SELECT BRAND ITCH RELIEF- diphenhydramine hydrochloride and zinc acetate spray Stati Uniti - inglese - NLM (National Library of Medicine)

select brand itch relief- diphenhydramine hydrochloride and zinc acetate spray

select brand - diphenhydramine hydrochloride (unii: tc2d6jad40) (diphenhydramine - unii:8gts82s83m), zinc acetate (unii: fm5526k07a) (zinc cation - unii:13s1s8sf37) - diphenhydramine hydrochloride .2 g in 10 ml - - temporarily relieves pain and itching due to: insect bites minor burns sunburn minor skin irritations minor cuts scrapes rashes due to poison ivy, poison oak, and poison sumac - insect bites - minor burns - sunburn - minor skin irritations - minor cuts - scrapes - rashes due to poison ivy, poison oak, and poison sumac - dries the oozing and weeping of poison • ivy • oak • sumac

SELECT BRAND MEDICATED VAPORIZING STEAM (camphor- synthetic liquid Stati Uniti - inglese - NLM (National Library of Medicine)

select brand medicated vaporizing steam (camphor- synthetic liquid

select brand - camphor (synthetic) (unii: 5tjd82a1et) (camphor (synthetic) - unii:5tjd82a1et) - camphor (synthetic) .623 g in 10 ml - cough suppressant temporarily relieves cough ocurring with a cold

JADENU- deferasirox tablet, film coated
JADENU- deferasirox granule Stati Uniti - inglese - NLM (National Library of Medicine)

jadenu- deferasirox tablet, film coated jadenu- deferasirox granule

novartis pharmaceuticals corporation - deferasirox (unii: v8g4mof2v9) (deferasirox - unii:v8g4mof2v9) - deferasirox 90 mg - jadenu is indicated for the treatment of chronic iron overload due to blood transfusions (transfusional hemosiderosis) in patients 2 years of age and older. jadenu is indicated for the treatment of chronic iron overload in patients 10 years of age and older with non-transfusion-dependent thalassemia (ntdt) syndromes and with a liver iron concentration (lic) of at least 5 milligrams of iron per gram of liver dry weight (mg fe/g dw) and a serum ferritin greater than 300 mcg/l. the safety and efficacy of jadenu when administered with other iron chelation therapy have not been established. jadenu is contraindicated in patients with: - estimated gfr less than 40 ml/min/1.73 m2  [see dosage and administration (2.5), warnings and precautions (5.1)] ; - poor performance status [see warnings and precautions (5.1, 5.3)] ; - high-risk myelodysplastic syndromes (this patient population was not studied and is not expected to benefit from chelation therapy) ; - advanced malignancies [see warnings and precautions (5.1, 5.3)] ; - platelet counts less than 50 x 109 /l [see warnings and precautions (5.3, 5.4)] ; - known hypersensitivity to deferasirox or any component of jadenu [see warnings and precautions (5.7), adverse reactions (6.2)] . risk summary there are no studies with the use of jadenu in pregnant women to inform drug-associated risks. administration of deferasirox to rats during pregnancy resulted in decreased offspring viability and an increase in renal anomalies in male offspring at doses that were about or less than the recommended human dose on a mg/m2 basis. no fetal effects were noted in pregnant rabbits at doses equivalent to the human recommended dose on an mg/m2 basis. jadenu should be used during pregnancy only if the potential benefit justifies the potential risk to the fetus. the background risk of major birth defects and miscarriage for the indicated population is unknown. all pregnancies had a background risk of birth defect, loss, or other adverse outcomes. however, the background risk in the u.s. general population of major birth defects is 2% to 4% and of miscarriage is 15% to 20% of clinically recognized pregnancies. data animal data in embryo-fetal developmental studies, pregnant rats and rabbits received oral deferasirox during the period of organogenesis at doses up to 100 mg/kg/day in rats and 50 mg/kg/day in rabbits (1.2 times the maximum recommended human dose (mrhd) on an mg/m2 basis). these doses resulted in maternal toxicity but no fetal harm was observed. in a prenatal and postnatal developmental study, pregnant rats received oral deferasirox daily from organogenesis through lactation day 20 at doses of 10, 30, and 90 mg/kg/day (0.1, 0.3, and 1.0 times the mrhd on a mg/m2 basis). maternal toxicity, loss of litters, and decreased offspring viability occurred at 90 mg/kg/day (1.0 times the mrhd on a mg/m2 basis), and increases in renal anomalies in male offspring occurred at 30 mg/kg/day (0.3 times the mrhd on a mg/m2 basis). risk summary no data are available regarding the presence of jadenu or its metabolites in human milk, the effects of the drug on the breastfed child, or the effects of the drug on milk production. deferasirox and its metabolites were excreted in rat milk. because many drugs are excreted in human milk and because of the potential for serious adverse reactions in a breastfeeding child from deferasirox and its metabolites, a decision should be made whether to discontinue breastfeeding or to discontinue the drug, taking into account the importance of the drug to the mother. contraception counsel patients to use non-hormonal method(s) of contraception since jadenu can render hormonal contraceptives ineffective [see drug interactions (7.2)] . transfusional iron overload the safety and effectiveness of jadenu have been established in pediatric patients 2 years of age and older for the treatment of transfusional iron overload [see dosage and administration (2.1)] . safety and effectiveness have not been established in pediatric patients less than 2 years of age for the treatment of transfusional iron overload. pediatric approval for treatment of transfusional iron overload was based on clinical studies of 292 pediatric patients 2 years to less than 16 years of age with various congenital and acquired anemias. seventy percent of these patients had beta-thalassemia [see indications and usage (1), dosage and administration (2.1), clinical studies (14)] . in those clinical studies, 173 children (ages 2 to < 12 years) and 119 adolescents (ages 12 to < 17 years) were exposed to deferasirox. a trial conducted in treatment naïve pediatric patients, 2 to < 18 years of age with transfusional iron overload (nct02435212) did not provide additional relevant information about the safety or effectiveness of the deferasirox granules dosage form (jadenu sprinkle) compared to the deferasirox oral tablets for suspension dosage form (exjade). iron overload in non-transfusion-dependent thalassemia syndromes the safety and effectiveness of jadenu have been established in patients 10 years of age and older for the treatment of chronic iron overload with non-transfusion-dependent thalassemia (ntdt) syndromes [see dosage and administration (2.2)] . safety and effectiveness have not been established in patients less than 10 years of age with chronic iron overload in ntdt syndromes. pediatric approval for treatment of ntdt syndromes with liver iron (fe) concentration (lic) of at least 5 mg fe per gram of dry weight and a serum ferritin greater than 300 mcg/l was based on 16 pediatric patients treated with deferasirox therapy (10 years to less than 16 years of age) with chronic iron overload and ntdt. use of jadenu in these age groups is supported by evidence from adequate and well-controlled studies of deferasirox in adult and pediatric patients [see indications and usage (1.2), dosage and administration (2.2), clinical studies (14)] . in general, risk factors for deferasirox-associated kidney injury include preexisting renal disease, volume depletion, overchelation, and concomitant use of other nephrotoxic drugs. acute kidney injury, and acute liver injury and failure has occurred in pediatric patients. in a pooled safety analysis, pediatric patients with higher deferasirox exposures had a greater probability of renal toxicity and decreased renal function, resulting in increased deferasirox exposure and progressive renal toxicity/kidney injury. higher rates of renal aes have been identified among pediatric patients receiving exjade doses greater than 25 mg/kg/day equivalent to 17.5 mg/kg/day jadenu when their serum ferritin values were less than 1,000 mcg/l [see dosage and administration (2.5), warnings and precautions (5.1, 5.6), adverse reactions (6.1, 6.2)] . monitoring recommendations for all pediatric patients with transfusional iron overload and ntdt it is recommended that serum ferritin be monitored every month to assess the patient’s response to therapy and to minimize the risk of overchelation [see warnings and precautions (5.6)] . monitor renal function by estimating gfr using an egfr prediction equation appropriate for pediatric patients and evaluate renal tubular function. monitor renal function more frequently in pediatric patients in the presence of renal toxicity risk factors, including episodes of dehydration, fever and acute illness that may result in volume depletion or decreased renal perfusion. use the minimum effective dose [see warnings and precautions (5.1)] . interrupt jadenu in pediatric patients with transfusional iron overload, and consider dose interruption in pediatric patients with non-transfusion-dependent iron overload, for acute illnesses, which can cause volume depletion, such as vomiting, diarrhea, or prolonged decreased oral intake, and monitor more frequently. resume therapy as appropriate, based on assessments of renal function, when oral intake and volume status are normal. evaluate the risk benefit profile of continued jadenu use in the setting of decreased renal function. avoid use of other nephrotoxic drugs [see dosage and administration (2.5), warnings and precautions (5.1)] . juvenile animal toxicity data renal toxicity was observed in adult mice, rats, and marmoset monkeys administered deferasirox at therapeutic doses. in a neonatal and juvenile toxicity study in rats, deferasirox was administered orally from postpartum day 7 through 70, which equates to a human age range of term neonate through adolescence. increased renal toxicity was identified in juvenile rats compared to adult rats at a dose based on mg/m2 approximately 0.4 times the recommended dose of 20 mg/kg/day. a higher frequency of renal abnormalities was noted when deferasirox was administered to non-iron overloaded animals compared to iron overloaded animals. four hundred thirty-one (431) patients greater than or equal to 65 years of age were studied in clinical trials of deferasirox in the transfusional iron overload setting. two hundred twenty-five (225) of these patients were between 65 and 75 years of age while 206 were greater than or equal to 75 years of age. the majority of these patients had myelodysplastic syndrome (mds) (n = 393). in these trials, elderly patients experienced a higher frequency of adverse reactions than younger patients. monitor elderly patients for early signs or symptoms of adverse reactions that may require a dose adjustment. elderly patients are at increased risk for toxicity due to the greater frequency of decreased hepatic, renal, or cardiac function, and of concomitant disease or other drug therapy. dose selection for an elderly patient should be cautious, usually starting at the low end of the dosing range. in elderly patients, including those with mds, individualize the decision to remove accumulated iron based on clinical circumstances and the anticipated clinical benefit and risks of deferasirox tablets for oral suspension therapy. jadenu is contraindicated in patients with egfr less than 40 ml/min/1.73 m2  [see contraindications (4)] . for patients with renal impairment (egfr 40 to 60 ml/min/1.73 m2 ), reduce the starting dose by 50% [see dosage and administration (2.4), clinical pharmacology (12.3)]. exercise caution in pediatric patients with an egfr between 40 and 60 ml/min/1.73 m2  [see dosage and administration (2.4)]. if treatment is needed, use the minimum effective dose with enhanced monitoring of glomerular and renal tubular function. individualize dose titration based on improvement in renal injury [see dosage and administration (2.4, 2.5)] . jadenu can cause glomerular dysfunction, renal tubular toxicity, or both, and can result in acute renal failure. monitor all patients closely for changes in egfr and renal tubular dysfunction during jadenu treatment. if either develops, consider dose reduction, interruption or discontinuation of jadenu until glomerular or renal tubular function returns to baseline [see dosage and administration (2.4, 2.5), warnings and precautions (5.1)] . avoid use in patients with severe (child-pugh c) hepatic impairment. for patients with moderate (child-pugh b) hepatic impairment, reduce the starting dose by 50%. closely monitor patients with mild (child-pugh a) or moderate (child-pugh b) hepatic impairment for efficacy and adverse reactions that may require dose titration [see dosage and administration (2.4), warnings and precautions (5.2), clinical pharmacology (12.3)] .

GILENYA- fingolimod hcl capsule Stati Uniti - inglese - NLM (National Library of Medicine)

gilenya- fingolimod hcl capsule

novartis pharmaceuticals corporation - fingolimod hydrochloride (unii: g926ec510t) (fingolimod - unii:3qn8byn5qf) - fingolimod 0.5 mg - gilenya is indicated for the treatment of relapsing forms of multiple sclerosis (ms), to include clinically isolated syndrome, relapsing-remitting disease, and active secondary progressive disease, in patients 10 years of age and older. gilenya is contraindicated in patients who have: - in the last 6 months experienced myocardial infarction, unstable angina, stroke, transient ischemic attack (tia), decompensated heart failure requiring hospitalization or class iii/iv heart failure - a history or presence of mobitz type ii second-degree or third-degree av block or sick sinus syndrome, unless patient has a functioning pacemaker [see warnings and precautions (5.1)] - a baseline qtc interval ≥ 500 msec - cardiac arrhythmias requiring anti-arrhythmic treatment with class ia or class iii anti-arrhythmic drugs - had a hypersensitivity reaction to fingolimod or any of the excipients in gilenya. observed reactions include rash, urticaria and angioedema upon treatment initiation [see warnings and precautions (5.14)] . pregnancy exposure registry there is a pregnancy exposure registry that monitors pregnancy outcomes in women exposed to gilenya during pregnancy. physicians are encouraged to enroll pregnant patients, or pregnant women may register themselves in the gilenya pregnancy registry by calling 1-877-598-7237, sending an email to gpr@quintiles.com, or visiting www.gilenyapregnancyregistry.com. risk summary based on findings from animal studies, gilenya may cause fetal harm when administered to a pregnant woman. data from prospective reports to the gilenya pregnancy registry (gpr) are currently not sufficient to allow for an adequate assessment of the drug-associated risk for birth defects and miscarriage in humans. in oral studies conducted in rats and rabbits, fingolimod demonstrated developmental toxicity, including an increase in malformations (rats) and embryolethality, when given to pregnant animals. in rats, the highest no-effect dose was less than the recommended human dose of 0.5 mg/day on a body surface area (mg/m2 ) basis. the most common fetal visceral malformations in rats were persistent truncus arteriosus and ventricular septal defect. the receptor affected by fingolimod (sphingosine 1-phosphate receptor) is known to be involved in vascular formation during embryogenesis (see data ). advise pregnant women of the potential risk to a fetus. in the us general population, the estimated background risk of major birth defects and miscarriage in clinically recognized pregnancies is 2% to 4% and 15% to 20%, respectively. the background risk of major birth defects and miscarriage for the indicated population is unknown. clinical considerations in females planning to become pregnant, gilenya should be stopped 2 months before planned conception. the possibility of severe increase in disability should be considered in women who discontinue or are considering discontinuation of gilenya because of pregnancy or planned pregnancy. in many of the cases in which increase in disability was reported after stopping gilenya, patients had stopped gilenya because of pregnancy or planned pregnancy [see warnings and precautions (5.9)] . data animal data when fingolimod was orally administered to pregnant rats during the period of organogenesis (0, 0.03, 0.1, and 0.3 mg/kg/day or 0, 1, 3, and 10 mg/kg/day), increased incidences of fetal malformations and embryofetal deaths were observed at all but the lowest dose tested (0.03 mg/kg/day), which is less than the recommended human dose (rhd) on a mg/m2 basis. oral administration to pregnant rabbits during organogenesis (0, 0.5, 1.5, and 5 mg/kg/day) resulted in increased incidences of embryofetal mortality and fetal growth retardation at the mid and high doses. the no-effect dose for these effects in rabbits (0.5 mg/kg/day) is approximately 20 times the rhd on a mg/m2 basis. when fingolimod was orally administered to female rats during pregnancy and lactation (0, 0.05, 0.15, and 0.5 mg/kg/day), pup survival was decreased at all doses and a neurobehavioral (learning) deficit was seen in offspring at the high dose. the low-effect dose of 0.05 mg/kg/day is similar to the rhd on a mg/m2 basis. risk summary there are no data on the presence of fingolimod in human milk, the effects on the breastfed infant, or the effects of the drug on milk production. fingolimod is excreted in the milk of treated rats. when a drug is present in animal milk, it is likely that the drug will be present in human milk. the developmental and health benefits of breastfeeding should be considered along with the mother’s clinical need for gilenya and any potential adverse effects on the breastfed infant from gilenya or from the underlying maternal condition. pregnancy testing the pregnancy status of females of reproductive potential should be verified prior to starting treatment with gilenya [see use in specific populations (8.1)] . contraception before initiation of gilenya treatment, females of reproductive potential should be counseled on the potential for a serious risk to the fetus and the need for effective contraception during treatment with gilenya [see warnings and precautions (5.8) and use in specific populations (8.1)] . since it takes approximately 2 months to eliminate the compound from the body after stopping treatment, the potential risk to the fetus may persist and women should use effective contraception during this period [see warnings and precautions (5.8, 5.13)] . safety and effectiveness of gilenya for the treatment of relapsing forms of multiple sclerosis in pediatric patients 10 to less than 18 years of age were established in one randomized, double-blind clinical study in 215 patients (gilenya n = 107; intramuscular interferon (ifn) beta-1a n = 108) [see clinical studies (14.2)] . in the controlled pediatric study, the safety profile in pediatric patients (10 to less than 18 years of age) receiving gilenya 0.25 mg or 0.5 mg daily was similar to that seen in adult patients. in the pediatric study, cases of seizures were reported in 5.6% of gilenya-treated patients and 0.9% of interferon beta-1a-treated patients. it is recommended that pediatric patients, if possible, complete all immunizations in accordance with current immunization guidelines prior to initiating gilenya therapy. safety and effectiveness of gilenya in pediatric patients below the age of 10 years have not been established. juvenile animal toxicity data in a study in which fingolimod (0.3, 1.5, or 7.5 mg/kg/day) was orally administered to young rats from weaning through sexual maturity, changes in bone mineral density and persistent neurobehavioral impairment (altered auditory startle) were observed at all doses. delayed sexual maturation was noted in females at the highest dose tested and in males at all doses. the bone changes observed in fingolimod-treated juvenile rats are consistent with a reported role of s1p in the regulation of bone mineral homeostasis. when fingolimod (0.5 or 5 mg/kg/day) was orally administered to rats from the neonatal period through sexual maturity, a marked decrease in t-cell dependent antibody response was observed at both doses. this effect had not fully recovered by 6-8 weeks after the end of treatment. overall, a no-effect dose for adverse developmental effects in juvenile animals was not identified. clinical ms studies of gilenya did not include sufficient numbers of patients aged 65 years and over to determine whether they respond differently than younger patients. gilenya should be used with caution in patients aged 65 years and over, reflecting the greater frequency of decreased hepatic, or renal, function and of concomitant disease or other drug therapy. because fingolimod, but not fingolimod-phosphate, exposure is doubled in patients with severe hepatic impairment, patients with severe hepatic impairment should be closely monitored, as the risk of adverse reactions may be greater [see warnings and precautions (5.5), clinical pharmacology (12.3)] . no dose adjustment is needed in patients with mild or moderate hepatic impairment. the blood level of some gilenya metabolites is increased (up to 13-fold) in patients with severe renal impairment [see clinical pharmacology (12.3)] . the toxicity of these metabolites has not been fully explored. the blood level of these metabolites has not been assessed in patients with mild or moderate renal impairment.

NEORAL- cyclosporine capsule, liquid filled
NEORAL- cyclosporine solution Stati Uniti - inglese - NLM (National Library of Medicine)

neoral- cyclosporine capsule, liquid filled neoral- cyclosporine solution

novartis pharmaceuticals corporation - cyclosporine (unii: 83hn0gtj6d) (cyclosporine - unii:83hn0gtj6d) - cyclosporine 25 mg - neoral is indicated for the prophylaxis of organ rejection in kidney, liver, and heart allogeneic transplants. neoral has been used in combination with azathioprine and corticosteroids. neoral is indicated for the treatment of patients with severe active, rheumatoid arthritis where the disease has not adequately responded to methotrexate. neoral can be used in combination with methotrexate in rheumatoid arthritis patients who do not respond adequately to methotrexate alone. neoral is indicated for the treatment of adult, nonimmunocompromised patients with severe (i.e., extensive and/or disabling), recalcitrant, plaque psoriasis who have failed to respond to at least one systemic therapy (e.g., puva, retinoids, or methotrexate) or in patients for whom other systemic therapies are contraindicated or cannot be tolerated. while rebound rarely occurs, most patients will experience relapse with neoral as with other therapies upon cessation of treatment. neoral is contraindicated in patients with a hypersensitivity to cyclosporine or to any of the ingredients of the formulation. rheumatoid arthritis patients with abnormal renal function, uncontrolled hypertension, or malignancies should not receive neoral. psoriasis patients who are treated with neoral should not receive concomitant puva or uvb therapy, methotrexate or other immunosuppressive agents, coal tar or radiation therapy. psoriasis patients with abnormal renal function, uncontrolled hypertension, or malignancies should not receive neoral. although no adequate and well-controlled studies have been completed in children, transplant recipients as young as one year of age have received neoral with no unusual adverse effects. the safety and efficacy of neoral treatment in children with juvenile rheumatoid arthritis or psoriasis below the age of 18 have not been established. in rheumatoid arthritis clinical trials with cyclosporine, 17.5% of patients were age 65 or older. these patients were more likely to develop systolic hypertension on therapy, and more likely to show serum creatinine rises ≥ 50% above the baseline after 3 to 4 months of therapy. clinical studies of neoral in transplant and psoriasis patients did not include a sufficient number of subjects aged 65 and over to determine whether they respond differently from younger subjects. other reported clinical experiences have not identified differences in response between the elderly and younger patients. in general, dose selection for an elderly patient should be cautious, usually starting at the low end of the dosing range, reflecting the greater frequency of decreased hepatic, renal, or cardiac function, and of concomitant disease or other drug therapy.

TASIGNA- nilotinib capsule Stati Uniti - inglese - NLM (National Library of Medicine)

tasigna- nilotinib capsule

novartis pharmaceuticals corporation - nilotinib (unii: f41401512x) (nilotinib - unii:f41401512x) - nilotinib 200 mg - tasigna is indicated for the treatment of adult and pediatric patients greater than or equal to 1 year of age with newly diagnosed philadelphia chromosome positive chronic myeloid leukemia (ph+ cml) in chronic phase. tasigna is indicated for the treatment of adult patients with chronic phase and accelerated phase philadelphia chromosome positive chronic myelogenous leukemia (ph+ cml) resistant or intolerant to prior therapy that included imatinib. tasigna is indicated for the treatment of pediatric patients greater than or equal to 1 year of age with chronic phase and accelerated phase philadelphia chromosome positive chronic myeloid leukemia (ph+ cml) with resistance or intolerance to prior tyrosine-kinase inhibitor (tki) therapy. tasigna is contraindicated in patients with hypokalemia, hypomagnesemia, or long qt syndrome [see boxed warning]. risk summary based on findings from animal studies and the mechanism of action, tasigna can cause fetal harm when administered to a pregnant woman [see clinical pharmacology (12.1)] . there are no available data in pregnant women to inform the drug-associated risk. in animal reproduction studies, administration of nilotinib to pregnant rats and rabbits during organogenesis caused adverse developmental outcomes, including embryo-fetal lethality, fetal effects, and fetal variations in rats and rabbits at maternal exposures (auc) approximately 2 and 0.5 times, respectively, the exposures in patients at the recommended dose (see data ). advise pregnant women of the potential risk to a fetus. the background risk of major birth defects and miscarriage for the indicated population is unknown. all pregnancies have a background risk of birth defect, loss, or other adverse outcomes. in the u.s. general population, the estimated background risk of major birth defects and miscarriage in clinically recognized pregnancies are 2% to 4% and 15% to 20%, respectively. data animal data in embryo-fetal development studies in rats and rabbits, pregnant animals received oral doses of nilotinib up to 100 mg/kg/day and 300 mg/kg/day, respectively, during the period of organogenesis. in rats, oral administration of nilotinib produced embryo-lethality/fetal effects at doses ≥ 30 mg/kg/day. at ≥ 30 mg/kg/day, skeletal variations of incomplete ossification of the frontals and misshapen sternebra were noted, and there was an increased incidence of small renal papilla and fetal edema. at 100 mg/kg/day, nilotinib was associated with maternal toxicity (decreased gestation weight, gravid uterine weight, net weight gain, and food consumption) and resulted in a single incidence of cleft palate and two incidences of pale skin were noted in the fetuses. a single incidence of dilated ureters was noted in a fetus also displaying small renal papilla at 100 mg/kg/day. additional variations of forepaw and hindpaw phalanx unossified, fused sternebra, bipartite sternebra ossification, and incomplete ossification of the cervical vertebra were noted at 100 mg/kg/day. in rabbits, oral administration of nilotinib resulted in the early sacrifice of two females, maternal toxicity and increased resorption of fetuses at 300 mg/kg/day. fetal skeletal variations (incomplete ossification of the hyoid, bent hyoid, supernumerary short detached ribs and the presence of additional ossification sites near the nasals, frontals and in the sternebral column) were also increased at this dose in the presence of maternal toxicity. slight maternal toxicity was evident at 100 mg/kg/day but there were no reproductive or embryo-fetal effects at this dose. at 30 mg/kg/day in rats and 300 mg/kg/day in rabbits, the maternal systemic exposure (auc) were 72700 ng*hr/ml and 17100 ng*hr/ml, respectively, representing approximately 2 and 0.5 times the exposure in humans at the highest recommended dose 400 mg twice daily. when pregnant rats were dosed with nilotinib during organogenesis and through lactation, the adverse effects included a longer gestational period, lower pup body weights until weaning and decreased fertility indices in the pups when they reached maturity, all at a maternal dose of 60 mg/kg (i.e., 360 mg/m2 , approximately 0.7 times the clinical dose of 400 mg twice daily based on body surface area). at doses up to 20 mg/kg (i.e., 120 mg/m2 , approximately 0.25 times the clinical dose of 400 mg twice daily based on body surface area) no adverse effects were seen in the maternal animals or the pups. risk summary there are no data on the presence of nilotinib or its metabolites in human milk or its effects on a breastfed child or on milk production. however, nilotinib is present in the milk of lactating rats. because of the potential for serious adverse reactions in a breastfed child, advise women not to breastfeed during treatment with tasigna and for 14 days after the last dose. animal data after a single 20 mg/kg of [14 c] nilotinib dose to lactating rats, the transfer of parent drug and its metabolites into milk was observed. the overall milk-to-plasma exposure ratio of total radioactivity was approximately 2, based on the auc0-24h or auc0-inf values. no rat metabolites of nilotinib were detected that were unique to milk. based on animal studies, tasigna can cause fetal harm when administered to a pregnant woman [see use in specific populations (8.1)] . pregnancy testing females of reproductive potential should have a pregnancy test prior to starting treatment with tasigna. contraception females advise females of reproductive potential to use effective contraception during treatment with tasigna and for 14 days after the last dose. infertility the risk of infertility in females or males of reproductive potential has not been studied in humans. in studies in rats and rabbits, the fertility in males and females was not affected [see nonclinical toxicology (13.1)] . the safety and effectiveness of tasigna have been established in pediatric patients greater than or equal to 1 year of age with newly diagnosed and resistant or intolerant ph+ cml in chronic phase [see clinical studies (14.5)] . there are no data for pediatric patients under 2 years of age. use of tasigna in pediatric patients 1 year to less than 2 years of age with newly diagnosed or resistant or intolerant ph+ cml in chronic phase is supported by efficacy in pediatric patients 2 to 6 years of age for these indications. the safety and effectiveness of tasigna have been established in pediatric patients greater than or equal to 1 year of age with resistant or intolerant ph+ cml in accelerated phase based on evidence of effectiveness from an adequate and well-controlled single-arm study in adults [see clinical studies (14.2)] with safety data from two pediatric studies as described in the next paragraph. use of tasigna in pediatric patients 1 to less than 18 years of age is supported by evidence from two clinical trials [see clinical studies (14.5)] . the 25 patients with newly diagnosed ph+ cml-cp were in the following age groups: 6 children (age 2 to less than 12 years) and 19 adolescents (age 12 to less than 18 years). the 44 patients with resistant or intolerant ph+ cml-cp included 18 children (age 2 to less than 12 years) and 26 adolescents (age 12 to less than 18 years). all pediatric patients received tasigna treatment at a dose of 230 mg/m2 twice daily, rounded to the nearest 50 mg dose (to a maximum single dose of 400 mg). no differences in efficacy or safety were observed between the different age subgroups in the two trials. the frequency, type, and severity of adverse reactions observed were generally consistent with those observed in adults, with the exception of the laboratory abnormalities of hyperbilirubinemia (grade 3/4: 16%) and transaminase elevation (ast grade 3/4: 2.9%, alt grade 3/4: 10%), which were reported at a higher frequency in pediatric patients than in adults [see adverse reactions (6.1)] . for pediatric growth and development, growth retardation has been reported in pediatric patients with ph+ cml-cp treated with tasigna [see warnings and precautions (5.14), adverse reactions (6.1)] . the safety and effectiveness of tasigna in pediatric patients below the age of 1 year with newly diagnosed, or resistant or intolerant ph+ cml in chronic phase and accelerated phase, have not been established. in the clinical trials of tasigna (patients with newly diagnosed ph+ cml-cp and resistant or intolerant ph+ cml-cp and cml-ap), approximately 12% and 30% of patients were 65 years or over, respectively. - patients with newly diagnosed ph+ cml-cp: there was no difference in major molecular response between patients aged less than 65 years and those greater than or equal to 65 years. - patients with resistant or intolerant cml-cp: there was no difference in major cytogenetic response rate between patients aged less than 65 years and those greater than or equal to 65 years. - patients with resistant or intolerant cml-ap: the hematologic response rate was 44% in patients less than 65 years of age and 29% in patients greater than or equal to 65 years. no major differences for safety were observed in patients greater than or equal to 65 years of age as compared to patients less than 65 years. in the clinical trials, patients with a history of uncontrolled or significant cardiovascular disease, including recent myocardial infarction, congestive heart failure, unstable angina or clinically significant bradycardia, were excluded. caution should be exercised in patients with relevant cardiac disorders [see boxed warning, warnings and precautions (5.2)]. reduce the tasigna dosage in patients with hepatic impairment and monitor the qt interval closely in these patients [see dosage and administration (2.7), clinical pharmacology (12.3)] .

TAFINLAR- dabrafenib capsule
TAFINLAR- dabrafenib tablet, for suspension Stati Uniti - inglese - NLM (National Library of Medicine)

tafinlar- dabrafenib capsule tafinlar- dabrafenib tablet, for suspension

novartis pharmaceuticals corporation - dabrafenib mesylate (unii: b6dc89i63e) (dabrafenib - unii:qgp4ha4g1b) - dabrafenib 50 mg - tafinlar®  is indicated as a single agent for the treatment of patients with unresectable or metastatic melanoma with braf v600e mutation as detected by an fda-approved test. tafinlar is indicated, in combination with trametinib, for the treatment of patients with unresectable or metastatic melanoma with braf v600e or v600k mutations, as detected by an fda-approved test [see dosage and administration (2.1)] . tafinlar is indicated, in combination with trametinib, for the adjuvant treatment of patients with melanoma with braf v600e or v600k mutations, as detected by an fda-approved test, and involvement of lymph node(s), following complete resection [see dosage and administration (2.1)] . tafinlar is indicated, in combination with trametinib, for the treatment of patients with metastatic non-small cell lung cancer (nsclc) with braf v600e mutation as detected by an fda-approved test [see dosage and administration (2.1)] . tafinlar is indicated, in combination with trametinib, for the treatment of patients with locally advanced or metastatic anaplastic thyroid cancer (atc) with braf v600e mutation and with no satisfactory locoregional treatment options [see dosage and administration (2.1)] . tafinlar is indicated, in combination with trametinib, for the treatment of adult and pediatric patients 1 year of age and older with unresectable or metastatic solid tumors with braf v600e mutation who have progressed following prior treatment and have no satisfactory alternative treatment options [see dosage and administration (2.1)] . this indication is approved under accelerated approval based on overall response rate (orr) and duration of response (dor) [see clinical studies (14.6)] . continued approval for this indication may be contingent upon verification and description of clinical benefit in a confirmatory trial(s). tafinlar is indicated, in combination with trametinib, for the treatment of pediatric patients 1 year of age and older with low-grade glioma (lgg) with a braf v600e mutation who require systemic therapy [see dosage and administration (2.1)] . - tafinlar is not indicated for treatment of patients with colorectal cancer because of known intrinsic resistance to braf inhibition [see indications and usage (1.6), clinical pharmacology (12.1)] . - tafinlar is not indicated for treatment of patients with wild-type braf solid tumors [see warnings and precautions (5.2)] . none. risk summary based on findings from animal reproduction studies and its mechanism of action [see clinical pharmacology (12.1)] , tafinlar can cause fetal harm when administered to a pregnant woman. there is insufficient data in pregnant women exposed to tafinlar to assess the risks. dabrafenib was teratogenic and embryotoxic in rats at doses three times greater than the human exposure at the recommended adult clinical dose of 150 mg twice daily (see data) . advise pregnant women of the potential risk to a fetus. in the u.s. general population, the estimated background risk of major birth defects and miscarriage in clinically recognized pregnancies is 2% to 4% and 15% to 20%, respectively. data animal data in a combined female fertility and embryo-fetal development study in rats conducted during the period of organogenesis, developmental toxicity consisted of embryo-lethality, ventricular septal defects, and variation in thymic shape at a dabrafenib dose of 300 mg/kg/day [approximately three times the human exposure at the recommended adult dose based on area under the curve (auc)]. at doses of 20 mg/kg/day or greater (equivalent to the human exposure at the recommended adult dose based on auc), rats demonstrated delays in skeletal development and reduced fetal body weight. risk summary there are no data on the presence of dabrafenib in human milk, or the effects of dabrafenib on the breastfed child or on milk production. because of the potential for serious adverse reactions in breastfed children, advise women not to breastfeed during treatment with tafinlar and for 2 weeks following the last dose. pregnancy testing verify pregnancy status in females of reproductive potential prior to initiating tafinlar. contraception based on data from animal studies and its mechanism of action, tafinlar can cause fetal harm when administered to pregnant women [see use in specific populations (8.1 )]. females advise female patients of reproductive potential to use effective contraception during treatment with tafinlar and for 2 weeks after the last dose. counsel patients to use a non-hormonal method of contraception since tafinlar can render hormonal contraceptives ineffective [see drug interactions (7.2)] . males to avoid potential drug exposure to pregnant partners and female partners of reproductive potential, advise male patients (including those who have had vasectomies) with female partners of reproductive potential to use condoms during treatment with tafinlar and for 2 weeks after the last dose. infertility females advise female patients of reproductive potential that tafinlar may impair fertility. a reduction in fertility was observed in female rats at dose exposures equivalent to the human exposure at the recommended adult dose. a reduction in the number of corpora lutea was noted in pregnant rats at dose exposures approximately three times the human exposure at the recommended adult dose [see nonclinical toxicology (13.1)]. males advise male patients of the potential risk for impaired spermatogenesis which may be irreversible. effects on spermatogenesis have been observed in animals treated with dabrafenib at dose exposures up to three times the human exposure at the recommended adult dose [see nonclinical toxicology (13.1)]. braf v600e mutation-positive unresectable or metastatic solid tumors and lgg the safety and effectiveness of tafinlar in combination with trametinib have been established in pediatric patients 1 year of age and older with unresectable or metastatic solid tumors with braf v600e mutation who have progressed following prior treatment and have no satisfactory alternative treatment options; or with lgg with braf v600e mutation who require systemic therapy. use of tafinlar in combination with trametinib for these indications is supported by evidence from studies x2101 and g2201 that enrolled 171 patients (1 to < 18 years) with braf v600 mutation-positive advanced solid tumors, of which 4 (2.3%) patients were 1 to < 2 years of age, 39 (23%) patients were 2 to < 6 years of age, 54 (32%) patients were 6 to < 12 years of age, and 74 (43%) patients were 12 to < 18 years of age [see adverse reactions (6.1), clinical pharmacology (12.3), clinical studies (14.6, 14.7)] . the safety and effectiveness of tafinlar in combination with trametinib have not been established for these indications in pediatric patients less than 1 year old. the safety and effectiveness of tafinlar as a single agent in pediatric patients have not been established. juvenile animal toxicity data in a repeat-dose toxicity study in juvenile rats, an increased incidence of kidney cysts and tubular deposits were noted at doses as low as 0.2 times the human exposure at the recommended adult dose based on auc. additionally, forestomach hyperplasia, decreased bone length, and early vaginal opening were noted at doses as low as 0.8 times the human exposure at the recommended adult dose based on auc. of the 586 patients with various solid tumors who received single agent tafinlar, 22% were aged 65 years and older. of the 187 patients with melanoma who received single-agent tafinlar in the break-3 study, 21% were aged 65 years or older [see clinical studies (14.1)] . no overall differences in the effectiveness or safety of tafinlar were observed between geriatric patients as compared to younger adults in the break-3 study. of the 994 patients with melanoma who received tafinlar plus trametinib in the combi-d, combi-v, and combi-ad studies [see clinical studies (14.2, 14.3)] , 21% were aged 65 years and older and 5% were aged 75 years and older. no overall differences in the effectiveness of tafinlar plus trametinib were observed in geriatric patients as compared to younger adults across these melanoma studies. the incidences of peripheral edema (26% vs. 12%) and anorexia (21% vs. 9%) were increased in geriatric patients as compared to younger adults in these studies. of the 171 patients with nsclc who received tafinlar in study brf113928, there were insufficient numbers of geriatric patients to determine whether they respond differently from younger adults [see clinical studies (14.4)] . of the 26 patients with atc who received tafinlar in study brf117019, 77% were aged 65 years and older, and 31% were aged 75 years and older [see clinical studies (14.5)] . this study in atc did not include sufficient numbers of younger adults to determine whether they respond differently compared to geriatric patients. dose adjustment is not recommended for patients with mild (bilirubin ≤ upper limit of normal (uln) and aspartate aminotransferase (ast) > uln or bilirubin > 1x to 1.5x uln and any ast) hepatic impairment. as hepatic metabolism and biliary secretion are the primary routes of elimination of dabrafenib and its metabolites, patients with moderate (bilirubin > 1.5x to 3x uln and any ast) to severe (bilirubin > 3x to 10x uln and any ast) hepatic impairment may have increased exposure. an appropriate dosage has not been established for patients with moderate to severe hepatic impairment [see clinical pharmacology (12.3)] . - read this “instructions for use” carefully before you prepare and take or give tafinlar oral suspension for the first time and each time you get a refill. there may be new information. - this “instructions for use” does not take the place of talking with your healthcare provider about your or your child’s medical condition and treatment. - your healthcare provider or pharmacist should show you how to prepare and take or give a dose of tafinlar oral suspension correctly. always take or give tafinlar exactly as your healthcare provider tells you to. - if you have any questions about how to prepare and take or give a dose of tafinlar oral suspension, talk to your healthcare provider or pharmacist. - always use the dosing cup that comes with your tafinlar pack. if your pack does not contain a dosing cup, contact your healthcare provider or pharmacist. - use only clean water to rinse. do not use soap or dishwashing liquid to clean the dosing cup. - if at any time tafinlar oral suspension gets on your or your child’s skin, wash the area well with soap and water. - if at any time tafinlar oral suspension gets in your or your child’s eyes, rinse the eyes well with cool water. - if you spill any tafinlar oral suspension, follow the instructions at the end of this “instructions for use” in section e. “how to clean up any spilled tafinlar oral suspension”. - you will receive the tafinlar tablets in a sealed bottle. the tafinlar tablets must be mixed in water before taking or giving a dose of tafinlar oral suspension. the tablets break apart (disperse) in the water and may not fully dissolve. follow the instructions below to mix the tablets in water. - the prescribed number of tablets - 1 dosing cup - 1 teaspoon - drinking water - add cool drinking water up to the markings on the dosing cup, as follows: - if the prescribed dose is 1 to 4 tablets, you will need about 5 ml of water. - if the prescribed dose is 5 to 15 tablets, you will need about 10 ml of water. - do not throw away (dispose of) the cap. - if you are opening the bottle for the first time, remove the seal from the bottle. - add the prescribed number of tablets into the water in your dosing cup. - the bottle contains 2 plastic canisters to keep the tablets dry. if either canister falls out when you are taking out the tablets, re-insert it back into the bottle. - with your other hand, gently stir the water and tablets with the handle of a teaspoon until the tablets break apart (disperse). - it may take 3 minutes or more for the tablets to disperse. after the tablets disperse, the tafinlar oral suspension should be cloudy white, but may contain small pieces. - take or give the tafinlar oral suspension no later than 30 minutes after the tablets have been dispersed in water. - if more than 30 minutes have passed, throw away the tafinlar oral suspension following the instructions in section d and restart from the beginning of section a. if you are not sure how to throw away the tafinlar oral suspension, ask your healthcare provider or pharmacist. - if the prescribed dose is 1 to 4 tablets: do steps 7 through 9 one time. - if the prescribed dose is 5 to 15 tablets: do steps 7 through 9 two times. - if the prescribed dose is 1 to 3 tablets, the feeding tube size that may be used is 10 french gauge or larger. - if the prescribed dose is 4 to 15 tablets, the feeding tube size that may be used is 12 french gauge or larger. - if any of the tafinlar oral suspension comes into contact with your skin or eyes when you are following the steps below, follow the instructions in the section “important information you need to know before taking or giving tafinlar tablets for oral suspension”. - if any of the tafinlar oral suspension spills, follow the instruction in section e. “how to clean up any spilled tafinlar oral suspension”. - wash and dry your hands before giving a dose of tafinlar oral suspension. - it is important to give all of the medicine residue that is left in the oral syringe and feeding tube. repeat steps 4 through 7 three times to make sure that you give a full dose of tafinlar. - shake off excess water then wipe dry using clean paper towels. - you can also wash the teaspoon in a dishwasher. - throw away unused tafinlar tablets or oral suspension, or old dosing cups into the trash. do not pour tafinlar oral suspension down the drain. - ask your healthcare provider or pharmacist about how to safely throw away tafinlar tablets or oral suspension if you are not sure. - store the bottle of tafinlar tablets for oral suspension at room temperature between 68°f to 77°f (20°c to 25°c). - store the bottle of tafinlar tablets for oral suspension, along with the two plastic canisters inside the original packaging, with the cap tightly closed. the canisters contain a drying agent (desiccant) to help keep your medicine dry. - tafinlar tablets for oral suspension come in a bottle with a child-resistant cap. - throw away any tafinlar oral suspension if it is not taken or given within 30 minutes after it is prepared. t2024-24

PROMACTA- eltrombopag olamine tablet, film coated
PROMACTA- eltrombopag olamine powder, for suspension Stati Uniti - inglese - NLM (National Library of Medicine)

promacta- eltrombopag olamine tablet, film coated promacta- eltrombopag olamine powder, for suspension

novartis pharmaceuticals corporation - eltrombopag olamine (unii: 4u07f515lg) (eltrombopag - unii:s56d65xj9g) - eltrombopag 12.5 mg - promacta is indicated for the treatment of thrombocytopenia in adult and pediatric patients 1 year and older with persistent or chronic immune thrombocytopenia (itp) who have had an insufficient response to corticosteroids, immunoglobulins, or splenectomy. promacta should be used only in patients with itp whose degree of thrombocytopenia and clinical condition increase the risk for bleeding. promacta is indicated for the treatment of thrombocytopenia in patients with chronic hepatitis c to allow the initiation and maintenance of interferon-based therapy. promacta should be used only in patients with chronic hepatitis c whose degree of thrombocytopenia prevents the initiation of interferon-based therapy or limits the ability to maintain interferon-based therapy. - promacta is indicated in combination with standard immunosuppressive therapy (ist) for the first-line treatment of adult and pediatric patients 2 years and older with severe aplastic anemia. - promacta is indicated for the treatment of patients with severe aplastic anemia who have had an insufficient response to immunosuppressive therapy. - promacta is not indicated for the treatment of patients with myelodysplastic syndromes (mds) [see warnings and precautions (5.3)] . - safety and efficacy have not been established in combination with direct-acting antiviral agents used without interferon for treatment of chronic hepatitis c infection. none. risk summary available data from a small number of published case reports and postmarketing experience with promacta use in pregnant women are insufficient to assess any drug-associated risks for major birth defects, miscarriage, or adverse maternal or fetal outcomes. in animal reproduction and developmental toxicity studies, oral administration of eltrombopag to pregnant rats during organogenesis resulted in embryolethality and reduced fetal weights at maternally toxic doses. these effects were observed at doses resulting in exposures that were six times the human clinical exposure based on area under the curve (auc) in patients with persistent or chronic itp at 75 mg/day, and three times the auc in patients with chronic hepatitis c at 100 mg/day (see data). the estimated background risk of major birth defects and miscarriage for the indicated population is unknown. all pregnancies have a background risk of birth defect, loss, or other adverse outcomes. in the u.s. general population, the estimated background risk of major birth defects and of miscarriage in clinically recognized pregnancies is 2% to 4% and 15% to 20%, respectively. data animal data in an early embryonic development study, female rats received oral eltrombopag at doses of 10, 20, or 60 mg/kg/day (0.8, 2, and 6 times, respectively, the human clinical exposure based on auc in patients with itp at 75 mg/day and 0.3, 1, and 3 times, respectively, the human clinical exposure based on auc in patients with chronic hepatitis c at 100 mg/day). increased pre- and post-implantation loss and reduced fetal weight were observed at the highest dose which also caused maternal toxicity. in an embryo-fetal development study eltrombopag was administered orally to pregnant rats during the period of organogenesis at doses of 10, 20, or 60 mg/kg/day (0.8, 2, and 6 times, respectively, the human clinical exposure based on auc in patients with itp at 75 mg/day and 0.3, 1, and 3 times, respectively, the human clinical exposure based on auc in patients with chronic hepatitis c at 100 mg/day). decreased fetal weights (6% to 7%) and a slight increase in the presence of cervical ribs were observed at the highest dose which also caused maternal toxicity. however, no evidence of major structural malformations was observed. in an embryo-fetal development study eltrombopag was administered orally to pregnant rabbits during the period of organogenesis at doses of 30, 80, or 150 mg/kg/day (0.04, 0.3, and 0.5 times, respectively, the human clinical exposure based on auc in patients with itp at 75 mg/day and 0.02, 0.1, and 0.3 times, respectively, the human clinical exposure based on auc in patients with chronic hepatitis c at 100 mg/day). no evidence of fetotoxicity, embryolethality, or teratogenicity was observed. in a pre- and post-natal developmental toxicity study in pregnant rats (f0), oral eltrombopag was administered from gestation day 6 through lactation day 20. no adverse effects on maternal reproductive function or on the development of the offspring (f1) were observed at doses up to 20 mg/kg/day (2 times the human clinical exposure based on auc in patients with itp at 75 mg/day and similar to the human clinical exposure based on auc in patients with chronic hepatitis c at 100 mg/day). eltrombopag was detected in the plasma of offspring (f1). the plasma concentrations in pups increased with dose following administration of drug to the f0 dams. risk summary there are no data regarding the presence of eltrombopag or its metabolites in human milk, the effects on the breastfed child, or the effects on milk production. however, eltrombopag was detected in the pups of lactating rats 10 days postpartum suggesting the potential for transfer during lactation. due to the potential for serious adverse reactions in a breastfed child from promacta, breastfeeding is not recommended during treatment. contraception based on animal reproduction studies, promacta can cause fetal harm when administered to a pregnant woman. sexually-active females of reproductive potential should use effective contraception (methods that result in less than 1% pregnancy rates) when using promacta during treatment and for at least 7 days after stopping treatment with promacta. the safety and efficacy of promacta have been established in pediatric patients 1 year and older with persistent or chronic itp and in pediatric patients 2 years and older with ist-naïve severe aplastic anemia (in combination with h-atg and cyclosporine). safety and efficacy in pediatric patients below the age of 1 year with itp have not been established. safety and efficacy in pediatric patients with thrombocytopenia associated with chronic hepatitis c and refractory severe aplastic anemia have not been established. the safety and efficacy of promacta in pediatric patients 1 year and older with persistent or chronic itp were evaluated in two double-blind, placebo-controlled trials [see adverse reactions (6.1), clinical studies (14.1)] . the pharmacokinetics of eltrombopag have been evaluated in 168 pediatric patients 1 year and older with itp dosed once daily [see clinical pharmacology (12.3)] . see dosage and administration (2.1) for dosing recommendations for pediatric patients 1 year and older. the safety and efficacy of promacta in combination with h-atg and cyclosporine for the first-line treatment of severe aplastic anemia in pediatric patients 2 years and older were evaluated in a single-arm, open-label trial [see adverse reactions (6.1), clinical studies (14.3)] . a total of 26 pediatric patients (ages 2 to < 17 years) were evaluated; 12 children (aged 2 to < 12 years) and 14 adolescents (aged 12 to < 17). see dosage and administration (2.3) for dosing recommendations for pediatric patients 2 years and older. the safety and efficacy of promacta in combination with h-atg and cyclosporine in pediatric patients younger than 2 years for the first-line treatment of severe aplastic anemia have not yet been established. in patients 2 to 16 years of age, 69% of patients experienced serious adverse events compared to 42% in patients 17 years and older. among the 12 patients who were 2 to 11 years of age in the promacta d1-m6 cohort and reached the 6-month assessment or withdrew earlier, the complete response rate at month 6 was 8% versus 46% in patients age 12 to 16 years and 50% in patients 17 years of age and older. of the 106 patients in two randomized clinical trials of promacta 50 mg in persistent or chronic itp, 22% were 65 years of age and over, while 9% were 75 years of age and over. of the 1439 patients in two randomized clinical trials of promacta in patients with chronic hepatitis c and thrombocytopenia, 7% were 65 years of age and over, while < 1% were 75 years of age and over. of the 196 patients who received promacta for the treatment of severe aplastic anemia, 18% were 65 years of age and over, while 3% were 75 years of age and over. no overall differences in safety or effectiveness were observed between these patients and younger patients. patients with persistent or chronic itp and severe aplastic anemia reduce the initial dose of promacta in patients with persistent or chronic itp (adult and pediatric patients 6 years and older only) or refractory severe aplastic anemia who also have hepatic impairment (child-pugh class a, b, c) [see dosage and administration (2.1, 2.3), warnings and precautions (5.2), clinical pharmacology (12.3)] . in a clinical trial in patients with severe aplastic anemia who had not received prior definitive immunosuppressive therapy, patients with baseline alt or ast > 5 x uln were ineligible to participate. if a patient with hepatic impairment (child-pugh class a, b, c) initiates therapy with promacta for the first-line treatment of severe aplastic anemia, reduce the initial dose [see dosage and administration (2.3), warnings and precautions (5.2), clinical pharmacology (12.3)] . patients with chronic hepatitis c no dosage adjustment is recommended in patients with chronic hepatitis c and hepatic impairment [see clinical pharmacology (12.3)] . reduce the initial dose of promacta for patients of east-/southeast-asian ancestry with itp (adult and pediatric patients 6 years and older only) or severe aplastic anemia [see dosage and administration (2.1, 2.3), clinical pharmacology (12.3)] . no reduction in the initial dose of promacta is recommended in patients of east-/southeast-asian ancestry with chronic hepatitis c [see clinical pharmacology (12.3)] .

SELECT BRAND LICE KILLING- piperonyl butoxide, pyrethrum extract shampoo Stati Uniti - inglese - NLM (National Library of Medicine)

select brand lice killing- piperonyl butoxide, pyrethrum extract shampoo

select brand - piperonyl butoxide (unii: lwk91tu9ah) (piperonyl butoxide - unii:lwk91tu9ah), pyrethrum extract (unii: zum06l90gv) (pyrethrum extract - unii:zum06l90gv) - piperonyl butoxide 4 g in 100 ml - treats head, pubic (crab), and body lice lice treatment