruciani F, Tamanti N, Abdolrahimzadeh S et al. Ocular toxicity of systemic chemotherapy with megadoses of carmustine and mitomycin. Ann Ophthalmol . 1994; 26:97-100. [PubMed 7944163]
147. Grossman SA, Sheidler VR, Gilbert MR. Decreased phenytoin levels in patients receiving chemotherapy. Am J Med . 1989; 87:505-10. [PubMed 2683764]
148. Food and Drug Administration. MedWatchSafety-related drug labeling changes: BiCNU (carmustine) [February 2003]. From FDA web site [Web].
149. Zackheim HS, Epstein EH Jr, Crain WR. Topical carmustine (BCNU) for cutaneous T cell lymphoma: a 15-year experience in 143 patients. J Am Acad Dermatol . 1990; 22:802-10. [PubMed 2347966]
150. Zackheim HS. Topical carmustine (BCNU) in the treatment of mycosis fungoides. Dermatol Ther . 2003; 16:299-302. [PubMed 14686972]
151. Guilford Pharmaceuticals Inc. Gliadel® wafer (polifeprosan 20 with carmustine implant) prescribing information. Baltimore, MD; 2000 Aug.
152. Westphal M, Hilt DC, Bortey E et al. A phase 3 trial of local chemotherapy with biodegradable carmustine (BCNU) wafers (Gliadel wafers) in patients with primary malignant glioma. Neuro-oncol . 2003; 5:79-88. [PubMedCentral][PubMed 12672279]