VA Class:IM900
REMS: FDA approved a REMS for thalidomide to ensure that the benefits outweigh the risks. The REMS may apply to one or more preparations of thalidomide and consists of the following: elements to assure safe use and implementation system. See the FDA REMS page ([Web]). |
Thalidomide, a synthetic glutamic acid derivative, is an immunomodulatory agent with anti-inflammatory, antiangiogenic, and sedative and hypnotic activity; the drug is also a potent teratogen.1,2,112,220
Thalidomide is labeled by the US Food and Drug Administration (FDA) for acute treatment of cutaneous manifestations of moderate to severe erythema nodosum leprosum (ENL) in leprosy patients and for maintenance therapy for prevention and suppression of cutaneous manifestations of ENL recurrence.1 The drug has been designated an orphan drug by FDA for the treatment of ENL and treatment and maintenance of reactional lepromatous leprosy.28 Thalidomide also is used for the treatment of multiple myeloma and is designated an orphan drug by FDA for use in this condition.1,28 Thalidomide also has been designated an orphan drug by FDA for treatment of wasting syndrome associated with human immunodeficiency virus (HIV) infection;28 prevention and treatment of severe recurrent aphthous stomatitis in severely, terminally immunocompromised patients;28 prevention and treatment of graft-versus-host disease in patients receiving bone marrow transplantation;28 treatment of clinical manifestations of mycobacterial infection caused by Mycobacterium tuberculosis and nontuberculous mycobacteria;28 treatment of Crohn disease;28 treatment of primary brain tumors;28 and hereditary hemorrhagic telangiectasia.28,264 In addition, thalidomide has been used for the treatment of a variety of inflammatory and/or dermatologic disorders,49,59,60,77,93,94,104,110,117,118,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,180,181,182,183,184 treatment of various HIV-associated conditions,12,30,40,41,50,55,56,104,112,140,141,142,143,144,146,147,170 and treatment of various malignancies.29,32,100,161,162,163,168,169,173,174,175 Use of thalidomide may not limit disease progression and/or death.1
Past and Current Therapeutic Perspective
In the late 1950s, thalidomide was marketed in several other countries, including the United Kingdom and Canada, and was available over-the-counter in some countries (e.g., Germany) for use as a sedative and for the treatment of various conditions, including asthma, hypertension, migraine, and common symptoms of early pregnancy (e.g., morning sickness), often in combination with other drugs.2,3,4,5,15,52,53,54 Thalidomide was being used in clinical studies in the US,4,15,16 but did not receive FDA approval at that time principally because of European reports of thalidomide-associated peripheral neuropathy that may be irreversible and unanswered questions about potential adverse effects on fetuses.2,4,5,15,16,52,54,58 Because of thalidomide's prompt sedative effects, lack of hangover, and apparent safety (in regards to overdosage), the drug was considered to be a safe alternative to barbiturates,2,4,5,15,16,52 and its manufacturer at that time promoted it as a nontoxic drug with no adverse effects, and completely safe for administration to pregnant women.16 However, reports of serious adverse effects soon appeared1,2,3,4,5,15,16,52,53,54 and use of thalidomide was linked to an epidemic of severe and often fatal fetal and neonatal malformations (most frequently phocomelia) that affected children in 46 countries.2,3,4,5,15,16,52,53,54 During that time, 17 babies were born in the US with thalidomide-associated phocomelia to mothers who received the drug from overseas sources or received premarketing samples distributed by drug company representatives.52 In 1961, the manufacturer withdrew thalidomide from the world market.2,3,4,5,15,16,53,54
A few years later, there were reports that leprosy patients with inflammatory lesions related to ENL reactions had experienced improvement in these lesions while receiving thalidomide as a sedative.2,4,5,16,52,53,104,112 The discovery of thalidomide's therapeutic efficacy in the treatment of ENL led to investigations that revealed the anti-inflammatory and immunomodulatory effects of the drug and prompted studies evaluating the drug in various autoimmune or inflammatory diseases and various conditions in HIV-infected patients.2,4,5,16,52,53,104,112 In addition, after in vitro and animal studies indicated that thalidomide has anti-angiogenesis activity, investigations were initiated to evaluate use of the drug for a variety of neoplastic diseases.2,112,115 Most information to date regarding use of thalidomide for the treatment of inflammatory or immune disorders or for the treatment of patients with HIV infection or malignancy consists of case reports or data obtained from uncontrolled studies in a limited number of patients.4,20,23,104,112 Well-controlled clinical studies are needed to evaluate the safety and efficacy of thalidomide for the treatment of these various indications.20,23,104,112
Thalidomide is used for acute treatment of cutaneous manifestations of moderate to severe erythema nodosum leprosum (ENL) reactions (lepra type 2 reactions) in leprosy patients,1,10,13,34,35,39,44,76,86,89,111,113,114,218,220,232,233,234,235,237,238 and also is used for maintenance therapy for prevention and suppression of cutaneous manifestations of ENL recurrence.1,10 Thalidomide should not be used alone as monotherapy for treatment of ENL reactions complicated by neuritis.1,53,54 In these situations, thalidomide generally is initiated in conjunction with systemic corticosteroid therapy and the corticosteroid is slowly withdrawn if neuritis resolves.1,54
ENL is a recurrent, immunologically mediated syndrome that occurs principally in patients with multibacillary leprosy.3,10,45,82,84,86,111 While ENL reactions have been reported to occur in 10-50% of lepromatous leprosy patients3,10,53,84,86,88,89,111 and 25-30% of borderline lepromatous patients,86,88,89 these reactions are being reported less frequently in patients receiving the currently recommended multidrug antileprosy regimens that include clofazimine than in those who received dapsone monotherapy.88,89,111 ENL usually occurs after initiation of anti-infective therapy for leprosy (generally within the first 2 years of treatment), although occasionally it may occur spontaneously in untreated lepromatous leprosy patients or after treatment is discontinued.10,44,81,82,84,85,86,88 These reactions are considered to be a manifestation of the disease rather than an adverse reaction to antileprosy regimens.84,86,88 Clinical symptoms include cutaneous papules or nodules that are painful or tender, erythematous, and histologically vasculitic;3,10,53,84,89 the papules may pustulate and ulcerate, appear as recurrent crops, and are widely distributed, generally appearing on extensor surfaces of the extremities and on the face.3,10,84 Cutaneous symptoms of ENL may be accompanied by peripheral neuritis (usually of the ulnar nerve), fever, malaise, wasting, uveitis, lymphadenitis orchitis, and glomerulonephritis.3,10,53,84,86,89 Histologically, ENL is an acute vasculitis or panniculitis that is thought to be secondary to immune (i.e., antigen-antibody) complex deposition.10,53,83,89
Depending on the severity of manifestations, ENL reactions generally are treated using analgesics, corticosteroids, and/or thalidomide;76,86,89,111 clofazimine also has anti-inflammatory effects and is beneficial in the treatment of ENL reactions.10,76,86,88 While mild ENL reactions in some patients may be adequately managed with a nonsteroidal anti-inflammatory agent (e.g., aspirin, indomethacin) and bedrest,89,111 moderate to severe ENL reactions generally are treated with corticosteroids and/or thalidomide, and hospitalization may be required.3,10,53,63,64,66,67,69,111,232,233 The antileprosy regimen usually is continued while the ENL reaction is treated.63,65,66,67,69 Corticosteroid therapy (usually prednisolone) generally is effective for the treatment of moderate and severe ENL and usually is necessary when ENL is complicated by neuritis; however, long-term therapy may be required and there is a risk that ENL patients (especially those with chronic ENL) may become steroid dependent.10,76,81,86,89,111
In patients already receiving clofazimine as part of a multidrug antileprosy regimen, a temporary increase in clofazimine dosage may allow a reduction in corticosteroid requirements; clofazimine dosage effective for the treatment of ENL reactions is slightly higher than that usually recommended for the treatment of leprosy.64,66,67,89,111 However, clofazimine is not as effective or as rapidly acting as corticosteroids or thalidomide in the treatment of ENL35,86,89,111 and should not be initiated as the sole agent for the treatment of severe ENL.35,111
Thalidomide is effective for the treatment of moderate to severe ENL reactions,34,35,39,44,45,76,89,111,113,114 and some clinicians suggest that thalidomide may be the drug of choice for the treatment of these reactions, especially severe, recurrent reactions, in part because a prompt response to thalidomide generally is obtained and because of concerns related to long-term use of corticosteroids in leprosy patients.10,86,89,111 However, the risks versus benefits of thalidomide therapy must be considered, especially in females of reproductive age.10,76,89,111 Early diagnosis and treatment of ENL is important since these reactions are associated with considerable morbidity, especially in chronic, recurrent ENL.10,76,89 Therapy for leprosy and leprosy reactional states should be undertaken in consultation with an expert in the treatment of leprosy.10,74 In the US, the National Hansen's Disease (Leprosy) Program at 800-642-2477 should be contacted for further information on the management of leprosy reactional states.74
Information regarding safety and efficacy of thalidomide for the treatment of cutaneous manifestations of moderate to severe ENL is derived from experience with the drug, open-label and controlled studies and case reports involving leprosy patients from many different countries, and a retrospective study of leprosy patients treated in the US by the US Public Health Service (USPHS).1,34,35,38,54,113,114 There is evidence that use of thalidomide in leprosy patients with moderate to severe ENL reactions can result in prompt resolution of the signs and symptoms of ENL.1 In one controlled study in patients with ENL cutaneous lesions who received 7-day regimens of thalidomide (400 mg daily) or placebo, a response (i.e., complete remission of symptoms or advanced remission of about 50% of existing ENL skin lesions) occurred in 66 or 9% of treatment courses in patients receiving thalidomide or placebo, respectively.1,39 In a similar study, a complete response (i.e., the absence of skin lesions) occurred in 75 or 25% of treatment courses in patients receiving thalidomide or aspirin, respectively.1,34,233
In a double-blind, placebo-controlled, randomized, dose-comparison study, 22 male patients with ENL received 6 capsules containing either 100 mg (group A; 12 patients) or 300 mg (group B; 10 patients) of thalidomide for 1 week.238 A 6-week, 4-capsules per day tapering followed in which patients in group A received a 50-mg thalidomide dosage daily in weeks 2 and 3, followed by placebo capsules in weeks 4 through 7, while patients in group B received gradual dosage decreases every 2 weeks.238 Comparable improvement was observed at day 7 in 12 or 7 patients in group A or B, respectively.238 However, slower tapering in group B was associated with less reemergence of ENL through week 7 than patients in group A.238 Most patients developed new lesions soon after discontinuing treatment.238 Some clinicians suggest that slower tapering from a higher initial thalidomide dosage may improve ENL responses, although high recurrence rates after discontinuance indicate that further studies are needed to identify better tapering regimens.238
Safety and efficacy of thalidomide have been evaluated in a prospective, comparative (versus prednisolone) study in 60 patients with moderate to severe ENL.235 Patients were randomized to receive a thalidomide dosage of 300 mg daily for 1 week, which was gradually reduced by 50 mg every 2 weeks (30 patients), or prednisolone 40 mg daily for 2 weeks, which was subsequently tapered by 10 mg every 2 weeks (30 patients).235 Faster clinical response (cutaneous and systemic), fewer relapses, and longer periods of remission were observed in patients receiving thalidomide compared with those receiving prednisolone.235 In addition, adverse effects were more frequently observed in patients receiving prednisolone than in those receiving thalidomide.235
Efficacy of thalidomide for the treatment of chronic ENL reactions has been evaluated in 2 limited crossover studies of hospitalized, corticosteroid-dependent ENL patients who were receiving dapsone and were treated with a 4- to 6-week regimen of thalidomide (300 mg daily) or placebo.1,35 In the first crossover study, the weekly corticosteroid dosage could be reduced in about 89% of patients while thalidomide was administered, but corticosteroid dosage had to be increased when placebo was administered.1,35 In the second crossover study, 100 or 12.5% of patients decreased their weekly dosage of corticosteroids while receiving a 6-week regimen of thalidomide or placebo, respectively.1,35
Information regarding safety and efficacy of thalidomide for the prevention of ENL relapse is limited, and labeling for this use is based on data obtained from a retrospective evaluation of ENL patients treated by the USPHS.1 A subset of these ENL patients receiving thalidomide had repeated relapse of ENL symptoms when thalidomide was discontinued, but remission of symptoms when treatment with the drug was reinitiated.1
Thalidomide is used in combination with dexamethasone for the treatment of newly diagnosed multiple myeloma.1,100,161,162,163,198,199,200,201,242 Efficacy of thalidomide in the treatment of newly diagnosed multiple myeloma has been studied in a randomized, multicenter, open label study and a randomized, multicenter, double-blind, placebo-controlled clinical trial.1,255
In the open-label study, patients were randomized to receive thalidomide (200 mg daily; 103 patients) and dexamethasone (40 mg once daily; 104 patients) on days 1-4, 9-12, and 17-20 every 28 days) or dexamethasone alone (40 mg once daily on days 1-4, 9-12, and 17-20 every 28 days).1,242 Each study population group received four 28-day cycles.1,242 Response rates, based on serum or urine paraprotein measurements, were higher in those receiving combination therapy with thalidomide and dexamethasone compared with those receiving dexamethasone alone (52 vs 36%, respectively).1
In the double-blind study, patients were randomized to receive thalidomide (50 mg daily escalated to 100 mg daily on day 15, followed by 200 mg daily beginning on day 1 of cycle 2) and dexamethasone (40 mg daily on days 1-4, 9-12, and 17-20 during cycles 1-4, followed by 40 mg daily on days 1-4) or dexamethasone and placebo; treatment cycles were repeated every 28 days.1,255 Treatment was continued until disease progression.1,255 Time to progression was significantly longer in patients receiving thalidomide and dexamethasone compared with those receiving placebo and dexamethasone (97.7 versus 28.3 weeks; hazard ratio 0.43 [95% CI 0.32-0.58]).1 Overall response rate (according to the Bladé criteria) was also higher in patients receiving thalidomide and dexamethasone compared with those receiving placebo and dexamethasone (63 versus 46%).1 No significant difference was observed between the two treatment groups in overall survival (24 versus 29%; hazard ratio 0.82 [95% CI 0.57-1.16]).1
Use of thalidomide in patients with multiple myeloma is associated with increased risk of thromboembolic events (e.g., deep venous thrombosis, pulmonary embolus).1 Such risk increases substantially when thalidomide is used in combination with standard chemotherapy, including dexamethasone.1 (See Thromboembolic Events under Cautions.)
Thalidomide, Bortezomib, and Dexamethasone
Thalidomide also has been studied in combination with bortezomib and dexamethasone (bortezomib-thalidomide-dexamethasone) as induction therapy for newly diagnosed multiple myeloma in transplant-eligible patients in several phase 3, open-label, randomized studies.10013,10014,10015,10027 The GIMEMA and PETHEMA/GEM studies compared bortezomib-thalidomide-dexamethasone with the combination of thalidomide and dexamethasone (thalidomide-dexamethasone),10013,10014 whereas the IFM2013-04 study compared bortezomib-thalidomide-dexamethasone with the combination of bortezomib, cyclophosphamide, and dexamethasone (bortezomib-cyclophosphamide-dexamethasone).10027
In the GIMEMA study in 480 patients with previously untreated multiple myeloma, postinduction complete plus near-complete response rate was 31% in patients receiving bortezomib-thalidomide-dexamethasone compared with 11% in those receiving thalidomide-dexamethasone; improved responses also were observed with bortezomib-thalidomide-dexamethasone following first (52 versus 31%) and second (55 versus 41%) autologous stem cell transplants and subsequent consolidation therapy (62 versus 45%).10013 The median time to best complete or near-complete response was shorter in patients receiving bortezomib-thalidomide-dexamethasone compared with those receiving thalidomide-dexamethasone (9 versus 14 months).10013 At a median follow-up of 36 months, progression-free survival was prolonged in patients who received bortezomib-thalidomide-dexamethasone compared with those who received thalidomide-dexamethasone (hazard ratio 0.63); however, no difference in estimated 3-year overall survival (86 versus 84%, respectively) was observed between the groups.10013 Subgroup analysis based on poor prognostic factors (i.e., older than 60 years of age, advanced disease stage, elevated serum LDH concentration, high infiltration of bone marrow plasma cells, chromosome 13q deletion [del(13q)], t(4;14) translocation with or without chromosome 17p deletion [del(17p)]) suggested that the progression-free survival benefit of bortezomib-thalidomide-dexamethasone was consistent across these subgroups; however, because the proportion of patients with the del(17p) chromosomal abnormality was substantially smaller than the proportion of patients with other high-risk cytogenetic abnormalities, the effect of bortezomib-thalidomide-dexamethasone on progression-free survival in patients with the del(17p) chromosomal abnormality was not evaluated.10013
In the PETHEMA/GEM study in 386 patients with newly diagnosed multiple myeloma, the complete response rate (35 versus 14%) was higher following induction therapy with bortezomib-thalidomide-dexamethasone compared with thalidomide-dexamethasone; improved complete response rates also were maintained following autologous stem cell transplantation (46 versus 24%).10014 Among patients with high-risk cytogenetic features, complete response rate was substantially higher in patients who received bortezomib-thalidomide-dexamethasone compared with those who received thalidomide-dexamethasone (35 versus 0%); complete responses were achieved in 38 or 58% of patients receiving bortezomib-thalidomide-dexamethasone who had t(4;14) or del(17p) chromosomal abnormalities, respectively, compared with 0% of patients receiving thalidomide-dexamethasone who had either chromosomal abnormality.10014 At a median follow-up of 35.2 months, median progression-free survival was prolonged in patients who received bortezomib-thalidomide-dexamethasone compared with those who received thalidomide-dexamethasone (56.2 versus 28.2 months); however, no difference in estimated 4-year overall survival (74 versus 65%, respectively) was observed between the treatment groups.10014
In the IFM2013-04 study in 340 patients with previously untreated multiple myeloma, the overall response rate (92.3 versus 83.4%) and the rate of very good partial response or better (66.3 versus 56.2%) were higher following induction therapy with bortezomib-thalidomide-dexamethasone compared with bortezomib-cyclophosphamide-dexamethasone; however, no significant difference in complete response rate was observed between the treatment groups.10027
Because peripheral neuropathy occurs frequently with thalidomide- and bortezomib-based induction regimens, a modified bortezomib-thalidomide-dexamethasone induction regimen using reduced dosages of thalidomide and bortezomib was compared with bortezomib-dexamethasone in another phase 3 study in 199 patients.10015 In this study, the rate of very good partial response or better following induction therapy was higher with bortezomib-thalidomide-dexamethasone compared with bortezomib-dexamethasone (49 versus 36%); however, postinduction complete response (13 and 12%, respectively) and overall response (88 and 81%, respectively) rates were similar for both treatment groups.10015 At a median follow-up of 32 months, median progression-free survival was similar in patients who received bortezomib-thalidomide-dexamethasone compared with those who received bortezomib-dexamethasone (26 versus 30 months, respectively).10015
Use of thalidomide in combination with bortezomib and dexamethasone also was investigated in an open-label, noncomparative phase 2 study in 98 patients randomized to receive bortezomib-thalidomide-dexamethasone with or without cyclophosphamide.10016,10020 In this study, postinduction rates of overall response and complete plus near-complete response were 100 and 51%, respectively, in patients receiving bortezomib-thalidomide-dexamethasone and 94 and 43%, respectively, in those receiving bortezomib-thalidomide-dexamethasone-cyclophosphamide.10016 At a median follow-up of 64.8 months, median progression-free survival was 56.3 months with bortezomib-thalidomide-dexamethasone and 36.3 months with bortezomib-thalidomide-dexamethasone-cyclophosphamide; the difference was not statistically significant and was largely related to an imbalance between treatment groups in the number of patients receiving subsequent therapy for relapsed disease prior to meeting the specified criteria for disease progression.10020
Use of bortezomib-thalidomide-dexamethasone as induction therapy for newly diagnosed multiple myeloma in transplant-eligible patients has improved postinduction response rates and prolonged progression-free survival in several randomized controlled studies;10013,10014,10016,10027 however, the same benefits were not observed in a study evaluating a modified bortezomib-thalidomide-dexamethasone regimen (i.e., reduced thalidomide and bortezomib dosages).10015 Therefore, the role of a modified bortezomib-thalidomide-dexamethasone regimen using reduced dosages of thalidomide and bortezomib10015 is unclear.10033 In addition, in the absence of studies including adequate numbers of patients with the del(17p) chromosomal abnormality, attempts to identify whether these patients might derive clinical benefit (e.g., prolonged progression-free survival) from bortezomib-thalidomide-dexamethasone induction therapy are speculative; additional studies are needed to identify subgroups of patients with high-risk cytogenetic features who might benefit from such therapy.10033 The AHFS Oncology Expert Committee concluded that use of thalidomide in combination with bortezomib and dexamethasone as induction therapy for newly diagnosed multiple myeloma in transplant-eligible patients may be considered a reasonable choice (accepted; with possible conditions);10033 however, use of a modified bortezomib-thalidomide-dexamethasone regimen using reduced dosages of thalidomide and bortezomib10015 is not fully established because of unclear risk/benefit and/or inadequate experience.10033 (See Thalidomide, Bortezomib, and Dexamethasone under Dosage and Administration.) Factors that should be considered when selecting a combination chemotherapy regimen for use as induction therapy for newly diagnosed multiple myeloma in transplant-eligible patients include performance status and preexisting conditions (e.g., peripheral neuropathy).10033
Use of thalidomide in patients with multiple myeloma is associated with increased risk of thromboembolic events (e.g., deep venous thrombosis, pulmonary embolus).1 Such risk increases substantially when thalidomide is used in combination with standard chemotherapy.1 (See Thromboembolic Events under Cautions.)
Inflammatory and/or Dermatologic Disorders
Based on the beneficial effects of thalidomide in the treatment of inflammatory dermatoses associated with ENL, the drug has been used for the treatment of a variety of inflammatory and/or dermatologic disorders, including Behcet syndrome,49,77,93,104,125,126,128,129,130,131,132,133,134,135,136,137,215,218,219,232,237 erosive lichen planus,104,127,215,219,232 erythema multiforme,181,182,215,218,219,232,237 lupus erythematosus (discoid lupus erythematosus, subacute or chronic cutaneous lupus erythematosus, cutaneous manifestations of systemic lupus erythematosus),3,4,53,59,104,110,121,122,123,124,196,215,218,219,220,229,237 prurigo nodularis,59,60,94,104,117,118,215,219,229,232 actinic prurigo,104,119,218,219,229,232 cutaneous Langerhans cell histiocytosis,4,104,183,184,232,237 uremic pruritus,104,180,215,237 porphyria cutanea tarda,195,215 and pyoderma gangrenosum.4,5,53,104,125,126,219,232,237 There is evidence that thalidomide can be beneficial and may result in improvements in the dermatologic and mucocutaneous manifestations of these conditions;59,117,121,122,123,180,195,196,215 however, there is recurrence of lesions and symptoms in many patients when the drug is discontinued and long-term maintenance therapy may be necessary.59,60,124 Most clinicians recommend that thalidomide be used for the treatment of inflammatory and/or dermatologic disorders only in selected patients with severe refractory disease unresponsive to other appropriate agents (e.g., corticosteroids).93,156,186,256
In patients with Behcet syndrome,3,4,5,6,49,52,53,77,93,125,126,128,129,130,131,132,134,137 a chronic inflammatory disease of unknown etiology characterized by recurrent orogenital ulceration, skin lesions, and arthritic symptoms that may be complicated by GI, ocular, and neurologic involvement,93,131 thalidomide therapy (100-400 mg daily) has resulted in resolution of dermatologic, mucocutaneous, and arthritic manifestations of the disease.49,77,93,125,126,128,129,130,131,134,137,216,219 However, symptoms and/or lesions generally recur within several weeks after the drug is discontinued.49,77 In a randomized, double-blind, placebo-controlled study in adult males with Behcet syndrome with mucocutaneous lesions (without major organ involvement), a complete response (resolution of orogenital ulcers and follicular lesions) was obtained in 6 or 16% of patients receiving thalidomide in a dosage of 100 or 300 mg daily, respectively; there were no complete responses in those receiving placebo.49 While there are some reports that GI, ophthalmic, and/or neurologic symptoms also may improve during thalidomide therapy,49,128,132,133,136 further study is needed to determine whether thalidomide has any clinically important effect on these manifestations of the disease.49
In a clinical trial, thalidomide has been used in a limited number of young patients (9-34 years of age) with juvenile-onset of severe Behcet syndrome (manifested by severe abdominal pain; intestinal ulceration, bleeding, perforation, or obstruction; recurrent stomatitis; oral aphthosis; genital ulceration; erythema nodosum in the extremities; ileocolitis; fever).210 All patients, but one, were receiving corticosteroids; severe steroid toxicity, osteoporosis, and/or steroid dependence was reported.210 The initial thalidomide dosage was 2 mg/kg daily; dosage was increased to 3 mg/kg daily or decreased to 0.5-1 mg/kg daily, according to patients' response.210 All patients showed improvement in clinical symptoms and all patients were able to discontinue corticosteroids.210 The end point of the trial was complete remission of the disease without using the drug; however, clinicians failed to withdraw thalidomide from all patients except one.210 Further large-scale clinical trials are needed to determine the appropriate duration of thalidomide therapy.210
Some clinicians state that in patients with Behcet syndrome with mucocutaneous involvement, thalidomide may be considered in select cases based on patient preference.258 Some clinicians also state that in patients with refractory and/or severe GI involvement (e.g., GI perforation, serious GI bleeding, obstruction), anti-tumor necrosis factor (anti-TNF; anti-TNF-α) monoclonal antibodies (e.g., infliximab, adalimumab) and/or thalidomide should be considered based on retrospective data indicating clinical benefit in patients with severe GI involvement refractory to conventional treatment modalities (e.g., azathioprine, corticosteroids).257,258
Thalidomide has not been effective when used for the treatment of toxic epidermal necrolysis.97 In a limited placebo-controlled study in adults with toxic epidermal necrolysis, there was no evidence that thalidomide had any beneficial effect on necrolysis, and patients receiving thalidomide had a higher mortality rate than those receiving placebo.97 In another randomized placebo-controlled trial in patients with toxic epidermal necrolysis, therapy with thalidomide was not shown to be effective and was associated with substantially higher mortality than placebo.211 Further randomized, controlled studies are needed to understand the mechanism of toxic epidermal necrolysis and to evaluate treatments of the disease especially in patients receiving high doses of steroids and IV immunoglobulins.211 In addition, there have been several reported cases of patients with malignancies developing toxic epidermal necrolysis while receiving thalidomide (usually with dexamethasone).197,212,214
Thalidomide has been studied for a variety of uses in HIV-infected patients, including treatment of HIV-associated aphthous ulcers,112,142,143,144,146,147,218,220,221,223 HIV-associated diarrhea,56,112 HIV-associated wasting syndrome,11,41,50,112,140,141,224 and Kaposi's sarcoma.30,140,141,187,218,219,232,237 The drug also has been used as an adjunct to anti-infective agents in the treatment of mycobacterial infections, including Mycobacterium tuberculosis and M. avium complex (MAC) infections, in HIV-infected patients.40,42,53,218 Although thalidomide may provide some benefits in HIV-infected patients (e.g., promote healing of aphthous ulcers, ameliorate weight loss), further study is needed to evaluate safety and efficacy of thalidomide in HIV-infected patients and to determine whether the drug may increase viral load (i.e., plasma HIV-1 RNA levels) and/or may be associated with an increased incidence of adverse effects in this patient population.5,11,40,41,42,55,58,104
HIV-associated Aphthous Ulcers
Thalidomide (100-300 mg once daily) has been effective for the treatment and prevention of oropharyngeal, esophageal, and anogenital aphthous ulcers in HIV-infected patients.3,4,5,6,52,53,55,139,142,143,144,145,146,147,221,223,237 The drug has been effective for the treatment of severe recurrent aphthous stomatitis in HIV-infected patients, and also has been effective for the treatment of severe recurrent aphthous stomatitis in immunocompetent patients or patients who are immunocompromised but not HIV-infected (see Uses: Recurrent Aphthous Stomatitis).138,139,148,149,219 Thalidomide is not considered a drug of first choice for the treatment of aphthous ulcers in HIV-infected patients,55,102,186 but may be effective in patients with recurrent ulcers refractory to other therapies (e.g., corticosteroids).55,142,143,144,147,221
Efficacy of thalidomide for the treatment of oral and esophageal aphthous ulcers in HIV-infected patients has been evaluated in a phase II double-blind, placebo-controlled study through the National Institute of Allergy and Infectious Diseases (NIAID) AIDS Clinical Trials Group (ACTG) (study ACTG 251).12,221,223 HIV-infected patients with oral aphthous ulcers were randomized to receive a 4-week regimen of thalidomide (200 mg once daily) or placebo; if healing of ulcers was not complete after this regimen, an additional 4-week regimen of thalidomide was administered.12 At 4 weeks, a complete response was obtained in 55 or 7% of patients receiving thalidomide or placebo, respectively.12
HIV-associated Wasting Syndrome
Thalidomide has been used for the treatment of wasting syndrome in a limited number of HIV-infected patients11,41,140,141,224 and also has been used for the treatment of cachexia in other severely immunocompromised patients (e.g., cancer patients). There is some evidence that thalidomide therapy (50-400 mg daily) may decrease elevated TNF-α plasma concentrations and may promote weight gain in HIV-infected patients.3,11,40,41,42,50,141,224 However, thalidomide may increase plasma TNF-α concentrations in some HIV-infected patients.1,12,186,224 The precise role of TNF-α in the wasting syndrome is unclear and additional study is necessary to evaluate the safety and efficacy of thalidomide in the treatment of HIV-associated wasting syndrome.1,11,41,42,43,112
Thalidomide has been used with some success in a limited number of patients for the treatment of HIV-associated diarrhea, including microsporidiosis.56,112 In one group of HIV-infected patients with chronic diarrhea and weight loss caused by Enterocytozoon bieneusi infection that was not responsive to albendazole, use of thalidomide (100 mg daily for 4 weeks) had a beneficial effect (i.e., complete or partial clinical response including increased weight and decreased stool frequency) in 55% of patients.56 Although there is evidence that thalidomide may relieve some symptoms of microsporidian enteritis in some patients, results of an in vitro study indicate that the drug has no direct activity against microsporidia.193
Safety and efficacy of thalidomide for the management of cutaneous Kaposi sarcoma have been evaluated in a phase II dose-escalating study through the National Cancer Institute (NCI).227 Twenty HIV-infected male patients (29-49 years of age) with biopsy-confirmed Kaposi sarcoma (having at least 5 assessable lesions and objective evidence of tumor progression over 2 months before enrollment), received an initial thalidomide dosage of 200 mg daily; dosage was increased by 200 mg daily every 14 days up to a maximum of 1 g daily (for up to 1 year).227 Dosage reductions or delays in dose escalation were permitted for toxicity.227 To be eligible for the study, patients were required to be either on stable antiretroviral therapy for at least 4 weeks or not receiving such therapy for at least 2 weeks.227 Seventeen patients were assessed for response, using the NIAID ACTG criteria.227 Eight patients achieved partial response (defined as no progressive disease and at least a 4-week persistence of either a 50% decrease in the sum of the cross products of the lesions, a 50% reduction in the total number of lesions, or flattening of 50% or more of the nodular lesions) for an overall major response rate of 47%.227 When all 20 patients were assessed on an intent-to-treat basis, the response rate (partial and complete [defined as the absence of any evident disease for 4 weeks] responses) was 40%.227 Seven of the 20 patients achieved the maximum thalidomide dosage of 1 g daily used for a median of 19.9 weeks.227 Overall, the results suggest that thalidomide has activity against Kaposi sarcoma at dosages that can be tolerated for 6 months or more.227 The severity of adverse effects was graded according to toxicity criteria established by the NCI.227 The principal toxicity that limited dose escalation was drowsiness.227
Preliminary results from a case report and 2 small trials have suggested that Kaposi sarcoma may respond to thalidomide dosages as low as 100 mg daily.227 Further studies are needed to assess the dose-response relationship of thalidomide for this condition.227
Because there is evidence that thalidomide is an inhibitor of angiogenesis, the drug is being evaluated for the treatment of a variety of malignancies, including advanced or metastatic breast cancer,32,173,175 Kaposi sarcoma30,115,170 (see Kaposi Sarcoma under Uses: Use in HIV-infected Patients), melanoma,175,232,237 ovarian cancer, myelodysplastic syndrome (MDS),229 advanced pancreatic cancer,229 primary brain tumors (recurrent high-grade astrocytomas and mixed gliomas),29,169,174,175,229 androgen-independent prostate cancer,168,229 and renal carcinoma.175,229
Thalidomide also has been used for the treatment of cachexia in patients with advanced cancer, and there is some evidence that the drug may provide benefits in these patients in terms of improvement in symptoms of insomnia, nausea, appetite, and feelings of well-being.172 (See HIV-associated Wasting Syndrome under Uses.)
Thalidomide has been used for the treatment of graft-versus-host disease (GVHD) in adult and pediatric bone marrow transplant recipients,75,101,109,151,152,153,155,218,219,232,237 but has been associated with increased morbidity and mortality when used for prophylaxis of chronic GVHD in adult bone marrow transplant recipients.189
Thalidomide has been used in allogeneic bone marrow transplant recipients for the treatment of chronic or refractory GVHD unresponsive to other therapies (e.g., corticosteroids, azathioprine, tacrolimus, cyclosporine, antithymocyte globulin);75,101,109,151,152,153,155 only limited information is available regarding use of the drug for the treatment of acute GVHD.75 In a study in adult and pediatric patients with chronic or refractory GVHD who received thalidomide therapy (800-1600 mg daily for a median duration of 240 days), a complete response was obtained in 32% and a partial response was obtained in 27%; the overall survival rate was 64%.153 Thalidomide has been effective when used for the treatment of chronic or corticosteroid-dependent GVHD in pediatric patients 0.5-17 years of age.75,109,151,152,153,155,160
When thalidomide was used for prophylaxis of chronic GVHD in a controlled study of adult allogeneic bone marrow transplant patients who had no evidence of active acute GVHD at study entry, those receiving thalidomide (200 mg twice daily beginning 80 days after transplant) experienced a higher incidence of chronic GVHD and had a substantially higher mortality rate compared with those receiving placebo.186,189 Chronic GVHD occurred in 64 or 38% of patients receiving thalidomide or placebo, respectively;189 overall mortality was about 39 or 8%, and mortality from GVHD was about 21 or 4% in those receiving thalidomide or placebo, respectively.189
Thalidomide also has been used in the treatment of GVHD in adult peripheral blood stem cell transplant recipients.236,239
Thalidomide has been effective when used for the treatment of severe oral ulcers in patients with recurrent aphthous stomatitis (RAS).138,139,148,149,219 The drug has been used effectively for the treatment of RAS in immunocompetent adults and pediatric patients and also has been effective when used in immunocompromised patients, including cancer patients and HIV-infected patients (see HIV-associated Aphthous Ulcers under Uses).138,139,148,149,150 In a placebo-controlled cross-over study in patients with severe RAS, a complete clinical remission was obtained in 48 or 9% of patients receiving thalidomide or placebo, respectively.150 Oral ulcers generally heal within 1-6 weeks after initiation of thalidomide therapy (100-300 mg daily), but higher dosage may be needed in some patients (400-600 mg daily) and ulcers may relapse when the drug is discontinued.148,149,150 In some patients, relapse may be prevented or treated using thalidomide maintenance therapy (50-100 mg daily).148,149,150,222 In one small clinical trial evaluating thalidomide (100 mg daily for 10 days, followed by 50 mg daily for 10 days, and then 25 mg daily for 10 days) compared with prednisone (0.4 mg/kg per day for 15 days followed by 0.2 mg/kg per day for 15 days), thalidomide prolonged the recurrence interval compared with prednisone at 3 months.259
Thalidomide has been used with some success in a limited number of patients for the treatment of refractory Crohn disease (e.g., in those with moderately to severely, chronically active or fistulizing disease).4,53,133,136,157,158,159,202,203,204,205,207,208,209,229,230 In patients who did not respond to or were intolerant of usually recommended therapies (e.g., corticosteroids, sulfasalazine, azathioprine, mercaptopurine, methotrexate), thalidomide therapy (50-300 mg daily) resulted in resolution of small bowel ulceration, bleeding, or pain157,158 and improvement related to quality of life or symptoms associated with fistulas.202,203,204,205 In at least one patient with Crohn disease, thalidomide therapy also resulted in resolution of severe, recurrent oral aphthous ulcers.95 Thalidomide has been used in a limited number patients with Crohn disease (i.e., active fistulous disease, luminal disease) who developed infliximab-induced delayed hypersensitivity reactions.231 Complete closure was observed in 2 patients, one with a single perirectal fistula, the other with 5 perianal fistulae at 4 or 12 weeks of thalidomide therapy, respectively.231 Two patients who continued thalidomide for more than 3 months remained in remission at 5 and 7 months.231
Limited data indicate that thalidomide (100 mg daily) may be effective in maintaining response in patients with chronically active and fistulizing refractory Crohn disease whose disease has responded to infliximab (dosage of 5 mg/kg administered as one or more infusions in patients with chronically active disease and 3 or more infusions in those with fistulizing disease).202,206 The median follow-up was 238 days from initiation of thalidomide therapy and 265 days after the last infusion of infliximab.206 Remission rates (full improvement) with thalidomide were 92, 83, or 83% at 3, 6, or 12 months, respectively, after the last dose of infliximab.202,206
Results of a 12-week, open-label study in a limited number of patients (20-77 years of age) with chronic inflammatory bowel disease (IBD; 5 with Crohn disease, 2 with ulcerative colitis, and 1 with indeterminate colitis) indicate that use of thalidomide can result in clinical response in patients with chronically active Crohn disease who have not responded to prior therapy (e.g., 5-amino-salicylic acid preparations, prednisone, azathioprine).230 Initially, patients were receiving 100 mg of thalidomide daily after which the daily dosage was increased to a maximum of 400 mg according to patients' response or adverse effect profile.230 Patients continued to receive other drugs for IBD during the duration of the trial.230 The Crohn Disease Activity Index (CDAI) decreased from 117 to 48; improvement included decreased stool frequency, erythrocyte sedimentation rate (ESR), and inflammation and histological grades.230 Two months after the trial, stool frequency returned to pretreatment levels indicating that the effect of thalidomide is of short duration.230 Long term studies are needed; minimizing the dosage over an extended period may be a potential solution.230
Thalidomide also has been studied in pediatric patients (2-18 years of age) with active Crohn disease despite other immunosuppressant therapy.260 A total of 56 children and adolescents were randomized to receive thalidomide or placebo for 8 weeks.260 Patients randomized to thalidomide weighing less than 30 kg, 30-60 kg, or greater than 60 kg received daily dosages of 50, 100, or 150 mg, respectively; however, 2 small patients received an individualized dosage of 2 mg/kg per day.260 Disease activity was measured using the Pediatric Crohn Disease Activity Index.260 At 8 weeks, clinical remission rates were significantly improved in patients receiving thalidomide compared with those receiving placebo (46.4 versus 11.5%; risk ratio 4.0; 95% CI, 1.2-12.5).260 The mean duration of clinical remission in patients receiving thalidomide was 181.1 weeks compared with 6.3 weeks in those receiving placebo.260
Thalidomide also has been used with some success in a limited number of patients for the treatment of ulcerative colitis, but additional study is needed.
Thalidomide has been used in a limited number of patients for the treatment of refractory ankylosing spondylitis;98,261 refractory rheumatoid arthritis;3,4,53,112,178,179 sarcoidosis;4,99,237,263 hereditary hemorrhagic telangiectasia;264 or polyneuropathy, organomegaly, endocrinopathy, M-protein, and skin changes (POEMS) syndrome262 , but additional study and experience are needed.
Because thalidomide is an inhibitor of angiogenesis and there is some evidence that loss of vision in the severe, wet form of macular degeneration may be related to angiogenesis in the blood vessels behind the macula, the drug has been investigated for the treatment of macular degeneration.3,52,53
Dispensing and Administration Precautions
Thalidomide is administered orally.1 Because administration with a high-fat meal substantially delays the time to peak plasma concentrations of thalidomide, the drug should be administered with water at least 1 hour after a meal.1
When thalidomide is given as a single daily dose, the manufacturer recommends that the dose preferably be given at bedtime (to minimize the sedative effects of the drug) with water and at least 1 hour after the evening meal.1 When high daily dosage of thalidomide is necessary (e.g., 400 mg daily), the daily dose may be given as a single dose at bedtime or, alternatively, in divided doses given with water at least 1 hour after meals.1
For acute treatment of cutaneous manifestations of moderate to severe erythema nodosum leprosum (ENL) in adults and children 12 years of age or older, thalidomide should be initiated at a dosage of 100-300 mg daily.1 Patients weighing less than 50 kg should receive a low initial dosage of thalidomide (e.g., 100 mg daily).1 For the treatment of severe cutaneous ENL reactions or in patients who previously required high thalidomide dosages to control an ENL reaction, the drug may be initiated at a dosage up to 400 mg daily.1
Thalidomide generally should be continued until signs and symptoms of active ENL reaction have subsided (usually at least 2 weeks), and dosage should then be gradually reduced.1 The manufacturer recommends that daily dosage of thalidomide be reduced in 50-mg increments every 2-4 weeks until the drug is discontinued or recurrence of ENL occurs.1 Patients who have recurrence of ENL while thalidomide dosage is being reduced and those who have a documented history indicating that prolonged maintenance treatment is necessary to prevent recurrence of cutaneous ENL should be maintained on the minimum dosage of the drug required to control the reaction.1 Thereafter, gradual decrease and discontinuance of maintenance dosage should be attempted every 3-6 months.1
Patients with moderate to severe neuritis associated with severe ENL reactions may receive corticosteroid therapy concomitantly with thalidomide; corticosteroid dosage may be tapered and the corticosteroid discontinued when neuritis has ameliorated.1
Thalidomide in combination with dexamethasone (in 28-day treatment cycles) is used for the treatment of newly diagnosed multiple myeloma in adults.1,100,161,162,163,198,199,200,201 The usual dosage of thalidomide is 200 mg given once daily.1 (See Administration under Dosage and Administration.) Dexamethasone is administered orally as 40-mg doses on days 1-4, 9-12, and 17-20 every 28 days.1
Thalidomide, Bortezomib, and Dexamethasone
Thalidomide has been used in several regimens in combination with bortezomib and dexamethasone as induction therapy for newly diagnosed multiple myeloma in transplant-eligible patients.10013,10014,10016,10027
When thalidomide has been used in combination with bortezomib and dexamethasone as induction therapy for newly diagnosed multiple myeloma in transplant-eligible adults, thalidomide 200 mg was administered orally daily (after initial dosage escalation during cycle 1 with 100 mg orally on days 1-14 followed by 200 mg orally daily thereafter) along with bortezomib 1.3 mg/m2 by IV injection twice weekly for 2 weeks (days 1, 4, 8, and 11) and dexamethasone 40 mg orally on days 1, 2, 4, 5, 8, 9, 11, and 12.10013 Treatment cycles were repeated every 21 days for 3 cycles.10013
Thalidomide 200 mg also has been administered orally daily (after initial dosage escalation during cycle 1 with 50 mg orally on days 1-14 followed by 100 mg orally on days 15-28) along with bortezomib 1.3 mg/m2 by IV injection twice weekly for 2 weeks (days 1, 4, 8, and 11) and dexamethasone 40 mg orally on days 1-4 and 9-12.10014 Treatment cycles were repeated every 4 weeks for 6 cycles.10014
Thalidomide 100 mg has been administered orally daily along with bortezomib 1.3 mg/m2 by subcutaneous or IV injection twice weekly for 2 weeks (days 1, 4, 8, and 11) and dexamethasone 40 mg orally on days 1-4 and 9-12.10016,10027 Treatment cycles were repeated every 3 weeks for 4 cycles.10016,10027
A modified regimen using reduced dosages of thalidomide and bortezomib10015 is not fully established.10033
For the treatment of recurrent aphthous stomatitis in adults, including those who are HIV-infected, thalidomide dosages of 100-300 mg daily have been used.148,149,150 Lower thalidomide dosages of 100 mg daily for 10 days, followed by 50 mg daily for 10 days, then 25 mg daily for 10 days also have been used.259 However, higher dosages (e.g., 400-600 mg daily) may be necessary in some patients.148,149,150 The optimum duration of thalidomide therapy has not been elucidated, but ulcers may relapse following discontinuance of the drug.148,149,150
For the management of refractory Crohn disease in adults, thalidomide dosages of 50-300 mg daily have been used.157,158,159,202,203,204,205,207,208 Thalidomide dosages of 100 mg daily have been used in maintenance therapy of chronically active or fistulizing refractory Crohn disease in patients whose disease has responded to infliximab.202,206 Although data in children and adolescents (2-18 years of age) are limited, a thalidomide daily dosage of 50, 100, or 150 mg in pediatric patients weighing less than 30 kg, 30-60 kg, or greater than 60 kg, respectively, or an individualized dosage of 2 mg/kg per day has have been used.260
For the treatment of graft-versus-host disease (GVHD), thalidomide dosages of 800-1600 mg daily have been used.153 The drug should not be used for prophylaxis of chronic GVHD.153 (See Graft-versus-host Disease under Uses.)
Dosage Modification for Toxicity
Temporary interruption of therapy, dosage reduction, and/or permanent discontinuance of thalidomide may be necessary in patients experiencing grade 3 or 4 adverse effects.1 If constipation, somnolence, or peripheral neuropathy occurs, temporarily interrupt thalidomide therapy and consider dosage reduction upon resumption of therapy.1
If angioedema, anaphylaxis, grade 4 rash, skin exfoliation, bullae, or any other severe dermatologic reaction (e.g., grade 4 rash, skin exfoliation, bullae) occurs, thalidomide therapy should be permanently discontinued.1 (See Cutaneous Reactions and also Hypersensitivity Reactions under Cautions.)
The manufacturer makes no specific dosage recommendations for patients with hepatic impairment.1
The manufacturer makes no specific dosage recommendations for patients with renal impairment.1
Because thalidomide is not excreted by the kidneys, experts state that dosage modification for renal impairment is not necessary.266,267,268
No dosage adjustment is necessary in patients with end-stage renal disease undergoing dialysis; however, clinicians should be aware of the risks of thalidomide-associated hyperkalemia in patients who require dialysis.245,266,267
The manufacturer makes no specific dosage recommendations for geriatric patients.1
Fetal/Neonatal Morbidity and Mortality
Thalidomide is a known human teratogen and has caused severe, life-threatening birth defects or fetal death when used during pregnancy.1,2,3,4,5,11,12,15,20,21,52,53,54,112 Mortality at or shortly after birth has been reported in approximately 40% of infants.1
The drug can cause teratogenic effects even if only a single dose of the drug (e.g., 50-100 mg; regardless of strength) is taken during pregnancy.1,2,3,4,5,15,52 Since the risks clearly outweigh any possible benefits in women who are or may become pregnant, thalidomide is contraindicated in such women.1 If the drug is inadvertently administered during pregnancy or if a patient becomes pregnant while receiving the drug, the drug should be discontinued immediately and the patient referred to an obstetrician-gynecologist experienced in reproductive toxicity for further evaluation and counseling.1 The patient should be informed that if their clinician is not available, information about emergency contraception (including information regarding clinicians who provide emergency contraceptive services) can be obtained by calling 888-668-2528 or by using other sources (e.g., [Web]).1,24 Any suspected exposure of a fetus to thalidomide should be reported to the FDA MedWatch Program (800-FDA-1088) and also to the manufacturer, Bristol Myers Squibb Corporation (1-888-423-5436).1 Because thalidomide may cause fetal harm and because of the possibility that the drug may be present in blood and be transfused into a woman who is pregnant, patients receiving thalidomide must not donate blood during therapy and for at least 4 weeks following discontinuance of the drug.1
To minimize risk of fetal exposure and help ensure the safe and effective use of thalidomide in the US, patients must be registered in the Thalomid REMS® program before they can receive commercially available drug.1,17,20,22 All female patients of reproductive potential and all sexually mature males receiving thalidomide must use effective contraceptive measures (which may include abstinence) to help ensure that fetal exposure to the drug does not occur.1 Contraceptive measures are indicated even in females with a history of infertility.1 The only females who do not need to observe mandatory contraceptive measures are those who have undergone hysterectomy.1 All female patients of reproductive potential must use 2 reliable forms of contraception simultaneously (unless the patient chooses to remain continuously abstinent from engaging in heterosexual sexual contact) beginning at least 4 weeks prior to initiation of therapy, during therapy (including treatment interruptions), and then for at least 4 weeks following discontinuance of thalidomide therapy.1 At least 1 contraceptive method should be a highly effective method (intrauterine device [IUD], hormonal contraceptives, tubal ligation, vasectomized partner); the other may be an effective barrier method (latex or synthetic condom, diaphragm, cervical cap).1 Sexually mature males receiving thalidomide must completely avoid unprotected sexual contact with women of reproductive potential and must not donate semen while receiving thalidomide and for 4 weeks after discontinuing the drug.1 While receiving thalidomide and for up to 4 weeks after discontinuing the drug, sexually mature males (including those who have successfully undergone vasectomy) must use a latex or synthetic condom each time they have sexual contact with a woman of reproductive potential.1
Therapy with thalidomide, presence of an underlying malignancy, and/or use of an estrogen-containing contraceptive can each increase the risk of thromboembolism; however, it is not known if these risks are additive.1 Each risk factor for thromboembolism should be taken into consideration when choosing contraceptive methods.1 (See Thromboembolic Events under Cautions.)
Because some patients may develop sudden, severe neutropenia and/or thrombocytopenia during thalidomide therapy, the risk for bleeding or infection may be increased in such patients at insertion, removal, or during use of an IUD or implantable contraception.1 (See Hematologic Effects under Cautions.)
All females of reproductive potential must be tested for pregnancy within 10-14 days and again within 24 hours immediately prior to initiation of thalidomide therapy.1 The prescribing clinician should not provide the patient with a prescription for thalidomide until reports of the pregnancy tests are available indicating that the results are negative.1 Pregnancy tests must then be repeated at regular intervals during thalidomide therapy (i.e., weekly during the first month, then every 2 or 4 weeks in women with irregular or regular menstrual cycles, respectively).1 Pregnancy tests and counseling also should be performed if a patient misses her period or if there is any abnormality in menstrual bleeding.1 The drug should be discontinued during the evaluation period.1
It has been estimated that there were more than 10,000 reported cases of infants born with birth defects related to use of thalidomide in pregnant women in other countries between 1957 (when thalidomide was first introduced in the world market) and 1963 (several years after the drug was removed from the world market).115 Although thalidomide was not commercially available in the US at that time, there were at least 17 reports of infants with thalidomide-associated birth defects being born to women in the US during those years; these women had received the drug from overseas sources or received premarketing samples distributed by drug company representatives.52
A variety of human fetal abnormalities related to thalidomide administration during pregnancy have been documented.1 The most common birth defects reported following fetal exposure to thalidomide are musculoskeletal fetal abnormalities involving the loss of all or part of one or more bones;1,2,3,4,5,15,21 however, fetal deformities may occur in almost any organ of the body15 and defects of internal organs have been reported with some skeletal deformities.4,5 The mortality rate at or shortly after birth in infants born with thalidomide-induced abnormalities has been reported to be about 40%;1,15 most fatalities are related to serious malformations of internal organs.1,15
Skeletal deformities caused by thalidomide include amelia (absence of legs and/or arms)1,5,15 and absence of bones;1,15 phocomelia (short legs and/or arms)1,4,5,15,54 and bone hypoplasia;1,15 hand deformities (e.g., radial club hand);15 thumb aplasia,5,15 hypoplasia,15 triphalangia,5,15 and nonopposability;15 finger aplasia,15 hypoplasia,15 fixed flexion,15 and syndactyly;15 and hip dysplasia5 and dislocation.5,15 Skeletal deformities appear to follow the craniocaudal progression of fetal morphogenesis.15 Most individuals with thalidomide defects of the upper limbs have normal lower limbs, some have defects of all limbs, but those with normal upper limbs and deformed lower limbs are rare.15 When substantial parts of bones are missing, the muscles normally attached to them are hypoplastic, but the degree of muscle hypoplasia does not always correspond to the level of bone loss (e.g., marked hypoplasia of shoulder and upper arm muscles may occur in a patient with a humerus of normal length).15 In general, more pronounced shortness of stature than that associated only with short leg bones occurs,15 resulting from poor growth, spinal osteochondritis, and progressive kyphosis.15 Thalidomide-induced skeletal deformities usually are bilaterally symmetric, similar to malformations reported with other teratogenic drugs.15 However, the extent of symmetry varies with the nature of the defect, both in the number of appreciably asymmetric deformities and in the closeness of the match between left and right.15
The next most common group of abnormalities reported following fetal exposure to thalidomide are craniofacial and orofacial fetal abnormalities involving the ear, eye, and nerve supplies to the face, external ocular muscles, and lacrimal glands;15,21 these deformities tend to occur in a variety of combinations and permutations.15 External ear deformities tend to be bilateral and symmetric,15 and include anotia,1,15 microtia or micropinna,1,15 and small or absent auditory canals.1,5,15 Individuals with anotia and a blind or absent external auditory canal (meatus) are profoundly deaf in the affected ear.15 Facial palsy may occur1,15 (usually unilaterally rather than bilaterally) and almost always is associated with anotia or microtia on the same side.15 Abnormalities of ocular structures occur frequently,2,5,15 including anophthalmos,1,15 microphthalmos1,15 coloboma of the iris and retina,15 and conjunctival dermoid cyst.15 Microphthalmos and coloboma often occur together, and are predominantly bilateral.15 Restricted ocular movements may occur;15 ocular movement defects are nearly always associated with otic defects, often associated with facial weakness, and tend to be bilateral.15 Tear-saliva syndrome (crocodile tears) has been reported, in which tears are secreted in association with eating (rather than saliva) but may not be secreted in association with crying.15 This syndrome results from incorrect neural connections (probably in the brain stem), is bilateral or unilateral, and usually is associated with otic abnormalities and ocular movement defects.15 Less common orofacial defects that have reported include cleft palate,15 high arched palate,15 bifid uvula,15 palatal palsy,15 cleft lip,15 choanal atresia,15 and mandibular and dental abnormalities.15
Other fetal deformities that have been associated with thalidomide exposure during pregnancy include congenital heart defects and other cardiovascular malformations (e.g., patent ductus arteriosus, ventricular and/or atrial septal defects, pulmonary stenosis);1,5,15,21 renal and urinary tract malformations (e.g., defects of the kidney, ureter, and bladder);15,21 genital malformations (e.g., hypospadias, vaginal atresia, fallopian tube interruption, bicornate uterus, hypoplasia of the scrotum or labia, and undescended, small, or absent testis);1,15,21 GI tract malformations (e.g., duodenal atresia, duodenal stenosis, pyloric stenosis, anorectal stenosis, imperforate anus with fistula, anteriorly placed anus); gallbladder aplasia;5,15 and respiratory tract malformations.5
Most teratogenic effects appear to have occurred as the result of exposure to thalidomide early in pregnancy, and there is some evidence that a distinct window of embryonic sensitivity to thalidomide's teratogenic effects exists (i.e., from about 34-50 days after the beginning of the last menstrual period).1,4,5,15 Some malformations appear to be associated with particular periods of embryonic exposure, including aplasia of the ear or stenosis of the rectum which appear to be associated with exposure to thalidomide at 34-38 or 49-50 days, respectively, after the beginning of the last menstrual period.5,15 However, incorrect reporting of the dates of last menstrual periods and/or thalidomide administration dates as well as atypical menstrual cycle duration may have resulted in considerable inaccuracy in these data.5 It has been suggested that limb defects may be secondary to an inhibition of blood vessel growth in the developing fetal limb bud,4 and studies of thalidomide's effects in fetal animal tissues have suggested that neural as well as limb development effects occur.4 The risk of potentially severe birth defects occurring following fetal thalidomide exposure later in pregnancy is unknown, but may be substantial.1 Animal studies to characterize the effects of thalidomide on late stage pregnancy have not been conducted.1 While numerous mechanisms for thalidomide-induced fetal abnormality and mortality have been proposed, some have been refuted and others have not been adequately studied;4,46 additional study is required to elucidate how thalidomide causes teratogenic effects in fetuses.1,4,46
The only type of thalidomide exposure known to have resulted in drug-associated birth defects was related to oral administration of the drug.1 Currently no specific data are available regarding cutaneous absorption or inhalation of thalidomide in women of childbearing potential and whether such exposures may result in any birth defect.1 Patients should be instructed not to handle extensively or open thalidomide capsules and to maintain capsules stored in blister packs until ingestion.1 If there has been contact with a non-intact thalidomide capsule or the powder contents, the exposed area should be washed with soap and water.1 Thalidomide has been shown to be present in the serum and semen of patients receiving thalidomide.1 If healthcare providers or other care givers are exposed to body fluids from patients receiving thalidomide, appropriate precautions should be taken (e.g., wearing gloves to prevent the potential cutaneous exposure to thalidomide, washing the exposed area with soap and water).1
Use of thalidomide in patients with multiple myeloma is associated with increased risk of arterial or venous thromboembolic events (e.g., deep-vein thrombosis, pulmonary embolism).1 Such risk increases substantially when thalidomide is used in combination with standard chemotherapy, including dexamethasone.1 In a controlled clinical trial, an increased incidence of venous thromboembolic events was observed in patients receiving thalidomide in combination with dexamethasone compared with those receiving dexamethasone alone (22.5 versus 4.9%).1 Ischemic heart disease, including myocardial infarction and stroke also has been reported in patients with previously untreated multiple myeloma receiving thalidomide and dexamethasone.1
Therapy with thalidomide, presence of an underlying malignancy, and/or use of an estrogen-containing contraceptive can each increase the risk of thromboembolism; however, it is not known if these risks of thromboembolism are additive.1 Each risk factor for thromboembolism should be taken into consideration when choosing contraceptive methods.1
Patients and clinicians are advised to watch for signs and symptoms of thromboembolism.1 Patients should be instructed to notify a clinician if they develop shortness of breath, chest pain, and/or arm or leg swelling.1
The manufacturer and some experts state that decisions regarding use of thromboprophylaxis and selection of an appropriate thromboprophylaxis regimen (e.g., aspirin, anticoagulant) should be based on careful assessment of the patient's risk factors for thromboembolism.253,254 The International Myeloma Working Group (IMWG) currently recommends thromboprophylaxis with aspirin for multiple myeloma patients receiving thalidomide who have one or no underlying individual and/or myeloma-related risk factors for venous thromboembolism and thromboprophylaxis with a low molecular weight heparin (LMWH) for those with 2 or more individual and/or myeloma-related risk factors.253 The IMWG also recommends that thromboprophylaxis with a LMWH be considered in thalidomide-treated patients receiving high-dose dexamethasone, doxorubicin, or multiple antineoplastic agents, independent of additional risk factors.253 The IMWG states that although full-dose warfarin (international normalized ratio [INR] 2-3) is an alternative to LMWHs, there is limited clinical experience with this approach.253 The American Society of Clinical Oncology (ASCO) currently recommends pharmacologic thromboprophylaxis for multiple myeloma patients receiving thalidomide in conjunction with dexamethasone or antineoplastic agents, and states that those at lower risk for thromboembolism may receive either aspirin or a LMWH, while those at higher risk should receive a low molecular weight heparin.254
Deep-vein thrombosis and pulmonary embolism also have been reported in patients with ENL.240
Other Warnings and Precautions
Increased mortality has been reported in clinical trials in patients with multiple myeloma receiving pembrolizumab in combination with a thalidomide analogue and dexamethasone.1 The manufacturer of thalidomide states that an anti-programmed death receptor-1 (anti-PD-1) or anti-programmed-death ligand-1 (anti-PD-L1) antibody should not be used in combination with a thalidomide analogue and dexamethasone in patients with multiple myeloma outside of a controlled clinical trial.1 FDA recommends that ongoing clinical trials evaluating an anti-PD-1 or anti-PD-L1 agent in combination with an immunomodulatory agent (e.g., thalidomide) be evaluated for permanent discontinuance or protocol amendments.265
Patients should be warned that thalidomide frequently causes drowsiness and somnolence, and may impair their ability to perform hazardous activities requiring mental alertness or physical coordination (e.g., operating machinery, driving a motor vehicle).1 Patients also should be warned not to take any other drugs that may cause drowsiness without consulting their clinician.1 Patients should be advised that thalidomide may potentiate the drowsiness caused by alcohol.1 (See CNS Agents under Drug Interactions.)
Thalidomide may cause potentially severe nerve damage (i.e., polyneuritis or peripheral neuropathy) that may be irreversible.1,3,4,5,20,41,53,54,57,59,60,61,62,232 The incidence of peripheral neuropathy in patients receiving thalidomide has been reported to range from less than 1% to more than 70%.4,6,61,112 Incidence data regarding this adverse effect has been difficult to obtain, in part because the drug often is used in patients with diseases that may be associated with neuropathy (e.g., leprosy patients with erythema nodosum leprosum [ENL], patients with HIV infection) and it may be difficult to differentiate the neuropathologic symptoms and changes caused by the underlying disease from those caused by the drug.1,5,6,61,62,89,112
Despite long-term experience with use of thalidomide in leprosy patients with ENL, there have been few reports of thalidomide-associated peripheral neuropathy in these patients;1 in some studies, less than 1% of patients receiving the drug for treatment of ENL experienced peripheral neuropathy.4,59,60,62 It has been suggested that this may not be an accurate reflection of the incidence of this adverse effect in these patients since peripheral neuritis was not recognized as a potential adverse effect of thalidomide in early studies of the drug,86,112 in part because ENL patients frequently have neuropathy in association with their disease and may not have reported such effects while receiving the drug.76,81,82,89 Therefore, preexisting neuropathy in ENL patients may complicate recognition of thalidomide-associated peripheral neuropathy.5,89,112 In addition, such symptoms may be difficult to monitor since ENL neuritis may improve during thalidomide therapy, but relapse after therapy with the drug is discontinued.35
In controlled studies in HIV-infected patients, neuropathy occurred in 7-8% of those receiving thalidomide.41 In one limited study in HIV-infected patients receiving thalidomide 400 mg daily for 3 months, 7% of patients experienced neuropathy symptoms requiring discontinuance of the drug.41 In other limited studies, 4% of HIV infected patients receiving 200-300 mg of thalidomide for 14-21 days experienced severe symptoms of neuropathy.58 It has been suggested that HIV-infected patients may be particularly susceptible to the development of thalidomide-associated polyneuritis.5 However, since axonal degeneration with clinical and electrophysiologic symptoms resembling those of thalidomide-associated polyneuropathies may occur in HIV-infected patients and other forms of polyneuropathy (including demyelinating and polyradiculopathy) also are reported in such patients, it may be difficult to differentiate neuropathologic changes caused by the underlying disease from those caused by thalidomide.5
In a retrospective study of patients receiving thalidomide for a variety of dermatologic conditions, thalidomide-associated peripheral neuropathy occurred in 21-50% of patients and was most frequent in women and geriatric patients.61 Limited data indicate that peripheral neuropathy occurs in up to 75% of patients receiving thalidomide for the treatment of prurigo nodularis.4,59,60,61,62
Thalidomide-associated peripheral neuropathy generally has been reported with chronic use of the drug over a period of months,53,60 but also has been reported with relatively short-term use of the drug.58,61,62 The correlation between peripheral neuropathy and the cumulative thalidomide dose is unclear;1,4,5,59,60,61,62 symptoms have been reported to begin after a cumulative dose of 40-50 g in some patients,4,60,61,139 but also have occurred with no apparent relationship to cumulative dose.49,59,61 There have been some cases when symptoms of thalidomide-associated peripheral neuropathy were not apparent until after the drug was discontinued.1
Polyneuropathic symptoms may improve in some patients when thalidomide is discontinued, but symptoms may resolve slowly or not at all after discontinuance of the drug.4,5,41,59,60,61,62 In a study of patients with thalidomide-induced peripheral neuropathy who were followed for 4-6 years after discontinuing the drug, 25% experienced full recovery, 25% had some improvement of symptoms, and 50% had no improvement in signs and symptoms of neuropathy.116
Clinical symptoms of thalidomide-induced neuropathy include predominantly symmetric painful paresthesia of the hands and feet, often accompanied by sensory loss in lower limbs.4,59,60,61,62 Muscle weakness and cramps, signs of pyramidal tract involvement, and carpal tunnel syndrome have been reported often.4,59,60,61,62 An unpleasant pedal symptom, described as tightness around the feet, was reported in 8-47% of patients in some studies.4,59 In early reports, thalidomide-associated peripheral neuropathies were characterized mostly by sensory symptoms, including hypoesthesia and hyperesthesia, hyperalgesia, impaired temperature sensitivity,4,5 and impaired vegetative (i.e., autonomic) nervous functions.5 These symptoms occasionally were accompanied by mild proximal muscle weakness,4,5,61,62 or evidence of pyramidal tract damage,4 but motor disturbances appeared to occur rarely.5 Muscle weakness rapidly improved after discontinuance of the drug, but sensory deficits were slow to improve, and occasionally worsened even while the patient was not receiving thalidomide.4 Because there was no substantial correlation between the severity of the condition and the total dose of the drug, and because discontinuance of thalidomide did not result in improvement in many cases, initially it could not be established that such neurologic disturbances were actually caused by thalidomide.5 Subsequently, the association between thalidomide and peripheral neuropathies was established in clinical studies of patients with discoid lupus erythematosus, prurigo nodularis, and aphthous stomatitis; in these studies, the neurologic status of patients was assessed before and after initiation of thalidomide therapy.5
Physiologically, thalidomide polyneuropathy is characterized by axonal degeneration without demyelinization (i.e., a dying back process).4,5,60,62 Electrophysiologic alteration principally is characterized by a decreased sensory action potential amplitude on medial nerves of the arm, and peroneal and sural (medial cutaneous) nerves of the leg, with relative conservation of conduction velocities;4,59,60,61,62 however, decreased sensory and motor conduction velocities have been observed.5,60 Also, increased somatosensory latency of activation after sural nerve stimulation may occur in the absence of clinical abnormalities in patients receiving thalidomide.4,5 Because electrophysiologic alterations may occur before the onset of subjective symptoms, the reported incidence of neuropathy in thalidomide studies is higher if neurophysiologic tests, rather than clinical symptoms, are used for diagnosis.5
Patients should be instructed to immediately report initial symptoms (e.g., numbness, tingling, pain or a burning sensation in the hands and feet) of peripheral neuropathy to their prescribing clinician.1 To ensure that early signs of neuropathy are detected, patients should be examined, counseled, and questioned regularly during thalidomide therapy (i.e., monthly for the first 3 months of thalidomide treatment, and periodically thereafter).1,4
Early detection and differential diagnosis of thalidomide-induced neuritis may be difficult in leprosy patients with ENL and in HIV-infected patients because neuritis associated with these diseases may be similar to that caused by thalidomide.1,5,34,61,62,89,112
To assist in detection of asymptomatic neuropathy, the manufacturer states that consideration should be given to using electrophysiologic testing, consisting of sensory nerve action potential (SNAP) amplitude measurement at baseline and every 6 months thereafter.1 If manifestations of peripheral neuropathy develop, thalidomide should be discontinued immediately (if clinically appropriate) to minimize further damage.1 Usually, treatment with thalidomide should be resumed only if manifestations of neuropathy return to baseline.1
Drugs known to be associated with peripheral neuropathy should be used with caution in patients receiving thalidomide.1 (See Drugs Associated with Peripheral Neuropathy under Drug Interactions.)
Dizziness and Orthostatic Hypotension
Because thalidomide may cause dizziness and orthostatic hypotension,1,53,54 patients receiving the drug should be instructed to sit upright for a few minutes before standing up from a reclining position.1
Because decreased leukocyte counts, including neutropenia,1,34,53,54 have been reported in patients receiving thalidomide, the drug should not be initiated in patients with absolute neutrophil counts (ANC) less than 750/mm3.1 Leukocyte counts and differentials should be routinely monitored, especially in patients who are prone to neutropenia (e.g., HIV-infected patients).1 If the ANC decreases to less than 750/mm3 while the patient is receiving thalidomide, the patient's drug regimen should be reevaluated and consideration given to withholding thalidomide if clinically appropriate.1,20
Because grade 3 or 4 thrombocytopenia has been reported, CBCs, including platelets, should be monitored.1 If hematologic toxicity occurs, dosage interruption, dosage reduction, or discontinuance of therapy may be required.1 Patients, particularly those receiving concomitant medications that may increase the risk of bleeding, should be monitored for signs and symptoms of bleeding (e.g., petechiae, epistaxis, GI bleed).1
Because some patients may develop sudden, severe neutropenia and/or thrombocytopenia during thalidomide therapy, the risk for bleeding or infection may be increased in such patients at insertion, removal, or during use of an intrauterine device (IUD) or implantable contraception.1
Increased HIV viral load (i.e., increased plasma HIV-1 RNA levels)1,12,53,54 has been reported in some HIV-infected patients receiving thalidomide. In one controlled study in HIV-infected patients, median plasma HIV-1 RNA levels increased by 0.42 log10 copies/mL in patients receiving thalidomide compared with a 0.05 log10-copies/mL increase in those receiving placebo.1,12 A similar trend of increased HIV load was reported in another study in HIV-infected patients.1 However, the clinical importance of these effects is unknown since the patients involved were not receiving the potent antiretroviral agent combination regimens currently recommended for HIV-infected patients.1,12,54
Plasma HIV-1 RNA levels should be measured after the first and third months of thalidomide treatment and every 3 months thereafter.1
Bradycardia, sometimes requiring medical intervention, has been reported in patients receiving thalidomide; however, the underlying etiology and clinical significance of this finding is unknown.1
Patients should be monitored for bradycardia and syncope.1 If bradycardia occurs, dosage reduction or discontinuance of therapy may be required.1 Concomitant medications that lower heart rate should be used with caution.1
Severe cases of cutaneous reactions (i.e., Stevens-Johnson syndrome [SJS], toxic epidermal necrolysis [TEN], drug reaction with eosinophilia and systemic symptoms [DRESS]) have been reported.1 DRESS may present with a cutaneous reaction (such as rash or exfoliative dermatitis), eosinophilia, fever, and/or lymphadenopathy with systemic complications such as hepatitis, nephritis, pneumonitis, myocarditis, and/or pericarditis.1 These reactions can be fatal.1 For grade 2 or 3 skin rash, temporary interruption or discontinuance of thalidomide should be considered.1 If grade 4 rash, exfoliative or bullous rash, or other severe cutaneous reactions (SJS, TEN, or DRESS) occur, thalidomide should be permanently discontinued.1
Seizures, including tonic-clonic (grand mal) seizures, have been reported during postmarketing experience with thalidomide. Because the reports of seizures in patients receiving thalidomide were submitted voluntarily from a population of unknown size, their frequency cannot be estimated.1 Most of the patients in whom seizures were reported had disorders that may have predisposed them to seizure activity, and it currently is not known whether thalidomide has any epileptogenic activity.1
Patients with a history of seizures or other risk factors for the development of seizures should be monitored closely for clinical changes that could precipitate acute seizure activity.1
Tumor lysis syndrome has been reported during postmarketing experience with thalidomide.1 Patients at risk of tumor lysis syndrome (e.g., high tumor burden at baseline) should be monitored for tumor lysis syndrome, and appropriate precautions should be instituted.1
Thalidomide is contraindicated in patients who are hypersensitive to the drug or any ingredient in the formulation.1 The drug should be discontinued if signs and symptoms of hypersensitivity (e.g., erythematous macular rash associated with fever, tachycardia, hypotension) occur and are severe; if thalidomide therapy is resumed and the reaction recurs, thalidomide should be permanently discontinued.1 Thalidomide therapy should not be resumed if angioedema or anaphylaxis occurs.1 Because hypersensitivity reactions characterized by cutaneous and/or febrile manifestations have been reported in HIV-infected patients receiving thalidomide and because rechallenge with the drug resulted in accelerated hypersensitivity (including hypotension) in some of these patients,58 some clinicians recommend that if thalidomide rechallenge is unavoidable in an HIV-infected patient (e.g., to elucidate a drug reaction in a patient receiving a complex drug regimen), the patient should be hospitalized when the drug is administered and for at least 24 hours following the rechallenge.58
Hypersensitivity reactions characterized by cutaneous and/or febrile symptoms have been reported in HIV-infected patients receiving thalidomide.58 In some of these HIV-infected patients, rechallenge with thalidomide resulted in accelerated hypersensitivity, including hypotension.58 Therefore, some clinicians state that if thalidomide rechallenge in an HIV-infected patient is unavoidable (e.g., to elucidate a drug reaction in a patient receiving a complex drug regimen), the patient should be hospitalized when the drug is administered and for at least 24 hours following the rechallenge.58
Thalidomide is a human teratogen and is contraindicated for use during pregnancy.1 (See REMS under Dosage and Administration and also see Fetal/Neonatal Morbidity and Mortality under Cautions.)
Females or Males of Reproductive Potential
Results of animal studies suggest that thalidomide may impair male fertility.1
Pregnancy must be excluded prior to treatment initiation and throughout therapy.1,22 Pregnancy must be prevented for ≥4 weeks prior to and during therapy and for 4 weeks after completion of therapy.1 (See Fetal/Neonatal Morbidity and Mortality under Cautions.)
It is not known whether thalidomide is distributed into human milk.1 Because many drugs are distributed into milk, and because of the potential for serious thalidomide-related adverse effects in nursing infants, a decision should be made whether to discontinue nursing or the drug, taking into account the importance of the drug to the mother.1
Although safety and efficacy of thalidomide in pediatric patients younger than 12 years of age have not been established,1 the drug has been used in a limited number of children 6 months to 12 years of age.75,77,119,149,151,159,210 When thalidomide was used for the treatment of graft-versus-host disease in a limited number of bone marrow transplant recipients 1.5-17 years of age, adverse effects of the drug included somnolence (which required discontinuance of the drug in at least one child), constipation, and a syndrome of rash, eosinophilia, and pancreatitis (which resolved after the drug was discontinued).75
When thalidomide was used for the treatment of Crohn disease in a limited number of children and adolescents 2-18 years of age, the most frequent severe adverse effect was peripheral neuropathy.260
In clinical trials evaluating thalidomide in combination with dexamethasone, 52% of patients were 65 years of age or older and 15% were 75 years of age or older.1 Atrial fibrillation, constipation, fatigue, nausea, hypokalemia, thromboembolism, hyperglycemia, and asthenia occurred at a higher incidence compared with younger adults.1
Thalidomide has not been studied in patients with hepatic impairment.1
Renal impairment is not expected to affect systemic exposure of thalidomide since less than 3.5% of the dose is excreted in urine as unchanged drug.1
When dialysis was performed 10-15 hours after administration of thalidomide, no significant difference in pharmacokinetics of thalidomide was observed on non-dialysis or dialysis days.245
Adverse effects reported in at least 20% of patients with multiple myeloma receiving thalidomide include fatigue, hypocalcemia, edema, constipation, sensory or motor neuropathy, dyspnea, muscle weakness, leukopenia, neutropenia, rash/desquamation, confusion, anorexia, nausea, anxiety/agitation, asthenia, tremor, fever, weight loss, thromboembolism, weight gain, dizziness, and dry skin.1
Adverse effects reported in at least 10% of patients with erythema nodosum leprosum (ENL) receiving thalidomide include somnolence, rash, and headache.1
In vitro, thalidomide does not inhibit or induce cytochrome P-450 (CYP) isoenzymes at clinically relevant concentrations.1 Thalidomide is not a substrate of CYP isoenzymes.1
Drugs Affecting or Metabolized by Hepatic Microsomal Enzymes
Drug interactions with thalidomide unlikely.1
Hormonal Contraceptives and Drugs that Interfere with Hormonal Contraceptives
Concomitant administration of thalidomide and oral contraceptives containing ethinyl estradiol and norethindrone does not appear to affect the pharmacokinetics of the hormones.1,78,79 In 2 studies in healthy women 21-45 years of age who received a 3-week regimen of thalidomide (200 mg of thalidomide daily) followed by a single oral dose of an estrogen-progestin combination oral contraceptive (70 mcg of ethinyl estradiol and 2 mg of norethindrone), the pharmacokinetics of both ethinyl estradiol and norethindrone were the same as those obtained in these women when the contraceptive was administered without thalidomide.78,79
While it is unlikely that thalidomide would affect efficacy of oral contraceptives since the drug does not appear to affect the pharmacokinetics of the hormones,79,186 other drugs that the patient may be receiving, including HIV protease inhibitors, griseofulvin, rifampin, rifabutin, phenytoin, carbamazepine, or herbal supplements such as St. John's wort, may reduce efficacy of oral contraceptives up to 1 month after discontinuance of these agents.1,25,26 Because it is mandatory that females of reproductive potential receiving thalidomide use effective contraceptive measures for at least 4 weeks prior to, throughout, and for at least 4 weeks after completion of thalidomide therapy, females of reproductive potential receiving thalidomide concomitantly with any other drug that can reduce efficacy of oral contraceptives must use 2 other effective methods of contraception or abstain from heterosexual sexual contact.1 (See Fetal/Neonatal Morbidity and Mortality under Cautions.)
The risk of thromboembolism may be increased in patients receiving estrogen-containing therapy; therefore, estrogen-containing therapy should be used with caution in patients with multiple myeloma receiving thalidomide in combination with dexamethasone.1
Thalidomide has been reported to enhance the sedative effects of some drugs, including opioids, antihistamines, antipsychotics, anti-anxiety agents, or other CNS depressants, and may potentiate the somnolence caused by alcohol.1
Drugs Associated with Bradycardia
The risk of bradycardia may be increased in patients receiving thalidomide concomitantly with drugs known to cause bradycardia (e.g., calcium-channel blocking agents, α- or β-adrenergic blocking agents, digoxin, histamine H2-receptor antagonists, lithium, tricyclic antidepressants, neuromuscular blocking agents).1
Drugs Associated with Peripheral Neuropathy
Because of the potential for additive effects, drugs known to be associated with peripheral neuropathy (e.g., bortezomib, amiodarone, cisplatin, docetaxel, paclitaxel, vincristine, disulfiram, phenytoin, metronidazole, alcohol) should be used with caution in patients receiving thalidomide.1
Drugs Associated with an Increased Risk of Thromboembolism
The risk of thrombosis may be increased in multiple myeloma patients receiving thalidomide and dexamethasone concomitantly with erythropoietic stimulating agents, estrogen-containing therapies, or other agents that increase the risk of thrombosis.1
In 16 healthy men, the pharmacokinetic profile of a single 0.5-mg dose of digoxin was similar with and without coadministration of thalidomide 200 mg/day at steady state levels.1 A single dose of digoxin also had no effect on the pharmacokinetic profile of thalidomide.1 The safety of long-term concomitant use of thalidomide and digoxin has not been evaluated.1
Following coadministration of a single 25-mg dose of warfarin and thalidomide 200 mg daily at steady-state, the pharmacokinetic profile of warfarin, international normalized ratio (INR), and prothrombin time were similar to those observed without coadministration of thalidomide.1 A single dose of warfarin had no effect on the pharmacokinetic profile of thalidomide.1
Thalidomide has immunomodulatory, anti-inflammatory, and antiangiogenic activities.1,2,3,4,7,12,53,90,112,164,167,232,237 The drug also has sedative and hypnotic effects.4,112 Thalidomide is structurally similar to, but pharmacologically different from, glutethimide.1,3,4,5,112 Thalidomide contains a phthalimide ring and a glutarimide ring;1,4 the glutarimide ring is similar to that contained in glutethimide and may be responsible for the sedative and hypnotic effects of thalidomide.4 Because the glutarimide ring has a single asymmetric carbon (chiral center), thalidomide may exist as either an optically active levorotatory or dextrorotatory enantiomer.1,4,90,106,112 The enantiomers rapidly interconvert in vivo and in vitro;4,90,106,108,112 it is unknown whether the enantiomers have distinct pharmacologic properties.4,106 Thalidomide is commercially available as a 1:1 racemic mixture of the 2 enantiomers with a net optical rotation of zero.1,4,112
The mechanism(s) of action of the immunomodulatory and anti-inflammatory effects of thalidomide are complex and have not been fully determined.2,3,5,6,12,14,41,42,43,53,112,230 Cellular activities of thalidomide are mediated through its target cereblon, a component of a cullin ring E3 ubiquitin ligase enzyme complex.1 Data from in vitro models suggest the effects of thalidomide appear to result in part from modulation of tumor necrosis factor alpha (TNF-α) levels,1,3,7,13,14,40,41,42,44,53,90,112,230,232,237 costimulatory or adjuvant effect on T-cells resulting in increased T-cell proliferation and increased production of interleukin-2 and interferon-γ,90,190,191,194,237,246 and/or modulation of leukocyte migration and chemotaxis.1,2,6,14,44,112,248 Other anti-inflammatory and immunomodulatory effects of the drug may include suppression of macrophage involvement of prostaglandin synthesis and modulation of interleukin-10 and interleukin-12 production by peripheral blood mononuclear cells.1,244
The immunomodulatory and anti-inflammatory effects of thalidomide differ from those of other immunosuppressive agents, including corticosteroids, cyclosporin (e.g., cyclosporine) or macrolide (e.g., tacrolimus) immunosuppressants, pentoxifylline, immunosuppressive purine analogs (e.g. azathioprine) and purine metabolism inhibitors (e.g., mycophenolic acid),2 and also differ from those of nonsteroidal anti-inflammatory agents.2 Thalidomide does not appear to interfere with important host antimicrobial mechanisms; the drug has no substantial inhibitory effect on lymphocyte proliferation, does not impair delayed-type hypersensitivity reactions, and does not impair granuloma formation.2,40,190,191
Thalidomide has no direct antibacterial activity against Mycobacterium leprae .2 In addition, results of an in vitro study using Enterocytozoon bieneusi , Encephalitozoon intestinalis , and E. cuniculi indicate that thalidomide has no direct activity against microsporidia.193
Use of thalidomide in the management of multiple myeloma, erythema nodosum leprosum (ENL], and various inflammatory and/or dermatologic disorders is based on the drug's immunomodulatory effects.2,112 Thalidomide's immunomodulatory effects may result from modulation of TNF-α levels,1,3,7,13,14,40,41,42,44,53,112,232,233 costimulatory or adjuvant effect on T-cells resulting in increased T-cell proliferation and increased production of interleukin-2 and interferon-γ,90,190,191,194,244 and/or modulation of leukocyte extravascular migration mechanisms.1,2,6,14,44,112 However, in vitro studies and preliminary clinical studies indicate that the immunomodulatory effects of thalidomide vary considerably under different conditions,1,2,41,42,43,90 and some evidence indicates that effects of the drug may be species specific.6
Treatment of thalidomide in patients with multiple myeloma is accompanied by an increase in the number of circulating natural killer cells, and an increase in plasma levels of interleukin-2 and interferon-gamma (T cell derived cytokines associated with cytotoxic activity).1 In patients with multiple myeloma, thalidomide inhibits TNF-α expression by bone marrow stromal cells, resulting in inhibition of growth of multiple myeloma cells.243,248,249 Thalidomide enhances T cell activation, releasing cytokines IL-2 and interferon-γ.1,248 These cytokines activate natural killer cells causing lysis of multiple myeloma cells.1,243,248
The difficulty in characterizing thalidomide's mechanism of action has been attributed in part to numerous in vivo metabolites that result from the combination of optical enantiomerism, hydrolysis, and hydroxylation of the drug.5,6 In studies of enantiomerically stable thalidomide analogs (with an α-methyl group attached to the chiral carbon), levorotatory analogs demonstrated more immunomodulatory, sedative, and teratogenic activity, and more inhibition of TNF-α release, than dextrorotatory analogs.2 Although apparent enantiomer-specific effects of thalidomide have been reported from in vitro and in vivo studies, data from such studies are limited by rapid racemization and the pharmacokinetics of the drug in the system being studied, and should be interpreted with caution.2,90 Spontaneous hydrolysis of thalidomide yields products that do not appear to have immunomodulatory activity;2,92 the extent of hepatic metabolism of the drug appears to be limited,1,2 and some experts state that insufficient evidence exists to support speculation that formation of active metabolites is required for thalidomide's effects.2
The immunomodulatory effects of thalidomide may be related to suppression of excessive TNF-α production;1,3,7,13,14,40,41,44,45,53,112,232 however, there also is some evidence that thalidomide is capable of enhancing TNF-α synthesis.1,12,90,186 The drug selectively inhibits the production of TNF-α by cultured human monocytes (possibly by inhibiting production and/or enhancing the degradation of TNF-α messenger RNA) without influencing either general protein synthesis or the expression of other monocyte-derived cytokines (e.g., IL-1, IL-6, GM-CSF).6,7,8,11,40,112 Administration of thalidomide has been reported to decrease circulating TNF-α levels in patients with erythema nodosum leprosum (ENL).1,6,8,44,112
Wasting syndrome and the associated systemic symptoms (e.g., anorexia, fever) also may be mediated by excessive TNF-α production,8,11,40,41 and there is limited evidence that thalidomide may reduce plasma TNF-α concentrations and ameliorate wasting in patients with HIV infection and/or tuberculosis.11,40,41,42 However, the role of TNF-α in HIV infection is unclear because evidence of TNF-α production in patients with HIV infection is inconsistent and includes reports of undetected, increased, or unchanged production of the cytokine.12,14,41,42,43,47 Also, serum and plasma TNF-α levels fluctuate because of the cytokine's short half-life, and may not accurately reflect the actual extent of TNF-α production and/or the degree of TNF-α-related toxicities.42 Such discrepancies in apparent TNF-α levels may result from the different assays used,41,43 or from defects in the methods used to determine in vitro and in vivo cytokine production.41 In addition, cytokines may be produced and act locally in tissue, and it is possible that circulating levels of TNF-α do not reflect tissue activity.41,112
The effect of thalidomide on TNF-α levels, HIV replication, and HIV disease manifestations such as wasting is unclear.1,8,11,12,41,42,43 In one limited controlled study of patients with wasting syndrome associated with HIV-1 infection (with or without tuberculosis), those receiving thalidomide experienced a substantially greater weight gain than those receiving placebo;42 however, only the patients with HIV and tuberculosis experienced decreased plasma TNF-α and decreased plasma HIV-1 RNA levels while receiving thalidomide, and these patients experienced a higher mean weight gain than those with HIV infection alone.42 Data from some studies in patients with HIV infection indicate that thalidomide inhibited TNF-α activation of latent HIV-1 in monocytes and reduced replication of the virus;8,11 however, some evidence indicates that thalidomide increases plasma TNF-α and soluble TNF-α type II receptor levels in patients with HIV infection, and may increase HIV replication.1,12 In a placebo-controlled study in HIV-infected patients, thalidomide therapy (200 mg daily for 4 weeks) apparently had no inhibitory effect on HIV replication and no effect on plasma TNF-α levels, but did increase plasma levels of soluble IL-2 receptor, soluble CD8 antigen, and IL-12.194 In addition, in vitro studies using purified T-cells from these patients indicated that exposure to thalidomide resulted in a costimulatory effect resulting in increased production of IL-2 and interferon-γ.194
There is evidence that thalidomide induces down-modulation of selected cell surface adhesion molecules involved in leukocyte migration,1,2,6,14,45 and the drug's immunomodulatory mechanism of action may result in part from modulation of cellular interactions involving direct physical contact (i.e., adhesion and detachment) of leukocytes with endothelial cells.2 Limited data indicate that thalidomide may modulate TNF-α induction of endothelial cell adhesion molecules; this effect may interfere with leukocyte adhesion to endothelium, and prevent the initiation of leukocytic extravasation into inflammatory foci.2 Also, once adhesion of leukocytes to vascular endothelium occurs, thalidomide may interfere with their detachment, and impede transmigration of leukocytes into extravascular tissue.2 Although thalidomide may up- or down-modulate different adhesion receptors, and its effects on various types of leukocytes is variable,2,14 treatment with thalidomide induces a prompt reduction in the number of neutrophils and CD4+ T-cells in the lesions of patients with ENL.14,44 Leukocyte infiltration and cytokine (especially TNF-α) responses are present in focal inflammatory lesions characterized by post-capillary vasculitis (e.g., ENL lesions and mucocutaneous aphthae),2 and these immunopathologic conditions are those most clearly responsive to thalidomide.2 Such antivasculitic effects also may be enhanced by thalidomide's modulation of TNF-α synthesis or release.2 However, the exact mechanism of action for the drug's clinical immunomodulatory activity is unclear,1,2,3,5,6,12,14 and further study is required to elucidate the drug's mechanism of action.2,5,6,14
Thalidomide inhibits angiogenesis,2,3,12,164,167 and it has been suggested that the teratogenic effects of thalidomide on fetal limbs may be related to inhibition of blood vessel growth in the developing fetal limb bud.3,12,112,164,167 Thalidomide's anti-angiogenic effects have been demonstrated in several animal angiogenesis models;112,164,167 however, there is evidence that the drug's anti-angiogenic effects may be species specific and possibly may be related to a species-specific metabolite and/or metabolic activation.112,164,167 Thalidomide reduced the area of vascularization in a rabbit corneal model of induced neovascularization.112,164 The drug also inhibited angiogenesis in a rat aorta model and in human aortic endothelial cells when human or rabbit microsomes were present, but not when rat microsomes were present.112,167
The mechanism of thalidomide's anti-angiogenic effects is unknown.164 It has been suggested that inhibition of cytokine synthesis (especially that of TNF-α) may contribute to thalidomide's anti-angiogenic effect;2,112,164 however, there is some evidence from animal models that thalidomide's effect on angiogenesis is independent of the drug's effect on TNF-α and possibly may result from a direct inhibitory effect on some component of angiogenesis.112
Thalidomide has CNS depressant effects and causes sedation.4,112,232 The drug has a prompt sedative effect, and does not cause a hangover.4,232 The glutarimide ring contained in thalidomide appears to be responsible for the sedative and hypnotic effects of the drug; the ring is structurally similar to ring moieties contained in some other sedative and hypnotic drugs (e.g., glutethimide).4 Thalidomide may activate a sleep center in the forebrain, a mechanism of action unlike that of barbiturates.4 Thalidomide has little acute CNS toxicity, and does not cause incoordination or respiratory depression even at large doses.2,4,15 While thalidomide initially was investigated for use as a sedative and hypnotic in the late 1950s,2,3,4,5,15,52,53,54 the drug is no longer promoted for use as a sedative and hypnotic because of its teratogenic risk.2,3,4,5,15,16,53,54 (See Uses: Overview.)
The pharmacokinetics of thalidomide have been studied in healthy adults,1,78,79,107 adults with leprosy,1 adults with human immunodeficiency virus (HIV) infection,1,51,176 and geriatric men with prostate cancer.96 While there is some evidence that bioavailability of oral thalidomide (i.e., peak plasma concentrations, area under the plasma concentration-time curve [AUC]) may be greater in leprosy patients than in healthy individuals,1 results of a single-dose study indicate that the pharmacokinetics of thalidomide in HIV-infected individuals are similar to those in healthy individuals.1 Age-related changes in the pharmacokinetics of thalidomide have not been observed in healthy individuals,1 leprosy patients 20-69 years of age,1 or prostate cancer patients 55-80 years of age.96 The pharmacokinetics of thalidomide have not been studied to date in individuals younger than 18 years of age.1 While limited data indicate that the pharmacokinetics of thalidomide are similar in males and females, specific comparative studies have not been performed to determine whether there are any gender- or race-related differences in the pharmacokinetics of the drug.1 The pharmacokinetics of thalidomide in patients with renal (except those with end-stage renal disease) or hepatic impairment have not been determined.1
Based on studies in healthy adults78,107 and HIV-infected patients,176 the pharmacokinetics of thalidomide can best be described by a single-compartment model with first-order absorption and elimination.78,107,112,176 Results of studies in healthy adults indicate that accumulation of thalidomide does not occur, and pharmacokinetic parameters are similar following single or multiple doses of the drug.1,78,79
The absolute bioavailability of thalidomide administered as the commercially available racemic mixture of the drug has not been determined to date, in part because racemic thalidomide has poor aqueous solubility.1,2,232 The limited aqueous solubility of racemic thalidomide does not appear to be due to a lipophilic structure, since the drug has a relative distribution between lipid and aqueous phases of about 2:1.2 However, the 4 carboxyl groups of the racemic form of the drug appear to interact through hydrogen bonding to result in relative aqueous insolubility.2 In contrast, the pure levorotatory or dextrorotatory enantiomers of thalidomide are 3-5 times more soluble than the racemic drug and are more readily absorbed.2,106,108
Following oral administration of racemic thalidomide, the drug is slowly absorbed from the GI tract,1,5,51,96,107 and some interindividual variation in absorption has been reported.1,186 The relative bioavailability of thalidomide capsules compared with an oral polyethylene glycol (PEG) solution of the drug is 90%.1 When thalidomide is administered in increasing doses in healthy individuals, the extent of absorption (as measured by the AUC) increases proportionally with increasing dose; however, peak plasma concentrations of the drug increase in a less than proportional manner and the time to peak plasma concentrations is delayed, indicating that thalidomide's poor aqueous solubility affects the rate of oral absorption.1
Mean peak plasma concentrations of thalidomide generally are attained 2.5-5 hours after an oral dose.1,5,51,78,107,112,176 In a study in healthy men 21-43 years of age who received a single 200-mg dose of thalidomide given as tablets (not commercially available in the US), mean peak plasma concentrations of 1.15 mcg/mL were attained at a mean of 4.4 hours.107,112 In healthy women 21-45 years of age who received a single 200-mg dose of thalidomide given as 50- or 100-mg capsules (100-mg capsules not commercially available in the US), mean peak plasma concentrations of 2.3-3.2 mcg/mL were attained within about 6 hours.78,79 In healthy adults who received a single 50-, 200-, or 400-mg oral dose of thalidomide as 50-mg capsules, mean peak plasma concentrations of 0.62, 1.76, or 2.82 mcg/mL, respectively, were attained at 2.9, 3.5, or 4.3 hours, respectively, after the dose.1
In adults with leprosy who received a single 400-mg oral dose of thalidomide as 50-mg capsules, peak plasma concentrations of 3.44 mcg/mL were attained 5.7 hours after the dose.1
In adults with HIV infection (without active opportunistic infections or concomitant disease that potentially could alter drug absorption) who received a single 100- or 300-mg dose of thalidomide as capsules, mean peak plasma concentrations of the drug were 1.2 or 3.5 mcg/mL, respectively, and were attained at a mean of 3.4 hours after either dose.51 In another study in asymptomatic HIV-infected adults who received single 100- or 200-mg doses of the drug, peak plasma concentrations averaged 1.2 or 1.9 mcg/mL, respectively, and were attained at 2.5 or 3.3 hours, respectively.176
In men 55-80 years of age with prostate cancer who received a single 200- or 800-mg oral dose of thalidomide as 50-mg capsules, median peak plasma concentrations were 1.97 or 4.42 mcg/mL, respectively, and were attained at a median of 3.32 or 4.42 hours, respectively, after the dose.96 When these patients received multiple doses of thalidomide, mean peak plasma concentrations at steady state were 1.8 mcg/mL in those receiving 200 mg once daily and 7.57 mcg/mL in those receiving 800 mg daily.96
Information on distribution of thalidomide in humans is not available.186 Although pharmacologic and adverse effects of thalidomide appear to be organ specific, such effects do not correspond well with pharmacokinetic distribution data obtained in animals.2 While results of some animal studies indicate that high concentrations of thalidomide are found in the GI tract, liver, and kidneys, and lower concentrations are found in muscle, brain, and adipose tissue,5 other distribution studies in animals have failed to detect substantial accumulation of the drug in any particular organ.2 When radiolabeled thalidomide was administered to animals in one study, there was an even distribution of radioactivity, except for slight enhancement of radioactivity in kidneys, liver, biliary tissue, white (but not grey) matter of the CNS, and peripheral nerve trunks.2 In rabbits, thalidomide concentrations in CSF are about 50% of concurrent plasma concentrations of the drug.2 Thalidomide is present in semen; in at least one study, the drug was detectable in the semen of HIV-infected men receiving thalidomide 100 mg daily.1
Thalidomide crosses the placenta in humans and in animals.1,2,5 Malformations of human fetuses exposed to thalidomide during the first trimester of pregnancy exhibit striking organ specificity.2,15 Fetal skeletal deformities involving the loss of part or all of one or more bones are most common,1,2,3,4,5,15,21 but fetal deformities may occur in almost any organ of the body15 and defects of internal organs accompany some skeletal deformities (e.g., amelia, phocomelia).4,5 Seemingly isolated neurodevelopmental regions (e.g., the eyes, ears, and possibly some brain stem neurons innervating facial muscles) are affected in cases of thalidomide embryopathy, but other neurodevelopmental defects rarely occur, and the skin and the lymphatic organs are notably not affected by fetal exposure to thalidomide.2,15 (See Fetal/Neonatal Morbidity and Mortality under Cautions.)
The apparent volume of distribution of thalidomide has been reported to be 69.9-82.7 L in HIV-infected adults.176
Results of an in vitro study indicate that the dextrorotatory and levorotatory enantiomers of thalidomide are 55 and 66%, respectively, bound to plasma proteins.1,108
The mean elimination half-life of thalidomide following a single 200-mg oral dose ranges from 3-6.7 hours,78,79,96 and the elimination half-life appears to be similar following multiple doses of the drug.1,78,96 In a study in healthy adults who received a single 50- to 400-mg oral dose of the drug, the mean elimination half-life of thalidomide was 5.5-7.3 hours, respectively.1 The mean elimination half-life of thalidomide was 4.6-6.5 hours in HIV-infected adults who received a single 100- to 300-mg dose.51,176
Numerous metabolites may be formed as a result of optical enantiomerism and hydrolysis and hydroxylation of the drug.2,5,6,105,106,108,232 Commercially available thalidomide is a 1:1 racemic mixture of the dextrorotatory and levorotatory enantiomers of the drug,4,112 and chiral inversion and spontaneous hydrolysis of the enantiomers occurs in vivo or in vitro.4,105,106,107,108 The half-life for racemization of an enantiomer in whole blood is about 2.25 hours.2,5,6 In vivo, chiral inversion appears to occur principally in the circulation and in albumin-rich extravascular sites.108 Chiral inversion and hydrolysis of thalidomide apparently occur by several different mechanisms.108
Hepatic metabolism of thalidomide is limited,1,4 and only the parent compound appears to be metabolized by cytochrome P-450 (CYP) isoenzymes.4
The principal metabolic pathway of thalidomide appears to be nonenzymatic spontaneous hydrolysis.1,2,4,6,108,112,232,245 All 4 amide bonds in thalidomide are susceptible to hydrolytic cleavage and, under physiologic conditions, ring opening occurs first in the phthalimide moiety.2,4 Spontaneous hydrolysis to form more than 10 metabolites occurs rapidly in vitro, with a mean half-life (dependent on incubation temperature and pH) of 2-5 hours for both optical isomers.2,6 There is evidence that thalidomide metabolites formed by spontaneous hydrolysis in vitro have no immunomodulatory activity;2,92 however, further study is required to characterize the pharmacologic activity, if any, of metabolites formed in vivo.2,5
Hydroxylation can occur at 4 sites in thalidomide (i.e., 2 sites in each of the phthalimide and glutarimide moieties).2 Metabolism and some pharmacologic effects of thalidomide may be species specific.2,46 Hydroxylated metabolites have been identified in species sensitive to the teratogenic effects of thalidomide (e.g., rabbit),2,6 and there is evidence that rat (a species not sensitive to teratogenic effects of the drug) microsomes are not able to produce a metabolite that is toxic to lymphocytes in other species.6,46 Intermediary forms (i.e., arene oxides or epoxides) proposed to be responsible for the teratogenic effects of the drug have not been demonstrated, hydroxylated metabolites resulting from epoxide activation have not been confirmed in human samples, and in vitro human microsomal preparations do not appear to catalyze the formation of hydroxylated metabolites.2
Following administration of a radiolabeled dose of thalidomide, 91.9% of the dose is excreted in urine, mainly as hydrolytic metabolites;1 some of the drug may be eliminated in the bile.232 Although total body clearance of the drug is about 170-207 mL/minute,4,5 unmetabolized thalidomide has a renal clearance of 1.15-1.38 mL/minute1,5 and less than 3.5% of the drug is excreted in urine as unchanged drug.1,5
Additional Information
Overview® (see Users Guide). For additional information on this drug until a more detailed monograph is developed and published, the manufacturer's labeling should be consulted. It is essential that the manufacturer's labeling be consulted for more detailed information on usual cautions, precautions, contraindications, potential drug interactions, laboratory test interferences, and acute toxicity. 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.
Because thalidomide is a known human teratogen and can cause severe, life-threatening birth defects if administered during pregnancy, distribution of thalidomide is restricted.1 (See REMS under Dosage and Administration.)
Routes | Dosage Forms | Strengths | Brand Names | Manufacturer |
---|---|---|---|---|
Oral | Capsules | 50 mg | Bristol Myers Squibb | |
100 mg | Thalomid® | Bristol Myers Squibb | ||
150 mg | Thalomid® | Bristol Myers Squibb | ||
200 mg | Thalomid® | Bristol Myers Squibb |
1. Bristol Myers Squibb. Thalomid® (thalidomide) capsules prescribing information. Princeton, NJ; 2021 Feb. [Web]
2. Zwingenberger K, Wnendt S. Immunomodulation by thalidomide: systematic review of the literature and of unpublished observations. J Inflamm . 1995-96; 46:177-211.
3. Anon. New uses of thalidomide. Med Lett Drugs Ther . 1996; 38:15-6. [PubMed 8592478]
4. Tseng S, Pak G, Washenik K et al. Rediscovering thalidomide: a review of its mechanism of action, side effects, and potential uses. J Am Acad Dermatol . 1996; 35:969-79. [PubMed 8959957]
5. Gunzler V. Thalidomide in human immunodeficiency virus (HIV) patients. A review of safety considerations. Drug Saf . 1992; 7:116-34. [PubMed 1605898]
6. Schuler U, Ehninger G. Thalidomide: rationale for renewed use in immunological disorders. Drug Saf . 1995; 12:364-9. [PubMed 8527011]
7. Sampaio, EP, Sarno EN et al. Thalidomide selectively inhibits tumor necrosis factor α production by stimulated human monocytes. J Exp Med . 1991; 173:699-703. [PubMedCentral] [PubMed 1997652]
8. Makonkawkeyoon S, Limson-Pobre RN, Moreira AL et al. Thalidomide inhibits the replication of human immunodeficiency virus type 1. Proc Natl Acad Sci USA . 1993; 90:5974-8. [PubMedCentral][PubMed 8327469]
10. Gelber RH. Leprosy (Hansen's disease). In: Mandell GL, Bennett JE, Dolin R, eds. Mandell, Douglas and Bennett's principles and practice of infectious diseases. 4th ed. New York, NY: Churchill Livingstone; 1995:2243-50.
11. Balog DL, Epstein ME, Amodio-Groton MI. HIV wasting syndrome: treatment update. Ann Pharmacother . 1998; 32:446-58. [PubMed 9562141]
12. Jacobson JM, Greenspan JS, Spritzler J et al. Thalidomide for the treatment of oral aphthous ulcers in patients with human immunodeficiency virus infection. N Engl J Med . 1997; 336:1487-93. [PubMed 9154767]
13. Sampaio EP, Moreira AL, Sarno EN et al. Prolonged treatment with recombinant interferon γ induces erythema nodosum leprosum in lepromatous leprosy patients. J Exp Med . 1992; 175:1729-37. [PubMedCentral] [PubMed 1588290]
14. Nogueira AC, Neubert R, Helge H et al. Thalidomide and the immune system. Simultaneous up- and down-regulation of different integrin receptors on human white blood cells. Life Sci . 1994; 55:77-92. [PubMed 7516993]
15. Smithells RW, Newman CG. Recognition of thalidomide defects. J Med Genet . 1992; 29:716-23. [PubMedCentral][PubMed 1433232]
16. Burkholz, H. Giving thalidomide a second chance. FDA Consumer . 1997; September-October:12-14.
17. Food and Drug Administration. FDA approves thalidomide for Hansen's disease side effect, imposes unprecedented restrictions on distribution. FDA Talk Paper . July 16, 1998.
18. US Food and Drug Administration. Thalomid® NDA #020785: Risk evaluation mitigation strategy. From FDA website. Accessed 2021, June 17. [Web]
20. Food and Drug Administration. Thalidomide (marketed as Thalomide®) information. 24 Jul 2015. From website ([Web]).
21. Food and Drug Administration Center for Drug Evaluation and Research. Thalidomide. Important patient information. July 17, 1998. From web site ([Web]).
22. Zeldis JB, Williams BA, Thomas SD et al. S.T.E.P.S.®: a comprehensive program for controlling and monitoring access to thalidomide. Clin Ther . 1999; 21:319-30. [PubMed 10211535]
23. Anon. Celgene Thalomid initial availability will be limited to 500 physicians. F-D-C Rep . 1998; Jul 20:3-4.
24. Stewart F. Promoting emergency contraception. Hosp Pract . 1998; (Aug 15):61-75.
25. Oral contraceptives interaction: ritonavir (Norvir). In: Hansten PD, Horn JR. Drug interactions analysis and management. Vancouver, WA: Applied Therapeutics, Inc; 1997: N.32.
26. Ethinyl estradiol (Ortho-Novum 1/35) drug interaction: Indinavir (Crixivan). In: Hansten PD, Horn JR. Drug interactions analysis and management. Vancouver, WA: Applied Therapeutics, Inc; 1997: N.49.
28. Food and Drug Administration. Search orphan drug designations and approvals. From FDA website. Accessed 2021 Jun 22. [Web]
29. Kaba SE, Kyritis AP. Recognition and management of gliomas. Drugs . 1997; 53:235-44. [PubMed 9028743]
30. Soler RA, Howard M, Brink NS et al. Regression of AIDS-related Kaposi's sarcoma during therapy with thalidomide. Clin Infect Dis . 1996; 23:501-3. [PubMed 8879772]
32. Gasparini G, Harris AL. Clinical importance of the determination of tumor angiogenesis in breast carcinoma: much more than a new prognostic tool. J Clin Oncol . 1995; 13:765-82. [PubMed 7533829]
34. Iyer CG, Languillon J, Ramanujam K et al. WHO co-ordinated short-term double-blind trial with thalidomide in the treatment of acute lepra reactions in male lepromatous patients. Bull World Health Organ . 1971; 45:719-32. [PubMedCentral][PubMed 4947831]
35. Waters MF. An internally-controlled double blind trial of thalidomide in severe erythema nodosum leprosum. Lepr Rev . 1971; 42:26-42. [PubMed 4338720]
38. Smithells D. Does thalidomide cause second generation birth defects? Drug Saf . 1998; 19:339-41.
39. Sheskin J, Convit J. Results of a double blind study of the influence of thalidomide on the lepra reaction. Int J Lepr Other Mycobact Dis . 1969; 37:135-46. [PubMed 4900407]
40. Tramontana JM, Utaipat U, Molloy A et al. Thalidomide treatment reduces tumor necrosis factor α production and enhances weight gain in patients with pulmonary tuberculosis. Mol Med . 1995; 1:384-97. [PubMedCentral] [PubMed 8521296]
41. Reyes-Teran G, Sierra-Madero JG, Martinez del Cerro V et al. Effects of thalidomide on HIV-associated wasting syndrome: a randomized, double-blind, placebo-controlled clinical trial. AIDS . 1996; 10:1501-7. [PubMed 8931784]
42. Klausner JD, Makonkawkeyoon S, Akarasewi P et al. The effect of thalidomide on the pathogenesis of human immunodeficiency virus type 1 and M. tuberculosis infection. J Acquir Immune Defic Syndr Hum Retrovirol . 1996; 11:247-57. [PubMed 8603261]
43. Aukrust P, Liabakk NB, Muller F et al. Serum levels of tumor necrosis factor-α (TNFα) and soluble TNF receptors in human immunodeficiency virus type 1 infectioncorrelations to clinical, immunologic, and virologic parameters. J Infect Dis . 1994; 169:420-4. [PubMed 7906293]
44. Sampaio EP, Kaplan G, Miranda A et al. The influence of thalidomide on the clinical and immunologic manifestation of erythema nodosum leprosum. J Infect Dis . 1993; 168:408-14. [PubMed 8335978]
45. Sarno EN, Grau GE, Vieira LM et al. Serum levels of tumour necrosis factor-alpha and interleukin 1β during leprosy reactional states. Clin Exp Immunol . 1991; 84:103-8. [PubMedCentral] [PubMed 2015700]
46. Gordon GB, Spielberg SP, Blake DA et al. Thalidomide teratogenesis: evidence for a toxic arene oxide metabolite. Proc Natl Acad Sci USA . 1981; 78:2545-8. [PubMedCentral][PubMed 6941308]
47. Munis JR, Richman DD, Kornbluth RS. Human immunodeficiency virus-1 infection of macrophages in vitro neither induces tumor necrosis factor (TNF)/cachectin gene expression nor alters TNF/cachectin induction by lipopolysaccharide. J Clin Invest . 1990; 85:591-6. [PubMedCentral][PubMed 2298923]
49. Hamuryudan V, Mat C, Saip S et al. Thalidomide in the treatment of the mucocutaneous lesions of the Behcet syndrome. Ann Intern Med . 1998; 128:443-50. [PubMed 9499327]
50. Sharp M, Getty J, Klausner JD. Thalidomide use is associated with weight gain in HIV-1 positive clients. J Acquir Immune Defic Syndr . 1997; 15:392.
51. Piscitelli SC, Figg WD, Hahn B et al. Single-dose pharmacokinetics of thalidomide in human immunodeficiency virus-infected patients. Antimicrob Agents Chemother . 1997; 41:2797-9. [PubMedCentral][PubMed 9420064]
52. Vanchieri C. Preparing for thalidomide's comeback. Ann Intern Med . 1997; 127:951-2. [PubMed 9446509]
53. Anon. Thalidomide. Med Lett Drugs Ther . 1998; 40:103-4. [PubMed 9813597]
54. Nightingale SL. From the Food and Drug Administration. Thalidomide approved for erythema nodosum leprosum. JAMA . 1998; 280:872. [PubMed 9739956]
55. Weidle PJ. Thalidomide for aphthous ulcers in patients infected with the human immunodeficiency virus. Am J Health-Syst Pharm . 1996; 53:368,371-2,378. [PubMed 8673657]
56. Sharpstone D, Rowbottom A, Francis N et al. Thalidomide: a novel therapy for microsporidiosis. Gastroenterology . 1997; 112:1823-9. [PubMed 9178672]
57. Gagnon B, Bruera E. A review of the drug treatment of cachexia associated with cancer. Drugs . 1998; 55:675-88. [PubMed 9585863]
58. Haslett P, Tramontana J, Burroughs M et al. Adverse reactions to thalidomide in patients infected with human immunodeficiency virus. Clin Infect Dis . 1997; 24:1223-7. [PubMed 9195087]
59. Clemmensen OJ, Olsen PZ, Andersen KE. Thalidomide neurotoxicity. Arch Dermatol . 1984; 120:338-41. [PubMed 6703734]
60. Wulff CH, Hoyer H, Asboe-Hansen G et al. Development of polyneuropathy during thalidomide therapy. Br J Dermatol . 1985; 112:475-80. [PubMed 2986670]
61. Ochonisky S, Verroust J, Bastuji-Garin S et al. Thalidomide neuropathy incidence and clinicoelectrophysiologic findings in 42 patients. Arch Dermatol . 1994; 130:66-9. [PubMed 8285742]
62. Aronson IK, Yu R, West DP et al. Thalidomide-induced peripheral neuropathy. Arch Dermatol . 1984; 120:1466-70. [PubMed 6093713]
63. Jacobus Pharmaceutical Company Inc. Dapsone USP prescribing information. Princeton, NJ; 1985 Jun.
64. Jacobson RR. Treatment. In: Hastings RC, ed. Leprosy. New York: Churchill Livingstone; 1985:193-222.
65. Geigy Pharmaceuticals. Lamprene (clofazimine) prescribing information. Ardsley, NY; 1986 Dec.
66. Jolliffe DS. Leprosy reactional states and their treatment. Br J Dermatol . 1977; 97:345-51. [PubMed 336075]
67. Beeching NJ, Ellis CJ. Leprosy and its chemotherapy. J Antimicrob Chemother . 1982; 10:81-7. [PubMed 6749788]
69. Yawalkar SJ, Vischer W. Lamprene (clofazimine) in leprosy. Lepr Rev . 1979; 50:135-44. [PubMed 396428]
74. National Hansen's Disease (Leprosy) Program Caring and Curing Since 1984. From Health Resources and Services Administration website. 2021 June.. [Web]
75. Mehta P, Kedar A, Graham-Pole S et al. Thalidomide in children undergoing bone marrow transplantation: series at a single institution and review of the literature. Pediatrics . 1999; 103:e44. [PubMedCentral]
76. Jacobson RR, Krahenbuhl JL. Leprosy. Lancet . 1999; 353:655-60. [PubMed 10030346]
77. Shek LP, Lee YS, Lee BW et al. Thalidomide responsiveness in an infant with Behcet's syndrome. Pediatrics . 1999; 103:1295-7. [PubMed 10353947]
78. Scheffler MR, Colburn W, Kook KA et al. Thalidomide does not alter estrogen-progesterone hormone single dose pharmacokinetics. Clin Pharmacol Ther . 1999; 65:483-90. [PubMed 10340913]
79. Trapnell CB, Donahue SR, Collins JM et al. Thalidomide does not alter the pharmacokinetics of ethinyl estradiol and norethindrone. Clin Pharmacol Ther . 1998; 64:597-602. [PubMed 9871424]
81. Sugumaran DS. Leprosy reactionscomplications of steroid therapy. Int J Lepr Other Mycobact Dis . 1998; 66:10-5. [PubMed 9614834]
82. Schreuder PAM. The occurrence of reactions and impairments in leprosy: experience in the leprosy control program of three provinces in Northeastern Thailand, 1978-1995. II. Reactions. Int J Lepr Other Mycobact Dis . 1998; 66:159-69. [PubMed 9728448]
83. Partida-Sanchez S, Favila-Castillo L, Pedraza-Sanchez S et al. IgG antibody subclasses, tumor necrosis factor and IFN-γ levels in patients with type II lepra reaction on thalidomide treatment. Int Arch Allergy Immunol . 1998; 116:60-6. [PubMed 9623511]
84. Bhargava P, Mal Kuldeep C, Mathur NK. Erythema nodosum leprosum in subgroups of lepromatous leprosy. Lepr Rev . 1998; 68:373-85.
85. de Carsalade GY, Wallach D, Spindler E et al. Daily multidrug therapy for leprosy; results of a fourteen-year experience. Int J Lepr Other Mycobact Dis . 1997; 65:37-44. [PubMed 9207752]
86. Meyerson MS. Erythema nodosum leprosum. Int J Dermatol . 1996; 35:389-92. [PubMed 8737869]
88. Willcox ML. The impact of multiple drug therapy on leprosy disabilities. Lepr Rev . 1997; 68:350-66. [PubMed 9503872]
89. Lockwood DNJ. The management of erythema nodosum leprosum: current and future options. Lepr Rev . 1996; 67:253-9. [PubMed 9033196]
90. Shannon EJ, Morales MJ, Sandoval F. Immunomodulatory assays to study structureactivity relationships of thalidomide. Immunopharmacology . 1997; 35:203-12. [PubMed 9043933]
92. Shannon EJ, Sandoval F, Krahenbuhl JL. Hydrolysis of thalidomide abrogates its ability to enhance mononuclear cell synthesis of IL-2 as well as its ability to suppress the synthesis of TNF-α. Immunopharmacology . 1997; 36:9-15. [PubMed 9129992]
93. Ghate JV, Jorizzo JL. Behcet's disease and complex aphthosis. J Am Acad Dermatol . 1999; 40:1-18. [PubMed 9922007]
94. Herranz P, Pizarro A, de Lucas R et al. Treatment of AIDS-associated prurigo nodularis with thalidomide. Clin Exp Dermatol . 1998; 23:233-4. [PubMed 10233821]
95. Weinstein TA, Sciubba JJ, Levine J. Thalidomide for the treatment of oral aphthous ulcers in Crohn's disease. J Ped Gastroenterol Nutr . 1999; 28:214-6.
96. Figg WD, Raje S, Bauer KS et al. Pharmacokinetics of thalidomide in an elderly prostate cancer population. J Pharm Sci . 1999; 88:121-5. [PubMed 9874712]
97. Wolkenstein P, Latarjet J, Roujeau JC et al. Randomised comparison of thalidomide versus placebo in toxic epidermal necrolysis. Lancet . 1998; 352:1586-9. [PubMed 9843104]
98. Breban M, Gombert B, Amor B et al. Efficacy of thalidomide in the treatment of refractory ankylosing spondylitis. Arthritis Rheumatism . 1999; 42:580-1. [PubMed 10088786]
99. Rousseau L, Beylot-Barry M, Doutre MS et al. Cutaneous sarcoidosis successfully treated with low doses of thalidomide. Arch Dermatol . 1998; 134:1045-6. [PubMed 9722748]
100. Berenson JR, Bergsagel PL, Munshi N. Initiation and maintenance of multiple myeloma. Semin Hematol . 1999; 36:9-13. [PubMed 9989483]
101. Forsyth CJ, Cremer PD, Torzillo P. Thalidomide responsive chronic pulmonary GVHD. Bone Marrow Transplant . 1996; 17:291-3. [PubMed 8640183]
102. Birnkrant D. Thalidomide for aphthous ulcers in HIV infection. N Engl J Med . 1997; 337:1086-7. [PubMed 9324641]
104. Stirling DI. Thalidomide and its impact in dermatology. Sem Cutan Med Surg . 1998; 17:231-42.
105. Eriksson T, Bjorkman S, Roth B et al. Hydroxylated metabolites of thalidomide: formation in-vitro and in-vivo in man. J Pharm Pharmacol . 1998; 50:1409-16. [PubMed 10052858]
106. Eriksson T, Bjorkman S, Roth B et al. Stereospecific determination, chiral inversion in vitro and pharmacokinetics in humans of the enantiomers of thalidomide. Chirality . 1995; 7:44-52. [PubMed 7702998]
107. Chen TL, Vogelsang GB, Petty BG et al. Plasma pharmacokinetics and urinary excretion of thalidomide after oral dosing in healthy male volunteers. Drug Metab Dispos . 1989; 17:402-5. [PubMed 2571480]
108. Eriksson T, Bjorkman S, Roth B et al. Enantiomers of thalidomide: blood distribution and the influence of serum albumin on chiral inversion and hydrolysis. Chirality . 1998; 10:223-8. [PubMed 9499573]
109. Heney D, Norfolk DR, Wheeldon J et al. Thalidomide treatment for chronic graft-versus-host disease. Br J Haematol . 1991; 78:23-7. [PubMed 2043478]
110. Lagueny A, Rommel A, Vignolly B et al. Thalidomide neuropathy: an electrophysiologic study. Muscle Nerve . 1986; 9:837-44. [PubMed 3023998]
111. WHO Expert Committee on Leprosy. WHO expert committee on Leprosy. World Health Organ Tech Rep Ser . 1998; 874:1-43. [PubMed 9627517]
112. Calabrese L, Resztak K. Thalidomide revisited: pharmacology and clinical applications. Exp Opin Invest Drugs. 1998; 7:2043.30.
113. Hastings RC, Trautman JR, Enna CD et al. Thalidomide in the treatment of erythema nodosum leprosum with a note on selected laboratory abnormalities in erythema nodosum leprosum. Clin Pharmacol Ther . 1970; 11:481-7. [PubMed 4913866]
114. Pearson JMH, Vedagiri M. Treatment of moderately severe erythema nodosum leprosum with thalidomidea double-blind controlled trial. Lepr Rev . 1969; 40:111-6. [PubMed 4893253]
115. National Institutes of Health, Food and Drug Administration, Centers for Disease Control and Prevention. Thalidomide: potential benefits and risks. Open public scientific workshop. Bethesda, MD: 1997 Sep 9-10.
116. Fullerton PM, O'sullivan DJ. Thalidomide neuropathy: a clinical, electrophysiological, and histological follow-up study. J Neurol, Neurosurg, Psy . 1968; 31:543.
117. Winkelmann RK, Connolly SM, Doyle JA et al. Thalidomide treatment of prurigo nodularis. Acta Derm Venereol . 1984; 64:412-7. [PubMed 6208720]
118. van den Broek H. Treatment of prurigo nodularis with thalidomide. Arch Dermatol . 1980; 116:571-2. [PubMed 7377791]
119. Lovell CR, Hawk JLM, Calnan CD et al. Thalidomide in actinic prurigo. Br J Dermatol . 1983; 108:467-71. [PubMed 6838772]
121. Stevens RJ, Andujar C, Edwards CJ et al. Thalidomide in the treatment of the cutaneous manifestations of lupus erythematosus: experience in sixteen consecutive patients. Br J Rheumatol . 1997; 36:353-9. [PubMed 9133968]
122. Naafs B, Faber WR. Thalidomide therapy: an open trial. Int J Derm . 1985; 24:131-4. [PubMed 2985516]
123. Holm AL, Bowers KE, McMeekin TO et al. Chronic cutaneous lupus erythematosus treated with thalidomide. Arch Dermatol . 1993; 129:1548-50. [PubMed 8250576]
124. Atra E, Sato EI. Treatment of the cutaneous lesions of systemic lupus erythematosus with thalidomide. Clin Exp Rheumatol . 1993; 11:487-93. [PubMed 8275583]
125. Munro CS, Cox NH. Pyoderma gangrenosum associated with Behcet's syndromeresponse to thalidomide. Clin Exp Dermatol . 1988; 13:408-10. [PubMed 3256462]
126. Rustin MHA, Gilkes JJH, Robinson JWE. Pyoderma gangrenosum associated with Behcet's disease: treatment with thalidomide. J Am Acad Dermatol . 1990; 23:941-4. [PubMed 2254485]
127. Dereure O, Basset-Sequin N, Guilhou JJ et al. Erosive lichen planus: dramatic results to thalidomide. Arch Dermatol . 1996; 132:1392-3. [PubMed 8915327]
128. Hamza MH. Treatment of Behcet's disease with thalidomide. Clin Rheumatol . 1986; 5:365-71. [PubMed 3780143]
129. Jorizzo JL, Schmalstieg FC, Solomon AR et al. Thalidomide effects in Behcet's syndrome and pustular vasculitis. Arch Intern Med . 1986; 146:878-81. [PubMed 3963978]
130. Eisenbud L, Horowitz I, Kay B et al. Recurrent aphthous stomatitis of the Behcet's syndrome: successful treatment with thalidomide. Oral Surg Oral Med Pathol . 1987; 64:289-2.
131. Mangelsdorf HC, White WL, Jorizzo JL. Behcet's disease: report of twenty-five patients from the United States with prominent mucocutaneous involvement. J Am Acad Dermatol . 1996; 34:745-50. [PubMed 8632067]
132. Ramselaar CG, Boone RM, Kluin-Nelemans HC. Thalidomide in the treatment of neuro-Behcet's syndrome. Br J Dermatol . 1986; 115:367-70. [PubMed 3756093]
133. Larsson H. Treatment of severe colitis in Behcet's syndrome with thalidomide (CG-217). J Intern Med . 1990; 228:405-7. [PubMed 2266351]
134. Lewis DA, Amerasinghe CN, Murphy SM. Successful treatment of Behcet's syndrome in an African female patient with thalidomide. Int J STD AIDS . 1996; 7:518-20. [PubMed 9116070]
135. Powell RJ, Allen BR, Jenkins JS et al. Investigation and treatment of orogenital ulceration; studies on a possible mode of action of thalidomide. Br J Dermatol . 1985; 113(Suppl 28):141-4. [PubMed 4040386]
136. Postema PTE, den Haan P, van Hagen PM et al. Treatment of colitis in Behcet's disease with thalidomide. Eur J Gastroenterol Hepatol . 1996; 8:929-31. [PubMed 8889464]
137. Saylan T, Saltik I. Thalidomide in the treatment of Behcet's syndrome. Arch Dermatol . 1982; 118:536. [PubMed 7103519]
138. Satnis HR. Aphthous stomatitis and its management. Curr Opin Dent . 1991; 1:763-8. [PubMed 1807480]
139. Bonnethlane JM, Royer C, Bedane C. Thalidomide and recurrent aphthous stomatitis: a follow-up study. Dermatology . 1996; 193:321-3. [PubMed 8993957]
140. Haslett P, Hempstead M, Seidman C et al. The metabolic and immunologic effects of short-term thalidomide treatment of patients infected with the human immunodeficiency virus. AIDS Res Hum Retroviruses . 1997; 13:1047-54. [PubMed 9264292]
141. Kaplan G, Schambelan M, Gottlieb M et al. Thalidomide reverses cachexia in HIV-wasting syndrome. 5th Conference on Retroviruses and Opportunistic Infections. 1998. Abstract No. 476.
142. Nicolau DP, West TE. Thalidomide: treatment of severe recurrent aphthous stomatitis in patients with AIDS. DICP . 1990; 24:1054-6. [PubMed 2275226]
143. Beliomo A, Schorr-Lesnick B. Thalidomide treatment for idiopathic esophageal ulcers in patients with HIV. Gastrointest Endosc . 1996; 44:729-1. [PubMed 8979067]
144. Ball SC, Sepkowitz KA, Jacobs JL. Thalidomide for treatment of oral aphthous ulcers in patients with human immunodeficiency virus: case report and review. Am J Gastroenterol . 1997; 92:169-70. [PubMed 8995965]
145. Ghigloiotti G, Repetto T, Farris A et al. Thalidomide: treatment of choice for aphthous ulcers in patients seropositive for human immunodeficiency virus. J Am Acad Dermatol . 1993; 28:271-2. [PubMed 8432932]
146. Naum SM, Molloy PJ, Kania RJ et al. Use of thalidomide in treatment and maintenance of idiopathic esophageal ulcers in HIV+ individuals. Dig Dis Sci . 1995; 40:1147-8. [PubMed 7729278]
147. Paterson DL, Georghiou PR, Allworth AM et al. Thalidomide as treatment of refractory aphthous ulceration related to human immunodeficiency virus infection. Clin Infect Dis . 1995; 20:250-4. [PubMed 7742424]
148. Grinspan D. Significant response of oral aphthosis to thalidomide treatment. J Am Acad Dermatol . 1985; 12:85-90. [PubMed 3980807]
149. Grinspan D, Blanco GF, Aguero S. Treatment of aphthae with thalidomide. J Am Acad Dermatol . 1989; 20:1060-3. [PubMed 2754056]
150. Revuz J, Guillaume JC, Janier M et al. Crossover study of thalidomide vs placebo in severe recurrent aphthous stomatitis. Arch Dermatol . 1990; 126:923-7. [PubMed 2193629]
151. Rovelli A, Arrigo C, Nesi F et al. The role of thalidomide int he treatment of refractory chronic graft-versus-host disease following bone marrow transplantation in children. Bone Marrow Transplant . 1998; 21:577-81. [PubMed 9543061]
152. Cole CH, Rogers PCJ, Pritchard S et al. Thalidomide in the management of chronic graft-versus-host disease in children following bone marrow transplantation. Bone Marrow Transplant . 1994; 14:937-42. [PubMed 7711671]
153. Vogelsang GB, Farmer ER, Hess AD et al. Thalidomide for the treatment of chronic graft-versus-host disease. N Engl J Med . 1992; 326:1055-8. [PubMed 1549151]
155. Parker PM, Chao N, Nademanee A et al. Thalidomide as salvage therapy for chronic graft-versus-host disease. Blood . 1995; 86:3604-9. [PubMed 7579470]
156. Stevens RJ. The place of thalidomide in the treatment of inflammatory disease. Lupus . 1996; 5:257-8. [PubMed 8869894]
157. Waters MF, Laing AB, Ambikapathy A et al. Treatment of ulcerative colitis with thalidomide. Br Med J . 1979; 1:792. [PubMedCentral][PubMed 435799]
158. Wettstein AR, Meagher AP. Thalidomide in Crohn's disease. Lancet . 1997; 350:1445-2. [PubMed 9371171]
159. Odeka EB, Miller V. Thalidomide in oral Crohn's disease refractory to conventional medical treatment. J Pediatr Gastroenterol . 1997; 25:250-1.
160. Kulkarni S, Powles R, Mehta J et al. Thalidomide in GVHDanti-GVHD effect separable from the antiangiogenesis? Blood . 1998; 92(Suppl 1):344b. Abstract No. 4482.
161. Singhal S, Mehta J, Eddlemon P et al. Marked anti-tumor effect from anti-angiogenesis (AA) therapy with thalidomide (T) in high risk refractory multiple myeloma (MM). Blood . 1998; 92(Suppl 1):318a.
162. Barlogie B, Desikan R, Munshi N et al. Single course D.T. pace anti-angiochemotherapy effects CR in plasma cell leukemia and fulminant multiple myeloma (MM). Blood . 1998; 92(Suppl 1):273b.
163. Lim SH, Maclean R. Thalidomide in IgA multiple myeloma. Blood . 1998; 92(Suppl 1):279b.
164. D'Amato RJ, Loughnan MS, Flynn E et al. Thalidomide is an inhibitor of angiogenesis. Proc Natl Acad Sci . 1994; 91:4082-5. [PubMedCentral][PubMed 7513432]
167. Bauer KS, Dixon SC, Figg WD. Inhibition of angiogenesis by thalidomide requires metabolic activation, which is species-dependent. Biochem Pharmacol . 1998; 55:1827-34. [PubMed 9714301]
168. Figg WD, Bergan R, Brawley O et al. Randomized phase II study of thalidomide in androgen-independent prostate cancer (AIPC). Proceedings of ASCO. 1997. Abstract No. 1189.
169. Fine HA, Loeffler JS, Kyritsis A et al. A phase II trial of the anti-angiogenic agent, thalidomide, in patients with recurrent high-grad gliomas. Proceedings of ASCO. 1997. Abstract No. 1372.
170. Politi P, Reboredo G, Losso M et al. Phase I trial of thalidomide (T) in AIDS-related kaposi sarcoma (KS). Proceedings of ASCO. 1998. Abstract No. 161.
171. Long G, Vredenburgh J, Rizzieri DA et al. Pilot trial of thalidomide post-autologous peripheral blood progenitor cell transplantation (PBPC) in patients with metastatic breast cancer. Proceedings of ASCO. 1998. Abstract No. 697.
172. Watanabe S, Pituskin E, Calder K et al. Thalidomide (T) in the symptomatic treatment of cachexia in patients (pts) with terminal cancer. Proceedings of ASCO Atlanta, CA 1999. Abstract No. 180.
173. Baidas SM, Isaacs C, Crawford J et al. A phase II evaluation of thalidomide in patients with metastatic breast cancer. Proceedings of ASCO Atlanta, CA 1999. Abstract No. 475.
174. Glass J, Gruber ML, Nirenberg A et al. Phase I/II study of carboplatin and thalidomide in recurrent glioblastoma multiforme. Proceedings of ASCO Atlanta, CA 1999. Abstract No. 551.
175. Marx GM, Levi JA, Bell DR et al. A phase I/II trial of thalidomide as an antiangiogenic agent in the treatment of advanced cancer. Proceedings of ASCO Atlanta, CA 1999. Abstract No. 1751.
176. Noormohamed FH, Youle MS, Higgs CJ et al. Pharmacokinetics and hemodynamic effects of single oral doses of thalidomide in asymptomatic human immunodeficiency virus-infected subjects. AIDS Res Hum Retroviruses . 1999; 15:1047-52. [PubMed 10461824]
177. Thalidomide Victims Association of Canada. Living in a world with thalidomide: a new reality. Aug 20, 1999. From web site ([Web]).
178. Gutierrez-Rodriguez O. A promising new treatment for rheumatoid arthritis. Arthritis Rheum . 1984; 27:1118-21. [PubMed 6237660]
179. Huizinga TWJ, Dijkmans BAC, van der Velde E et al. An open study of pentoxyfylline and thalidomide as adjuvant therapy in the treatment of rheumatoid arthritis. Ann Rheum Dis . 1996; 55:833-6. [PubMedCentral][PubMed 8976641]
180. Silva SR, Viana PC, Lugon NV et al. Thalidomide for the treatment of uremic pruritus: a crossover randomized double-blind trial. Nephron . 1994; 67:270-3. [PubMed 7936015]
181. Engeser P, Klimm HD. Therapy of recurrent exudative erythema multiforme. Effectiveness of thalidomidereport of a case. Fortschr Med . 1999; 117:39-40. [PubMed 10365532]
182. Cherouati K, Claudy A, Souteyrand P et al. Treatment by thalidomide of chronic multiforme erythema: its recurrent and continuous variants. A retrospective study of 26 patients. Ann Dermatol Venereol . 1996; 123:375-7. [PubMed 8959059]
183. Thomas L, Ducros B, Secchi T et al. Successful treatment of adult's Langerhans cell histiocytosis with thalidomide. Report of two cases and literature review. Arch Dermatol . 1993; 129:1261-4. [PubMed 8215489]
184. Meunier L, Marck Y, Ribeyre C et al. Adult cutaneous Langerhans cell histiocytosis: remission with thalidomide. Br J Dermatol . 1995; 132:168. [PubMed 7756144]
186. Reviewer's comments (personal observations).
187. Welles L, Little R, Wyvill K et al. Preliminary results of a phase II study of oral thalidomide in patients with HIV infection and Kaposi's Sarcoma (KS). J Acquir Immune Defic Syndr . 1997; 14: A21.
188. Warren R, Cole G. Reports of second generation thalidomide children: media reports that thalidomide disabilities can be inherited are false! Thalidomide Victims Association of Canada. August 30, 1997. From web site, 1999 Nov 2. ([Web]).
189. Chao NJ, Parker PM, Niland JC et al. Paradoxical effect of thalidomide prophylaxis on chronic graft-vs.-host disease. Biol Blood Marrow Transplant . 1996; 2:86-92. [PubMed 9118303]
190. Haslett PAJ, Corral LG, Albert M et al. Thalidomide costimulates primary human T lymphocytes, preferentially inducing proliferation, cytokine production, and cytotoxic responses in the CD8+ subset. J Exp Med . 1998; 187:1885-92. [PubMedCentral] [PubMed 9607928]
191. Corral LG, Haslett PAJ, Muller GW et al. Differential cytokine modulation and T cell activation by two distinct classes of thalidomide analogues that are potent inhibitors of TNF-α. J Immunol . 1999; 163: 380-6. [PubMed 10384139]
192. Ashby J, Tinwell H, Callander RD et al. Thalidomide: lack of mutagenic activity across phyla and genetic endpoints. Mut Res . 1997; 396:45-64.
193. Ridoux O, Drancourt M. Lack of in vitro antimicrosporidian activity of thalidomide. Antimicrob Agents Chemother . 1999; 2305-6.
194. Haslett PAJ, Klausner JD, Makonkawkeyoon S et al. Thalidomide stimulates T cell responses and interleukin 12 production in HIV-infected patients. AIDS Res Hum Retroviruses . 1999; 15:1169-79. [PubMed 10480630]
195. Monastirli A, Georgiou S, Bolsen K et al. Treatment of porphyria cutanea tarda with oral thalidomide. Skin Pharmacol Appl Skin Physiol . 1999; 12:305-11. [PubMed 10545826]
196. Duong DJ, Spigel GT, Moxley RT et al. American experience with low-dose thalidomide therapy for severe cutaneous lupus erythematosus. Arch Dermatol . 1999; 135:1079-87. [PubMed 10490113]
197. Horowitz SB, Stirling AL. Thalidomide-induced toxic epidermal necrolysis. Pharmacotherapy . 1999; 19:1177-80. [PubMed 10512068]
198. Durie BGM, Stepan DE. Efficacy of low dose thalidomide (T) in multiple myeloma. Presented at 7th annual international multiple myeloma workshop. Stockholm, Sweden: 1999 Sep 1-5. Abstract No. O9.
199. Weber DM, Gavino M, Delasalle K et al. Thalidomide for refractory multiple myeloma. Presented at 7th annual international multiple myeloma workshop. Stockholm, Sweden: 1999 Sep 105. Abstract No. P77.
200. Singhal S, Mehta J, Desikan et al. Antitumor activity of thalidomide in refractory multiple myeloma. N Engl J Med . 1999; 341:1565-71. [PubMed 10564685]
201. Raje N, Anderson K. Thalidomide: a revival story. N Engl J Med . 1999; 341:606-9.
202. Scribano M, Prantera C. Review article: medical treatment of moderate to severe Crohn's disease. Aliment Pharmacol Ther . 2003; 17(Suppl. 2):23-30. [PubMed 12786609]
203. Podolsky DK. Inflammatory bowel disease. N Engl J Med . 2002; 347:417-29. [PubMed 12167685]
204. Vasiliauskas EA, Kam LY, Abreu-Martin MT et al. An open-label pilot study of low-dose thalidomide in chronically active, steroid-dependent Crohn's disease. Gastroenterology . 1999; 117:1278-87. [PubMed 10579968]
205. Ehrenpreis ED, Kane SV, Cohen LB et al. Thalidomide therapy for patients with refractory Crohn's disease: an open-label trial. Gastroenterology . 1999; 117:1271-7. [PubMed 10579967]
206. Sabate JM, Villarejo J, Lemann M et al. An open-label study of thalidomide for maintenance therapy in responders to infliximab in chronically active and fistulizing refractory Crohn's disease. Aliment Pharmacol Ther . 2002; 16:1117-24. [PubMed 12030953]
207. Hershfield NB.. Disappearance of Crohn's ulcers in the terminal ileum after thalidomide therapy. Can J Gastroenterol . 2004; 18:101-4. [PubMed 14997219]
208. Bousvaros A, Mueller B. Thalidomide in gastrointestinal disorders. Drugs . 2001; 61:777-87. [PubMed 11398909]
209. Bauditz J, Wedel S, Lochs H. Thalidomide reduces tumour necrosis factor α and interleukin 12 production in patients with chronic active Crohn's disease. Gut . 2002; 50:196-200. [PubMedCentral] [PubMed 11788559]
210. Yasui K, Uchida N, Akazawa Y et al. Thalidomide for treatment of intestinal involvement of juvenile-onset Behçet disease. Inflamm Bowel Dis. 2008; 14:396-400. [PubMed 17973303]
211. Majumdar S, Mockenhaupt M, Roujeau J et al. Interventions for toxic epidermal necrolysis. Cochrane Database of Syst Rev. 2002; 4:CD001435.
212. Hall VC, El-Azhary RA, Bouwhuis S et al. Dermatologic side effects of thalidomide in patients with multiple myeloma. J Am Acad Dermatol. 2003; 48:548-52.
213. Wines NY, Cooper AJ, Wines MP. Thalidomide in dermatology. Australas J Dermatol. 2002; 43:229-38. [PubMed 12423428]
214. Rajkumar SV, Gertz MA, Witzig TE. Life-threatening toxic epidermal necrolysis with thalidomide therapy for myeloma. N Engl J Med. 2000; 343:972-3. [PubMed 11012329]
215. Paghdal K, Schwartz R. Thalidomide and its dermatologic uses. Acta Dermatovenerol Croat . 2007; 15:39-44. [PubMed 17433179]
216. Shek LPC, Lim DLC. Thalidomide in Behçet disease. Biomed Pharmacother. 2002; 56:31-5. [PubMed 11908493]
217. Colagrande M, Di Ianni M, Coletti G et al. Toxic epidermal necrolysis in a patient with primary myelofibrosis receiving thalidomide therapy. Int J Hematol. 2009; 89:76-9. [PubMed 19052692]
218. Nasca MR, Micali G, Cheigh NH et al. Dermatologic and nondermatologic uses of thalidomide. Ann Pharmacother. 2003; 37:1307-20. [PubMed 12921515]
219. Wu JJ, Huang DB, Pang KR et al. Thalidomide: dermatological indications, mechanisms of action and side-effects. Br J Dermatol. 2005; 153:254-273. [PubMed 16086735]
220. Moraes M, Russo G. Thalidomide and its dermatologic uses. Am J Med Sci. 2001; 321:321-6. [PubMed 11370795]
221. Shetty. Thalidomide in the management of recurrent aphthous ulcerations in patients who are HIV-positive: a review and case reports. Spec Care Dentist. 2005; 25:236-41.
222. Letsinger JA, McCarty MA, Jorizzo JI. Complex aphthosis: a large case series with evaluation algorithm and therapeutic ladder from topicals to thalidomide. J Am Acad Dermatology. 2005; 52:500-8.
223. Shetty KM. Current role of thalidomide in HIV-positive patients with recurrent aphthous ulcerations. Gen Dent. 2007; 55:537-42. [PubMed 18050580]
224. Corcoran C, Grinspoon S. Treatment for wasting in patients with the Acquired Immunodeficiency Syndrome. N Engl J Med . 1999; 340:1740-50. [PubMed 10352167]
225. Kaplan G, Thomas S, Fierer DS et al. Thalidomide for the treatment of AIDS-associated wasting. IDS Res and Human Retroviruses . 2000; 16:1345-55.
226. M'barek LB, Fardet L, Mebazaa A et al. A retrospective analysis of thalidomide therapy in non-HIV-related Kaposi's sarcoma. Dermatology. 2007; 215:202-5.
227. Little RF, Wyvill KM, Pluda JM et al. Activity of thalidomide in AIDS-related Kaposi's sarcoma. J Clin Oncol. 2000; 18:2593-602. [PubMed 10893291]
228. Johnson L, Jarvis JN, Wilkins EGL et al. Thalidomide treatment for refractory HIV-associated colitis: a case series. Clin Infect Dis. 2008; 47:133-6. [PubMed 18494607]
229. Teo SK, Stirling DI, Zeldis JB. Thalidomide as a novel therapeutic agent: new uses for an old product. Drug Discov. 2005; 10:107-14.
230. Bariol C, Meagher AP, Vickers CR et al. Thalidomide for inflammatory bowel disease. J Gastroenterol Hepatol. 2002; 17:135-9. [PubMed 11966942]
231. Kane S, Stone LJ, Ehrenpreis E. Thalidomide as salvage therapy for patients with delayed hypersensitivity response to infliximab: a case series. J Clin Gastroenterol. 35:149-50.
232. Shanhbag PS, Viswanath V, Torsekar RG. Thalidomide: current status. Ind J Dermatol Venereol Lepro . 2006; 72:75-80.
233. Walker SL, Waters MF, Lockwood DN. The role of thalidomide in the management of erythema nodosum leprosum. Lepr Rev . 2007; 78:197-215. [PubMed 18035771]
234. Feuth M, Brandsma JW, Faber WR et al. Erythema nodosum leprosum in Nepal: a retrospective study of clinical features and response to treatment with prednisolone or thalidomide. Lepr Rev . 2008; 79:254-69. [PubMed 19009975]
235. Kaur I, Dogra S, Narang T et al. Comparative efficacy of thalidomide and prednisolone in the treatment of moderate to severe erythema nodosum leprosum; a randomized study. Australas J Dermatol . 2009; 50:181-5. [PubMed 19659979]
236. Bensinger WI. Role of autologous and allogeneic stem cell transplantation in myeloma. Leukemia . 2009; 23:442-8. [PubMedCentral][PubMed 19277049]
237. Rosenbach M, Werth VP. Dermatologic therapeutics: thalidomide. A practical guide. Dermatol Ther . 2007; 20:175-86. [PubMed 17970884]
238. Villahermosa LG, Fajardo TT, Jr., Abalos RM et al. A randomized, double-blind, double-dummy, controlled dose comparison of thalidomide for treatment of erythema nodosum leprosorum. Am J Trop Med Hyg . 2005; 72: 518 -26. [PubMed 15891124]
239. Nair V, Sharma A, Ghosh I et al. Extensive chronic graft-versus-host disease of skin successfully treated with thalidomide. J Assoc Physicians India . 2005; 53:988-90. [PubMed 16515241]
240. Fabi SG, Hill C, Witherspoon JN et al. Frequency of thromboembolic events associated with thalidomide in the non-cancer setting: a case report and review of the literature. J Drugs Dermatol . 2009; 8:765-9. [PubMed 19663116]
241. Howell E, Johnson SM. Venous thrombosis occurring after initiation of thalidomide for the treatment of cicatricial pemphigoid. J Drugs Dermatol . 2004; 3:83-5. [PubMed 14964754]
242. Rajkumar SV, Blood E, Vesole D et al. Phase III clinical trial of thalidomide plus dexamethasone compared with dexamethasone alone in newly diagnosed multiple myeloma: a clinical trial coordinated by the Eastern Cooperative Oncology Group. J Clin Oncol . 2006; 24:431-6. [PubMed 16365178]
243. Sirohi B, Powles R. Multiple myeloma. Lancet . 2004; 363:875-7. [PubMed 15031034]
244. Haslett PA, Roche P, Butlin CR et al. Effective treatment of erythema nodosum leprosum with thalidomide is associated with immune stimulation. J Infect Dis . 2005; 192:2045-53. [PubMed 16288366]
245. Eriksson T, Höglund P, Turesson I et al. Pharmacokinetics of thalidomide in patients with impaired renal function and while on and off dialysis. J Pharm Pharmacol . 2003; 55:1701-6. [PubMed 14738599]
246. Okafor MC. Thalidomide for erythema nodosum leprosum and other applications. Pharmacotherapy . 2003; 23:481-93. [PubMed 12680478]
247. Du W, Hattori Y, Hashiguchi A et al. Tumor angiogenesis in the bone marrow of mulitple myeloma patients and its alteration by thalidomide treatment. Pathol Int . 2004; 54:285-94. [PubMed 15086832]
248. Teo SK. Properties of thalidomide and its analogues: implications for anticancer therapy. AAPS J . 2005; 7:E14-9. [PubMedCentral][PubMed 16146335]
249. Richardson P, Schlossman R, Jagannath S et al. Thalidomide for patients with relapsed multiple myeloma after high-dose chemotherapy and stem-cell transplantation: results of an open-label multicenter phase 2 study of efficacy, toxicity, and biological activity. Mayo Clin Proc . 2004; 79:875-82. [PubMed 15244383]
250. Legnani P, Kornbluth A Newer therapies for inflammatory bowel disease. Curr Treat Options Gastroenterol . 2004; 7:161-7.
251. Celgene Corporation. Thalomid® (thalidomide) capsules prescribing information. Summit, NJ; 2013 Feb.
253. Palumbo A, Rajkumar SV, Dimopoulos MA et al. Prevention of thalidomide- and lenalidomide-associated thrombosis in myeloma. Leukemia . 2008; 22:414-23. [PubMed 18094721]
254. Key NS, Khorana AA, Kuderer NM et al. Venous Thromboembolism Prophylaxis and Treatment in Patients With Cancer: ASCO Clinical Practice Guideline Update. J Clin Oncol . 2020; 38:496-520. [PubMed 31381464]
255. Rajkumar SV, Rosiñol L, Hussein M et al. Multicenter, randomized, double-blind, placebo-controlled study of thalidomide plus dexamethasone compared with dexamethasone as initial therapy for newly diagnosed multiple myeloma. J Clin Oncol . 2008; 26:2171-7. [PubMedCentral][PubMed 18362366]
256. Chasset F, Tounsi T, Cesbron E et al. Efficacy and tolerance profile of thalidomide in cutaneous lupus erythematosus: A systematic review and meta-analysis. J Am Acad Dermatol . 2018; 78:342-350.e4. [PubMed 28989111]
257. Hatemi I, Hatemi G, Pamuk ON et al. TNF-alpha antagonists and thalidomide for the management of gastrointestinal Behçet's syndrome refractory to the conventional treatment modalities: a case series and review of the literature. Clin Exp Rheumatol . 2015; 33:S129-37. [PubMed 26486925]
258. Hatemi G, Christensen R, Bang D et al. 2018 update of the EULAR recommendations for the management of Behçet's syndrome. Ann Rheum Dis . 2018; 77:808-818. [PubMed 29625968]
259. Zeng Q, Shi X, Yang J et al. The efficacy and safety of thalidomide on the recurrence interval of continuous recurrent aphthous ulceration: A randomized controlled clinical trial. J Oral Pathol Med . 2020; 49:357-364. [PubMed 31514233]
260. Lazzerini M, Martelossi S, Magazzù G et al. Effect of thalidomide on clinical remission in children and adolescents with refractory Crohn disease: a randomized clinical trial. JAMA . 2013; 310:2164-73. [PubMed 24281461]
261. Deng X, Zhang J, Zhang J et al. Thalidomide reduces recurrence of ankylosing spondylitis in patients following discontinuation of etanercept. Rheumatol Int . 2013; 33:1409-13. [PubMed 23143621]
262. Misawa S, Sato Y, Katayama K et al. Safety and efficacy of thalidomide in patients with POEMS syndrome: a multicentre, randomised, double-blind, placebo-controlled trial. Lancet Neurol . 2016; 15:1129-37. [PubMed 27496680]
263. Droitcourt C, Rybojad M, Porcher R et al. A randomized, investigator-masked, double-blind, placebo-controlled trial on thalidomide in severe cutaneous sarcoidosis. Chest . 2014; 146:1046-1054. [PubMed 24945194]
264. Invernizzi R, Quaglia F, Klersy C et al. Efficacy and safety of thalidomide for the treatment of severe recurrent epistaxis in hereditary haemorrhagic telangiectasia: results of a non-randomised, single-centre, phase 2 study. Lancet Haematol . 2015; 2:e465-73. [PubMedCentral][PubMed 26686256]
265. Food and Drug Administration. FDA drug safety communication: FDA alerts healthcare professionals and oncology clinical investigators about two clinical trials on hold evaluating Keytruda® (pembrolizumab) in patients with multiple myeloma. Silver Spring, MD; 2017 Sep 20. From FDA website. [Web]
266. Fakhouri F, Guerraoui H, Presne C et al. Thalidomide in patients with multiple myeloma and renal failure. Br J Haematol . 2004; 125:96-7. [PubMed 15015981]
267. Dimopoulos MA, Sonneveld P, Leung N et al. International Myeloma Working Group Recommendations for the Diagnosis and Management of Myeloma-Related Renal Impairment. J Clin Oncol . 2016; 34:1544-57. [PubMed 26976420]
268. Mikhael J, Ismaila N, Cheung MC et al. Treatment of Multiple Myeloma: ASCO and CCO Joint Clinical Practice Guideline. J Clin Oncol . 2019; 37:1228-1263. [PubMed 30932732]
10013. Cavo M, Tacchetti P, Patriarca F et al. Bortezomib with thalidomide plus dexamethasone compared with thalidomide plus dexamethasone as induction therapy before, and consolidation therapy after, double autologous stem-cell transplantation in newly diagnosed multiple myeloma: a randomised phase 3 study. Lancet . 2010; 376:2075-85. [PubMed 21146205]
10014. Rosiñol L, Oriol A, Teruel AI et al. Superiority of bortezomib, thalidomide, and dexamethasone (VTD) as induction pretransplantation therapy in multiple myeloma: a randomized phase 3 PETHEMA/GEM study. Blood . 2012; 120:1589-96. [PubMed 22791289]
10015. Moreau P, Avet-Loiseau H, Facon T et al. Bortezomib plus dexamethasone versus reduced-dose bortezomib, thalidomide plus dexamethasone as induction treatment before autologous stem cell transplantation in newly diagnosed multiple myeloma. Blood . 2011; 118:5752-8; quiz 5982. [PubMed 21849487]
10016. Ludwig H, Viterbo L, Greil R et al. Randomized phase II study of bortezomib, thalidomide, and dexamethasone with or without cyclophosphamide as induction therapy in previously untreated multiple myeloma. J Clin Oncol . 2013; 31:247-55. [PubMed 23091109]
10020. Ludwig H, Greil R, Masszi T et al. Bortezomib, thalidomide and dexamethasone, with or without cyclophosphamide, for patients with previously untreated multiple myeloma: 5-year follow-up. Br J Haematol . 2015; 171:344-54. [PubMed 26153365]
10027. Moreau P, Hulin C, Macro M et al. VTD is superior to VCD prior to intensive therapy in multiple myeloma: results of the prospective IFM2013-04 trial. Blood . 2016; 127:2569-74. [PubMed 27002117]
10033. AHFS final determination of medical acceptance: Off-label use of bortezomib in combination with thalidomide and dexamethasone as induction therapy for newly diagnosed multiple myeloma in transplant-eligible patients. Published 17 May 2018.