Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Blood ; 135(7): 463-471, 2020 02 13.
Article in English | MEDLINE | ID: mdl-31841594

ABSTRACT

Ivosidenib (AG-120) is an oral, targeted agent that suppresses production of the oncometabolite 2-hydroxyglutarate via inhibition of the mutant isocitrate dehydrogenase 1 (IDH1; mIDH1) enzyme. From a phase 1 study of 258 patients with IDH1-mutant hematologic malignancies, we report results for 34 patients with newly diagnosed acute myeloid leukemia (AML) ineligible for standard therapy who received 500 mg ivosidenib daily. Median age was 76.5 years, 26 patients (76%) had secondary AML, and 16 (47%) had received ≥1 hypomethylating agent for an antecedent hematologic disorder. The most common all-grade adverse events were diarrhea (n = 18; 53%), fatigue (n = 16; 47%), nausea (n = 13; 38%), and decreased appetite (n = 12; 35%). Differentiation syndrome was reported in 6 patients (18%) (grade ≥3 in 3 [9%]) and did not require treatment discontinuation. Complete remission (CR) plus CR with partial hematologic recovery (CRh) rate was 42.4% (95% confidence interval [CI], 25.5% to 60.8%); CR 30.3% (95% CI, 15.6% to 48.7%). Median durations of CR+CRh and CR were not reached, with 95% CI lower bounds of 4.6 and 4.2 months, respectively; 61.5% and 77.8% of patients remained in remission at 1 year. With median follow-up of 23.5 months (range, 0.6-40.9 months), median overall survival was 12.6 months (95% CI, 4.5-25.7). Of 21 transfusion-dependent patients (63.6%) at baseline, 9 (42.9%) became transfusion independent. IDH1 mutation clearance was seen in 9/14 patients achieving CR+CRh (5/10 CR; 4/4 CRh). Ivosidenib monotherapy was well-tolerated and induced durable remissions and transfusion independence in patients with newly diagnosed AML. This trial was registered at www.clinicaltrials.gov as #NCT02074839.


Subject(s)
Glycine/analogs & derivatives , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/drug therapy , Mutation/genetics , Pyridines/therapeutic use , Aged , Aged, 80 and over , Blood Transfusion , Female , Glycine/adverse effects , Glycine/therapeutic use , Humans , Leukemia, Myeloid, Acute/genetics , Male , Middle Aged , Pyridines/adverse effects , Remission Induction , Survival Analysis , Translational Research, Biomedical , Treatment Outcome
2.
Eur J Clin Pharmacol ; 75(8): 1099-1108, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31011758

ABSTRACT

PURPOSE: To assess the effect of ethnicity, food, and itraconazole (strong CYP3A4 inhibitor) on the pharmacokinetics of ivosidenib after single oral doses in healthy subjects. METHODS: Three phase 1 open-label studies were performed. Study 1: Japanese and Caucasian subjects received single doses of 250, 500, or 1000 mg ivosidenib (NCT03071770). Part 1 of study 2 (a two-period crossover study): subjects received 500 mg ivosidenib after either an overnight fast or a high-fat meal. Subjects received 1000 mg ivosidenib after an overnight fast in the single period of part 2 (NCT02579707). Study 3: in period 1, subjects received 250 mg ivosidenib; then, in period 2, subjects received oral itraconazole (200 mg once daily) on days 1-18, plus 250 mg ivosidenib on day 5 (NCT02831972). RESULTS: Ivosidenib was well tolerated in all three studies. Study 1: pharmacokinetic profiles were generally comparable, although AUC and Cmax were slightly lower in Japanese subjects than in Caucasian subjects, by ~ 30 and 17%, respectively. Study 2: AUC increased by ~ 25% and Cmax by ~ 98%, when ivosidenib was administered with a high-fat meal compared with a fasted state. Study 3: co-administration of itraconazole increased ivosidenib AUC by 169% (90% CI 145-195) but had no effect on ivosidenib Cmax. CONCLUSIONS: No ivosidenib dose adjustment is deemed necessary for Japanese subjects. High-fat meals should be avoided when ivosidenib is taken with food. When co-administered with strong CYP3A4 inhibitors, monitoring for QT interval prolongation (a previously defined adverse event of interest) is recommended and an ivosidenib dose interruption or reduction may be considered. CLINICALTRIALS.GOV : NCT03071770, NCT02579707, and NCT02831972.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Glycine/analogs & derivatives , Itraconazole/pharmacology , Long QT Syndrome/epidemiology , Pyridines/pharmacokinetics , Administration, Oral , Adult , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Area Under Curve , Asian People , Cross-Over Studies , Dose-Response Relationship, Drug , Drug Interactions/ethnology , Female , Food-Drug Interactions/ethnology , Glycine/administration & dosage , Glycine/adverse effects , Glycine/pharmacokinetics , Healthy Volunteers , Humans , Itraconazole/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Long QT Syndrome/diagnosis , Long QT Syndrome/etiology , Male , Middle Aged , Pyridines/administration & dosage , Pyridines/adverse effects
3.
J Inherit Metab Dis ; 39(6): 807-820, 2016 11.
Article in English | MEDLINE | ID: mdl-27469509

ABSTRACT

D-2-hydroxyglutaric aciduria (D2HGA) type II is a rare neurometabolic disorder caused by germline gain-of-function mutations in isocitrate dehydrogenase 2 (IDH2), resulting in accumulation of D-2-hydroxyglutarate (D2HG). Patients exhibit a wide spectrum of symptoms including cardiomyopathy, epilepsy, developmental delay and limited life span. Currently, there are no effective therapeutic interventions. We generated a D2HGA type II mouse model by introducing the Idh2R140Q mutation at the native chromosomal locus. Idh2R140Q mice displayed significantly elevated 2HG levels and recapitulated multiple defects seen in patients. AGI-026, a potent, selective inhibitor of the human IDH2R140Q-mutant enzyme, suppressed 2HG production, rescued cardiomyopathy, and provided a survival benefit in Idh2R140Q mice; treatment withdrawal resulted in deterioration of cardiac function. We observed differential expression of multiple genes and metabolites that are associated with cardiomyopathy, which were largely reversed by AGI-026. These findings demonstrate the potential therapeutic benefit of an IDH2R140Q inhibitor in patients with D2HGA type II.


Subject(s)
Brain Diseases, Metabolic, Inborn/drug therapy , Cardiomyopathies/drug therapy , Isocitrate Dehydrogenase/antagonists & inhibitors , Mutation/drug effects , Small Molecule Libraries/pharmacology , Animals , Brain Diseases, Metabolic, Inborn/genetics , Disease Models, Animal , Isocitrate Dehydrogenase/genetics , Mice , Mutation/genetics
4.
Blood ; 121(24): 4917-24, 2013 Jun 13.
Article in English | MEDLINE | ID: mdl-23641016

ABSTRACT

Cancer-associated isocitrate dehydrogenase (IDH) mutations produce the metabolite 2-hydroxyglutarate (2HG), but the clinical utility of 2HG has not been established. We studied whether 2HG measurements in acute myeloid leukemia (AML) patients correlate with IDH mutations, and whether diagnostic or remission 2HG measurements predict survival. Sera from 223 de novo AML patients were analyzed for 2HG concentration by reverse-phase liquid chromatography-mass spectrometry. Pretreatment 2HG levels ranged from 10 to 30 000 ng/mL and were elevated in IDH-mutants (median, 3004 ng/mL), compared to wild-type IDH (median, 61 ng/mL) (P < .0005). 2HG levels did not differ among IDH1 or IDH2 allelic variants. In receiver operating characteristic analysis, a discriminatory level of 700 ng/mL optimally segregated patients with and without IDH mutations, and on subsequent mutational analysis of the 13 IDH wild-type samples with 2HG levels >700 ng/mL, 9 were identified to have IDH mutations. IDH-mutant patients with 2HG levels >200 at complete remission had shorter overall survival compared to 2HG ≤200 ng/mL (hazard ratio, 3.9; P = .02). We establish a firm association between IDH mutations and serum 2HG concentration in AML, and confirm that serum oncometabolite measurements provide useful diagnostic and prognostic information that can improve patient selection for IDH-targeted therapies.


Subject(s)
Glutarates/blood , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/genetics , Mutation, Missense , Adolescent , Adult , DNA Mutational Analysis , Female , Follow-Up Studies , Humans , Isocitrate Dehydrogenase/metabolism , Male , Middle Aged
5.
Blood ; 120(23): 4649-52, 2012 Nov 29.
Article in English | MEDLINE | ID: mdl-23074281

ABSTRACT

Mutations of genes encoding isocitrate dehydrogenase (IDH1 and IDH2) have been recently described in acute myeloid leukemia (AML). Serum and myeloblast samples from patients with IDH-mutant AML contain high levels of the metabolite 2-hydroxyglutarate (2-HG), a product of the altered IDH protein. In this prospective study, we sought to determine whether 2-HG can potentially serve as a noninvasive biomarker of disease burden through serial measurements in patients receiving conventional therapy for newly diagnosed AML. Our data demonstrate that serum, urine, marrow aspirate, and myeloblast 2-HG levels are significantly higher in IDH-mutant patients, with a correlation between baseline serum and urine 2-HG levels. Serum and urine 2-HG, along with IDH1/2-mutant allele burden in marrow, decreased with response to treatment. 2-HG decrease was more rapid with induction chemotherapy compared with DNA-methyltransferase inhibitor therapy. Our data suggest that serum or urine 2-HG may serve as noninvasive biomarkers of disease activity for IDH-mutant AML.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Glutarates/metabolism , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/metabolism , Acute Disease , Aged , Azacitidine/administration & dosage , Azacitidine/analogs & derivatives , Cytarabine/administration & dosage , DNA Mutational Analysis , Decitabine , Female , Glutarates/blood , Glutarates/urine , Granulocyte Precursor Cells/metabolism , Humans , Idarubicin/administration & dosage , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Leukemia, Myeloid/genetics , Male , Middle Aged , Mutation , Prospective Studies , Time Factors
6.
Toxicol Appl Pharmacol ; 262(1): 1-10, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22676972

ABSTRACT

Anthracycline-based regimens are a mainstay of early breast cancer therapy, however their use is limited by cardiac toxicity. The potential for cardiotoxicity is a major consideration in the design and development of combinatorial therapies incorporating anthracyclines and agents that target the HER2-mediated signaling pathway, such as trastuzumab. In this regard, HER2-targeted liposomal doxorubicin was developed to provide clinical benefit by both reducing the cardiotoxicity observed with anthracyclines and enhancing the therapeutic potential of HER2-based therapies that are currently available for HER2-overexpressing cancers. While documenting the enhanced therapeutic potential of HER2-targeted liposomal doxorubicin can be done with existing models, there has been no validated human cardiac cell-based assay system to rigorously assess the cardiotoxicity of anthracyclines. To understand if HER2-targeting of liposomal doxorubicin is possible with a favorable cardiac safety profile, we applied a human stem cell-derived cardiomyocyte platform to evaluate the doxorubicin exposure of human cardiac cells to HER2-targeted liposomal doxorubicin. To the best of our knowledge, this is the first known application of a stem cell-derived system for evaluating preclinical cardiotoxicity of an investigational agent. We demonstrate that HER2-targeted liposomal doxorubicin has little or no uptake into human cardiomyocytes, does not inhibit HER2-mediated signaling, results in little or no evidence of cardiomyocyte cell death or dysfunction, and retains the low penetration into heart tissue of liposomal doxorubicin. Taken together, this data ultimately led to the clinical decision to advance this drug to Phase I clinical testing, which is now ongoing as a single agent in HER2-expressing cancers.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/drug therapy , Doxorubicin/pharmacology , Drug Delivery Systems , Myocytes, Cardiac/drug effects , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/toxicity , Breast Neoplasms/pathology , Doxorubicin/administration & dosage , Doxorubicin/toxicity , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Nude , Myocytes, Cardiac/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction/drug effects
7.
Int J Cancer ; 129(8): 1963-9, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21154746

ABSTRACT

Soft tissue sarcomas (STS) represent a diverse group of histologic subtypes with targetable molecular alterations, often treated as a single disease. Sunitinib malate is a multitargeted receptor tyrosine kinase inhibitor active in other solid tumors carrying similar alterations (i.e., imatinib mesylate-refractory gastrointestinal stromal tumors). This single-institution phase II study investigated the safety and efficacy of sunitinib malate in three common STS subtypes. Patients with documented unresectable or metastatic STS (liposarcoma, leiomyosarcoma and malignant fibrous histiocytoma [MFH]), measurable disease, and 3 or less prior lines of therapy were eligible. Treatment consisted of sunitinib malate, 50 mg daily, for 4 weeks every 6 weeks. Forty-eight patients were enrolled, and 35% were heavily pretreated (≥ 2 prior lines of chemotherapy). The safety profile resembled previously known sunitinib malate toxicities. Median progression-free and overall survivals for liposarcoma, leiomyosarcoma, and MFH were 3.9 and 18.6, 4.2 and 10.1 and 2.5 and 13.6 months, respectively. The 3-month progression-free rates in the untreated and pretreated (chemotherapy) patients with liposarcoma, leiomyosarcoma and MFH were 75% and 69.2%, 60%, and 62.5% and 25% and 44.4%, respectively. With the caveats that a minority of patients with potentially indolent or low-grade disease could have been included and the small numbers, a 3-month progression-free rate of >40% suggests activity for sunitinib malate at least in liposarcomas and leiomyosarcomas. Thus, we believe that further investigation in these susceptible STS subtypes is warranted.


Subject(s)
Indoles/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrroles/therapeutic use , Sarcoma/drug therapy , Disease-Free Survival , Drug Administration Schedule , Drug Resistance, Neoplasm , Female , Histiocytoma, Malignant Fibrous/drug therapy , Humans , Indoles/adverse effects , Leiomyosarcoma/drug therapy , Liposarcoma/drug therapy , Male , Neoplasm Metastasis , Protein Kinase Inhibitors/adverse effects , Pyrroles/adverse effects , Recurrence , Retreatment , Sarcoma/pathology , Sunitinib , Survival Rate
8.
Cancer Chemother Pharmacol ; 85(5): 959-968, 2020 05.
Article in English | MEDLINE | ID: mdl-32296873

ABSTRACT

PURPOSE: Isocitrate dehydrogenase (IDH) mutations lead to formation of the oncometabolite 2-hydroxyglutarate (2-HG), which is elevated in several solid and liquid tumors. Ivosidenib (AG-120) is a targeted, potent, oral inhibitor of the mutant IDH1 protein. We describe detailed pharmacokinetics and pharmacodynamics of ivosidenib in patients with advanced hematologic malignancies with an IDH1 mutation treated in a phase I study (ClinicalTrials.gov NCT02074839). METHODS: Patients received single and multiple oral doses of ivosidenib from 100 mg twice daily to 1200 mg once daily (QD) in 28-day continuous cycles. Concentrations of ivosidenib and 2-HG in plasma, and 2-HG in bone marrow, were assessed at routine intervals. Plasma 4ß-hydroxycholesterol/cholesterol ratios were assessed as a marker of CYP3A activity. RESULTS: Ivosidenib was rapidly absorbed and slowly eliminated (half-life 72-138 h) after single and multiple dosing. Ivosidenib exhibited dose- and time-dependent pharmacokinetics, with exposure increasing sub-proportionally to dose, and clearance increasing with increasing dose. Plasma 2-HG concentrations were maximally and persistently inhibited in the majority of patients receiving 500-mg QD ivosidenib, to concentrations close to those observed in healthy subjects. Ivosidenib pharmacokinetics were not affected by mild or moderate renal impairment, mild hepatic impairment, age, weight, sex, race, or co-administration of weak CYP3A4 inhibitors or inducers. Moderate-to-strong CYP3A4 inhibitors decreased ivosidenib clearance. Ivosidenib also induced CYP3A enzyme activity, with increases in 4ß-hydroxycholesterol/cholesterol ratios of 119-168% at 500-mg QD ivosidenib. CONCLUSIONS: Ivosidenib 500-mg QD has favorable pharmacokinetic and pharmacodynamic profiles in patients with advanced hematologic malignancies with an IDH1 mutation. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov NCT02074839.


Subject(s)
Glycine/analogs & derivatives , Hematologic Neoplasms , Isocitrate Dehydrogenase , Pyridines , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Monitoring/methods , Female , Glycine/administration & dosage , Glycine/adverse effects , Glycine/pharmacokinetics , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Humans , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Maximum Tolerated Dose , Mutation , Neoplasm Staging , Pyridines/administration & dosage , Pyridines/adverse effects , Pyridines/pharmacokinetics , Treatment Outcome
9.
Leukemia ; 33(11): 2575-2584, 2019 11.
Article in English | MEDLINE | ID: mdl-30967620

ABSTRACT

Older adults with acute myeloid leukemia (AML) who are not fit for standard chemotherapy historically have poor outcomes. Approximately 12-15% of older patients with AML harbor isocitrate dehydrogenase 2 (IDH2) gene mutations. Enasidenib is an oral inhibitor of mutant IDH2 proteins. Among 39 patients with newly diagnosed mutant-IDH2 AML who received enasidenib monotherapy in this phase I/II trial, median age was 77 years (range 58-87) and 23 patients (59%) had had an antecedent hematologic disorder. The median number of enasidenib treatment cycles was 6.0 (range 1-35). The most common treatment-related adverse events were indirect hyperbilirubinemia (31%), nausea (23%), and fatigue, decreased appetite, and rash (18% each). Treatment-related grade 3-4 cytopenias were reported for eight patients (21%); there was no treatment-related grade 3-4 infections. Twelve patients achieved a response (overall response rate 30.8% [95% CI 17.0%, 47.6%]), including seven patients (18%) who attained complete remission. At a median follow-up of 8.4 months, the median duration of any response was not reached (NR). Median overall survival for all patients was 11.3 months (95% CI 5.7, 15.1), and was NR for responders. Oral, outpatient targeted treatment with enasidenib may benefit older adults with newly diagnosed mutant-IDH2 AML who are not candidates for cytotoxic regimens.


Subject(s)
Aminopyridines/therapeutic use , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/drug therapy , Triazines/therapeutic use , Aged , Aged, 80 and over , Enzyme Inhibitors/therapeutic use , Female , Hematologic Diseases/complications , Humans , Male , Maximum Tolerated Dose , Middle Aged , Mutant Proteins/genetics , Mutation , Remission Induction , Treatment Outcome
10.
Lancet Gastroenterol Hepatol ; 4(9): 711-720, 2019 09.
Article in English | MEDLINE | ID: mdl-31300360

ABSTRACT

BACKGROUND: Isocitrate dehydrogenase-1 (IDH1) is mutated in up to 25% of cholangiocarcinomas, especially intrahepatic cholangiocarcinoma. Ivosidenib is an oral, targeted inhibitor of mutant IDH1 (mIDH1) approved in the USA for the treatment of mIDH1 acute myeloid leukaemia in newly diagnosed patients ineligible for intensive chemotherapy and patients with relapsed or refractory disease. Ivosidenib is under clinical evaluation in a phase 1 study that aims to assess its safety and tolerability in patients with mIDH1 solid tumours. Here we report data for the mIDH1-cholangiocarcinoma cohort. METHODS: We did a phase 1 dose-escalation and expansion study of ivosidenib monotherapy in mIDH1 solid tumours at 12 clinical sites in the USA and one in France. The primary outcomes were safety, tolerability, maximum tolerated dose, and recommended phase 2 dose. Eligible patients had a documented mIDH1 tumour based on local testing, an Eastern Cooperative Oncology Group performance status of 0 or 1, one or more previous lines of therapy, and evaluable disease by Response Evaluation Criteria in Solid Tumors version 1.1. During dose escalation, ivosidenib was administered orally at 200-1200 mg daily in 28-day cycles in a standard 3 + 3 design; during expansion, patients received the selected dose on the basis of pharmacodynamic, pharmacokinetic, safety, and activity data from dose escalation. Safety and clinical activity analyses were reported for all patients with mIDH1-cholangiocarcinoma who were enrolled and received at least one dose of study treatment. Enrolment is complete, and the study is ongoing. This trial is registered at ClinicalTrials.gov, number NCT02073994. FINDINGS: Between March 14, 2014 and May 12, 2017, 73 patients with mIDH1-cholangiocarcinoma were enrolled and received ivosidenib. No dose-limiting toxicities were reported and maximum tolerated dose was not reached; 500 mg daily was selected for expansion. Common (≥20%) adverse events, regardless of cause, were fatigue (31 [42%]; two [3%] grade ≥3), nausea (25 [34%]; one [1%] grade ≥3), diarrhoea (23 [32%]), abdominal pain (20 [27%]; two [3%] grade ≥3), decreased appetite (20 [27%]; one [1%] grade ≥3), and vomiting (17 [23%]). Common grade 3 or worse adverse events were ascites (four [5%]) and anaemia (three [4%]); the only treatment-related grade 3 or worse adverse event in more than one patient was fatigue (two [3%]). Two (3%) patients had serious adverse events leading to on-treatment death (Clostridioides difficile infection and procedural haemorrhage); neither was assessed by the investigator as related to treatment. 46 (63%) patients had adverse events deemed related to ivosidenib, of which four (5%) were grade 3 or higher (two [3%] for fatigue; one [1%] each for decreased blood phosphorus and increased blood alkaline phosphatase). One serious adverse event was considered possibly related to treatment (grade 2 supraventricular extrasystoles). Four (5%; 95% CI 1·5-13·4) patients had a partial response. Median progression-free survival was 3·8 months (95% CI 3·6-7·3), 6-month progression-free survival was 40·1% (28·4-51·6), and 12-month progression-free survival was 21·8% (12·3-33·0). Median overall survival was 13·8 months (95% CI 11·1-29·3); however, data were censored for 48 patients (66%). INTERPRETATION: Ivosidenib might offer a well tolerated option for patients with mIDH1-cholangiocarcinoma. An ongoing, global phase 3 study is evaluating ivosidenib versus placebo in patients with previously treated nonresectable or metastatic mIDH1-cholangiocarcinoma. FUNDING: Agios Pharmaceuticals, Inc.


Subject(s)
Antineoplastic Agents/administration & dosage , Bile Duct Neoplasms/drug therapy , Cholangiocarcinoma/drug therapy , Glycine/analogs & derivatives , Pyridines/administration & dosage , Adult , Aged , Aged, 80 and over , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/mortality , Cholangiocarcinoma/genetics , Cholangiocarcinoma/mortality , Dose-Response Relationship, Drug , Female , Glycine/administration & dosage , Humans , Isocitrate Dehydrogenase/genetics , Male , Middle Aged , Mutation , Progression-Free Survival
11.
J Periodontol ; 78(9): 1839-45, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17760557

ABSTRACT

BACKGROUND: Rhabdomyosarcoma is a malignant neoplasm of primitive mesenchyme exhibiting skeletal muscle differentiation. Oral rhabdomyosarcoma is rare and accounts for only 0.04% of all head and neck malignancies. METHODS: A 33-year-old woman presented with an erythematous gingival mass involving the anterior maxillary gingiva. The lesion had been present for > or =13 months before presentation, and in recent months, it had become intermittently painful. RESULTS: Clinical examination exhibited erythema and enlargement of the interdental papillae between the left maxillary canine, lateral incisor, and central incisor. The tissue was boggy and tender on palpation. Incisional biopsies were performed, and microscopic examination showed a cellular proliferation of spindle-shaped to ovoid cells with hyperchromatic, enlarged, and pleomorphic nuclei. Many of the tumor cells exhibited abundant eosinophilic cytoplasm. Immunohistochemical stains showed the tumor cells to be positive for desmin, myogenin, and myogenic differentiation 1 (MyoD1). A diagnosis of embryonal rhabdomyosarcoma was made. The patient was treated by surgical resection with postoperative chemotherapy and radiation. The patient had no evidence of disease at a follow-up examination 1 month after completion of therapy. CONCLUSIONS: Oral rhabdomyosarcoma can develop insidiously. Pain is a variable presenting symptom, and early lesions may be mistaken for benign neoplastic, inflammatory, or infectious processes. Over several decades, a multidisciplinary treatment approach that includes surgical removal if resectable, in combination with multiagent chemotherapy and possibly radiation therapy, has improved survival rates.


Subject(s)
Gingival Neoplasms/pathology , Rhabdomyosarcoma/pathology , Adult , Female , Humans , Maxilla
12.
J Clin Pharmacol ; 52(5): 691-703, 2012 May.
Article in English | MEDLINE | ID: mdl-21953571

ABSTRACT

Trastuzumab emtansine (T-DM1) is a HER2-targeted antibody-drug conjugate in development for treatment of HER2-positive cancers. T-DM1 has been tested as a single agent in a phase I and 2 phase II studies of patients with heavily pretreated metastatic breast cancer (MBC), with the maximum tolerated dose established at 3.6 mg/kg intravenously for every-3-week dosing. The authors present results from the population pharmacokinetics analysis for T-DM1. Population pharmacokinetics for T-DM1 were characterized using a clinical database of 273 patients from the 3 studies. Pharmacokinetics was best described by a linear 2-compartment model. Population estimates (interindividual variability [IIV]) for pharmacokinetic parameters were clearance, 0.7 L/d (21.0%); central compartment volume (V(c)), 3.33 L (13.2%); peripheral compartment volume (V(p)), 0.89 L (50.4%); and intercompartmental clearance, 0.78 L/d. Body weight, albumin, tumor burden, and aspartate aminotransferase levels were identified as statistically significant covariates accounting for interindividual variability in T-DM1 pharmacokinetics, with body weight having a greater effect on IIV of clearance and V(c) than other covariates. T-DM1 exposure was relatively consistent across the weight range following body weight-based dosing. This analysis suggests no further T-DM1 dose adjustments are necessary in heavily pretreated patients with MBC.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , Immunotoxins/pharmacokinetics , Maytansine/analogs & derivatives , Receptor, ErbB-2/metabolism , Tubulin Modulators/pharmacokinetics , Ado-Trastuzumab Emtansine , Adult , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Aspartate Aminotransferases/blood , Biomarkers, Tumor/immunology , Body Weight , Breast Neoplasms/blood , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Drug Administration Schedule , Drug Dosage Calculations , Female , Humans , Immunotoxins/administration & dosage , Immunotoxins/adverse effects , Infusions, Intravenous , Linear Models , Maximum Tolerated Dose , Maytansine/administration & dosage , Maytansine/adverse effects , Maytansine/pharmacokinetics , Middle Aged , Models, Biological , Protein Binding , Receptor, ErbB-2/immunology , Serum Albumin/metabolism , Trastuzumab , Treatment Outcome , Tubulin Modulators/administration & dosage , Tubulin Modulators/adverse effects , Tumor Burden
13.
Cancer Control ; 15(1): 55-62, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18094661

ABSTRACT

BACKGROUND: Overall, the survival rate for cancer patients has continued to improve over the past several decades. However, those aged 15 to 29 years have not experienced the same improvements in survival. This review explores some of the challenges faced by adolescent and young adult (AYA) cancer patients and their survivorship needs. METHODS: Using the OVID Medline database from 1966 to present, a variety of search terms including "adolescent," "young adult," and "cancer survivorship" were entered. Articles related to those obtained by the search were also collected. Additional data were obtained from the SEER database AYA monograph, the Childhood Cancer Survivorship Study, the Report of the Adolescent and Young Adult Oncology Progress Review Group, and the Long-Term Follow-Up Recommendations of the Children's Oncology Group. RESULTS: Cancer patients in this age-group are at increased risk for second malignancies, cardiotoxicity, and reproductive difficulties. Few data exist concerning intellectual and other psychosocial issues for this specific patient population. CONCLUSIONS: More research is needed to develop accurate data on treatment and survivorship for AYA patients. A separate cancer discipline focusing on improving outcomes in treatment and survivorship among AYA patients should be developed in major academic cancer centers.


Subject(s)
Neoplasms/complications , Neoplasms/psychology , Survivors/psychology , Survivors/statistics & numerical data , Adolescent , Adult , Clinical Trials as Topic , Heart Diseases/epidemiology , Heart Diseases/etiology , Humans , Infertility/epidemiology , Infertility/etiology , Neoplasms, Second Primary/epidemiology
14.
Cancer Control ; 15(1): 7-12, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18094656

ABSTRACT

BACKGROUND: Intraoperative pathologic diagnosis of bone and soft tissue lesions is an important yet challenging tool in clinical musculoskeletal oncology practice. There is limited information in the literature addressing the practical issues commonly encountered regarding intraoperative frozen section of musculoskeletal lesions. METHODS: A literature review and retrospective review of practical experience in intraoperative pathology consultation at our institute's sarcoma program were conducted to investigate the pitfalls and limitations of frozen section and potential solutions to overcome these problems. RESULTS: Frozen section evaluation is an essential and reliable procedure for guiding intraoperative decisions. Intraoperative cytology as an adjunct to frozen section enhances the accuracy of diagnosis of bone and soft tissue lesions. Cytology can accurately diagnose certain entities alone and is superior to frozen section for certain tumor types and for evaluating bone marrow margins. It is also invaluable in triaging cases for ancillary studies and for tumor banking. Practical working protocols can be developed to optimize the usefulness of intraoperative pathologic consultation. CONCLUSIONS: Intraoperative pathology consultation should be done in an interdisciplinary approach by correlating clinical, radiologic, and pathologic information. As an adjunct to frozen section, cytology and gross examination enhance the accuracy of diagnosis of musculoskeletal lesions.


Subject(s)
Bone Neoplasms/diagnosis , Frozen Sections , Interprofessional Relations , Pathology, Surgical , Soft Tissue Neoplasms/diagnosis , Aged , Female , Humans , Intraoperative Period , Male , Middle Aged , Referral and Consultation
15.
Cancer Control ; 15(1): 47-54, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18094660

ABSTRACT

BACKGROUND: Sarcomas are rare mesenchymally derived tumors for which there are limited treatment options. This paper discusses the current therapeutic potential of directed tyrosine kinase inhibitors (TKIs) in sarcoma. METHODS: The authors review antibody-based strategies and small molecular inhibitors of TKIs, with specific emphasis placed on the potential use of these targeted agents as therapeutic options for the treatment of sarcomas that are not gastrointestinal stromal tumors. RESULTS: Many TKs have been shown to be mutated or overexpressed in human sarcoma tumors and cell lines and may serve as potential targets for promising new sarcoma therapies. Furthermore, the novel mechanism of targeting TKs may complement the antitumor activity of existing sarcoma treatment options. CONCLUSIONS: TKIs such as imatinib, sunitinib, and sorefanib are promising new therapeutic options for the management of patients with soft tissue sarcoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/metabolism , Sarcoma/drug therapy , Sarcoma/enzymology , Animals , Clinical Trials as Topic , Humans , Protein-Tyrosine Kinases/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL