Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 156
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Cancer ; 139(7): 1648-57, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27194579

ABSTRACT

The eukaryotic translation initiation factor 4E (eIF4E) is a potent oncogene that is found to be dysregulated in 30% of human cancer, including colorectal carcinogenesis (CRC). ISIS 183750 is a second-generation antisense oligonucleotide (ASO) designed to inhibit the production of the eIF4E protein. In preclinical studies we found that EIF4e ASOs reduced expression of EIF4e mRNA and inhibited proliferation of colorectal carcinoma cells. An additive antiproliferative effect was observed in combination with irinotecan. We then performed a clinical trial evaluating this combination in patients with refractory cancer. No dose-limiting toxicities were seen but based on pharmacokinetic data and tolerability the dose of irinotecan was reduced to 160 mg/m(2) biweekly. Efficacy was evaluated in 15 patients with irinotecan-refractory colorectal cancer. The median time of disease control was 22.1 weeks. After ISIS 183750 treatment, peripheral blood levels of eIF4E mRNA were decreased in 13 of 19 patients. Matched pre- and posttreatment tumor biopsies showed decreased eIF4E mRNA levels in five of nine patients. In tumor tissue, the intracellular and stromal presence of ISIS 183750 was detected by IHC in all biopsied patients. Although there were no objective responses stable disease was seen in seven of 15 (47%) patients who were progressing before study entry, six of whom were stable at the time of the week 16 CT scan. We were also able to confirm through mandatory pre- and posttherapy tumor biopsies penetration of the ASO into the site of metastasis.


Subject(s)
Camptothecin/analogs & derivatives , Colorectal Neoplasms/therapy , Eukaryotic Initiation Factor-4E/antagonists & inhibitors , Oligonucleotides, Antisense/therapeutic use , Oligoribonucleotides/therapeutic use , Adult , Aged , Camptothecin/adverse effects , Camptothecin/blood , Camptothecin/therapeutic use , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Combined Modality Therapy , Eukaryotic Initiation Factor-4E/genetics , Female , HCT116 Cells , Humans , Irinotecan , Male , Middle Aged , Oligonucleotides , Oligonucleotides, Antisense/genetics , Oligoribonucleotides/genetics , RNA, Messenger/blood , RNA, Messenger/genetics
2.
Breast Cancer Res Treat ; 160(1): 51-59, 2016 11.
Article in English | MEDLINE | ID: mdl-27620882

ABSTRACT

PURPOSE: Improved therapies and imaging modalities are needed for the treatment of breast cancer brain metastases (BCBM). ANG1005 is a drug conjugate consisting of paclitaxel covalently linked to Angiopep-2, designed to cross the blood-brain barrier. We conducted a biomarker substudy to evaluate 18F-FLT-PET for response assessment. METHODS: Ten patients with measurable BCBM received ANG1005 at a dose of 550 mg/m2 IV every 21 days. Before and after cycle 1, patients underwent PET imaging with 18F-FLT, a thymidine analog, retention of which reflects cellular proliferation, for comparison with gadolinium-contrast magnetic resonance imaging (Gd-MRI) in brain metastases detection and response assessment. A 20 % change in uptake after one cycle of ANG1005 was deemed significant. RESULTS: Thirty-two target and twenty non-target metastatic brain lesions were analyzed. The median tumor reduction by MRI after cycle 1 was -17.5 % (n = 10 patients, lower, upper quartiles: -25.5, -4.8 %) in target lesion size compared with baseline. Fifteen of twenty-nine target lesions (52 %) and 12/20 nontarget lesions (60 %) showed a ≥20 % decrease post-therapy in FLT-PET SUV change (odds ratio 0.71, 95 % CI: 0.19, 2.61). The median percentage change in SUVmax was -20.9 % (n = 29 lesions; lower, upper quartiles: -42.4, 2.0 %), and the median percentage change in SUV80 was also -20.9 % (n = 29; lower, upper quartiles: -49.0, 0.0 %). Two patients had confirmed partial responses by PET and MRI lasting 6 and 18 cycles, respectively. Seven patients had stable disease, receiving a median of six cycles. CONCLUSIONS: ANG1005 warrants further study in BCBM. Results demonstrated a moderately strong association between MRI and 18F-FLT-PET imaging.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Paclitaxel/analogs & derivatives , Peptides/therapeutic use , Adult , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Biomarkers , Biomarkers, Tumor , Brain Neoplasms/diagnosis , Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , Combined Modality Therapy , Female , Fluorodeoxyglucose F18 , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Magnetic Resonance Imaging , Middle Aged , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Paclitaxel/therapeutic use , Peptides/administration & dosage , Peptides/adverse effects , Positron-Emission Tomography , Treatment Outcome
3.
ScientificWorldJournal ; 2014: 919570, 2014.
Article in English | MEDLINE | ID: mdl-24563633

ABSTRACT

Blood supply is essential for development and growth of tumors and angiogenesis is the fundamental process of new blood vessel formation from preexisting ones. Angiogenesis is a prognostic indicator for a variety of tumors, and it coincides with increased shedding of neoplastic cells into the circulation and metastasis. Several molecules such as cell surface receptors, growth factors, and enzymes are involved in this process. While antiangiogenic therapy for cancer has been proposed over 20 years ago, it has garnered much controversy in recent years within the scientific community. The complex relationships between the angiogenic signaling cascade and antiangiogenic substances have indicated the angiogenic pathway as a valid target for anticancer drug development and VEGF has become the primary antiangiogenic drug target. This review discusses the basic and clinical perspectives of angiogenesis highlighting the importance of comparative biology in understanding tumor angiogenesis and the integration of these model systems for future drug development.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Signal Transduction/drug effects , Animals , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Humans , Neoplasms/physiopathology , Neoplastic Stem Cells/metabolism , Neovascularization, Pathologic/physiopathology , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism
4.
Pharmacogenomics J ; 10(3): 191-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20038957

ABSTRACT

The anticancer agent docetaxel shows significant inter-individual variation in its pharmacokinetic and toxicity profile. Thalidomide is an active anticancer agent and also shows wide pharmacological variation. Past pharmacogenetic research has not explained this variation. Patients with prostate cancer enrolled in a randomized phase II trial using docetaxel and thalidomide versus docetaxel alone were genotyped using the Affymetrix DMET 1.0 platform, which tests for 1256 genetic variations in 170 drug disposition genes. Genetic polymorphisms were analyzed for associations with clinical response and toxicity. In all, 10 single-nucleotide polymorphisms (SNPs) in three genes were potentially associated with response to therapy: peroxisome proliferator-activated receptor-delta (PPAR-delta), sulfotransferase family, cytosolic, 1C, member 2 (SULT1C2) and carbohydrate (chondroitin 6) sulfotransferase 3 (CHST3). In addition, 11 SNPs in eight genes were associated with toxicities to treatment: spastic paraplegia 7 (pure and complicated autosomal recessive) (SPG7), CHST3, cytochrome P450, family 2, subfamily D, polypeptide 6 (CYP2D6), N-acetyltransferase 2 (arylamine N-acetyltransferase) (NAT2), ATP-binding cassette, sub-family C (CFTR/MRP), member 6 (ABCC6), ATPase, Cu++ transporting, alpha polypeptide (ATP7A), cytochrome P450, family 4, subfamily B, polypeptide 1 (CYP4B1) and solute carrier family 10 (sodium/bile acid cotransporter family), member 2 (SLC10A2). Genotyping results between drug metabolizing enzymes and transporters (DMET) and direct sequencing showed >96% of concordance. These findings highlight the role that non-CYP450 metabolizing enzymes and transporters may have in the pharmacology of docetaxel and thalidomide.


Subject(s)
PPAR delta/genetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Sulfotransferases/genetics , Taxoids/therapeutic use , Thalidomide/therapeutic use , Adult , Aged , Docetaxel , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis/methods , Orchiectomy , Pharmacogenetics , Polymorphism, Single Nucleotide , Taxoids/adverse effects , Taxoids/pharmacokinetics , Thalidomide/adverse effects , Thalidomide/pharmacokinetics , Carbohydrate Sulfotransferases
5.
J Transl Sci ; 5(2)2019 Apr.
Article in English | MEDLINE | ID: mdl-30906574

ABSTRACT

INTRODUCTION: Tumors over-expressing the human epithelial receptor 2 (HER2) or exhibiting amplification or mutation of its proto-oncogene have a poorer prognosis. Using trastuzumab and/or other HER2 targeted therapies can increase overall survival in patients with HER2(+) tumors making it critical to accurately identify patients who may benefit. We report on a Phase 0 study of the imaging agent, 111In-CHX-A"-DTPA trastuzumab, in patients with known HER2 status to evaluate its safety and biodistribution and to obtain preliminary data regarding its ability to provide an accurate, whole-body, non-invasive means to determine HER2 status. METHODS: 111In-CHX-A"-DTPA trastuzumab was radiolabeled on-site and slowly infused into 11 patients who underwent single (n=5) or multiple (n=6) ɣ-camera (n=6) and/or SPECT (n=8) imaging sessions. RESULTS: No safety issues were identified. Visual and semi-quantitative imaging data were concordant with tissue HER2 expression profiling in all but 1 patient. The biodistribution showed intense peak liver activity at the initial imaging timepoint (3.3h) and a single-phase clearance fit of the average time-activity curve (TAC) estimated t1/2=46.9h (R2=0.97; 95%CI 41.8 to 53h). This was followed by high gastrointestinal (GI) tract activity peaking by 52h. Linear regression predicted GI clearance by 201.2h (R2 =0.96; 95%CI 188.5 to 216.9h). Blood pool had lower activity with its maximum on the initial images. Non-linear regression fit projected a t1/2=34.2h (R2 =0.96; 95%CI 25.3 to 46.3h). Assuming linear whole-body clearance, linear regression projected complete elimination (x-intercept) at 256.5hr (R2=0.96; 95%CI 186.1 to 489.2h). CONCLUSION: 111In-CHX-A"-DTPA trastuzumab can be safely imaged in humans. The biodistribution allowed for visual and semiquantitative analysis with results concordant with tissue expression profiling in 10 of 11 patients. Advances in Knowledge and Implications for Patient Care Using readily available components and on-site radiolabeling 111In-CHX-A"-DTPA trastuzumab SPECT imaging may provide an economical, non-invasive means to detect HER2 over-expression.

6.
Clin Pharmacol Ther ; 84(4): 526-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-19238658

ABSTRACT

The field of clinical pharmacology is the discipline engaged in optimal drug discovery, development, and use, based on an in-depth knowledge of human pharmacology and therapeutics. Although many think clinical pharmacology is simply the study of pharmacokinetics, pharmacodynamics, and pharmacogenetics, the role of the clinical pharmacologists covers all avenues of drug discovery, development, therapeutics, optimal drug therapy, and education. This expanded understanding is important as we start to address the overwhelming mandate and the unfortunate shortage of qualified investigators in the field (both physicians and non-physicians).


Subject(s)
Biomedical Research , Education, Pharmacy, Graduate/organization & administration , Pharmacology, Clinical , Schools, Pharmacy/organization & administration , Biomedical Research/organization & administration , Humans , United States , Workforce
7.
Br J Cancer ; 98(10): 1630-2, 2008 May 20.
Article in English | MEDLINE | ID: mdl-18475295

ABSTRACT

Administration of BAS 100, a novel mechanism-based CYP3A4 inhibitor isolated from grapefruit juice, resulted in a 2.1-fold increase in erlotinib exposure following oral administration to wild-type and humanized CYP3A4 transgenic mice. This study illustrates the potential of BAS 100 to increase the low and variable oral bioavailability of erlotinib in cancer patients.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors , Enzyme Inhibitors/pharmacology , Esters/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Quinazolines/pharmacokinetics , Spiro Compounds/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Beverages , Biological Availability , Citrus paradisi , Erlotinib Hydrochloride , Female , Mice , Mice, Inbred BALB C , Mice, Transgenic , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/blood , Quinazolines/administration & dosage , Quinazolines/blood , Random Allocation
8.
Clin Pharmacol Ther ; 81(1): 76-82, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17186002

ABSTRACT

To explore retrospectively the relationships between paclitaxel pharmacokinetics and three known, non-synonymous single-nucleotide polymorphisms (SNPs) in SLCO1B3, the gene encoding organic anion transporting polypeptide (OATP)1B3. Accumulation of [(3)H]paclitaxel was studied in Xenopus laevis oocytes injected with cRNA of Oatp1b2, OATP1A2, OATP1B1, OATP1B3, OAT1, OAT3, OCT1, and NTCP. The 334T>G (Ser112Ala), 699G>A (Met233Ile), and 1564G>T (Gly522Cys) loci of SLCO1B3 were screened in 475 individuals from five ethnic groups and 90 European Caucasian cancer patients treated with paclitaxel. Only OATP1B3 was capable of transporting paclitaxel to a significant extent (P=0.003). The 334T>G and 699G>A SNPs were less common in the African-American and Ghanaian populations (P<0.000001). Paclitaxel pharmacokinetics were not associated with the studied SNPs or haplotypes (P>0.3). The studied SNPs in SLCO1B3 appear to play a limited role in the disposition of paclitaxel, although their clinical significance in other ethnic populations remains to be investigated.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacokinetics , Organic Anion Transporters, Sodium-Independent/genetics , Paclitaxel/pharmacokinetics , Racial Groups , Animals , Dose-Response Relationship, Drug , Female , Genotype , Humans , In Vitro Techniques , Oocytes , Polymorphism, Single Nucleotide , Retrospective Studies , Solute Carrier Organic Anion Transporter Family Member 1B3 , Xenopus laevis
9.
Leukemia ; 19(7): 1253-61, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15858615

ABSTRACT

Thalidomide alone or in combination with steroids has significant activity in multiple myeloma (MM). However, given its teratogenic potential, analogs have been synthesized, retaining the anti-MM activity without these side effects. We examined the anti-MM activity of two thalidomide analogs, CPS11 and CPS49. Direct cytotoxicity of the drugs on myeloma cell lines and patient myeloma cells was examined using thymidine uptake. Tumor cell apoptosis was evaluated by flow cytometry as well as Western blotting for caspase and PARP cleavage. Cellular signaling events were examined by immunoblotting for phosphorylated proteins. Both drugs inhibit proliferation of several MM cell lines sensitive and resistant to conventional therapies. They decrease secretion of IL-6, IGF, and VEGF by marrow stromal cells. Importantly, they inhibit proliferation of MM cells adherent to stromal cells. These drugs induce caspase-mediated apoptosis in MM cell lines, as well as patient MM cells. They inhibit the PI3K/Akt and JAK/STAT (signal transducers and activators of transcription) pathways in MM cells and are antiangiogenic in matrigel-based assays. CPS11 and CPS49 have potent antimyeloma activity and can overcome protective effects of the tumor microenvironment. They have potent antiangiogenic activity and direct effect on bone marrow stroma. These encouraging preclinical data provide the basis for further evaluation in the clinic.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Multiple Myeloma/drug therapy , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Apoptosis/drug effects , Cell Adhesion/drug effects , Cell Line, Tumor , DNA/biosynthesis , DNA/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Stromal Cells/drug effects
10.
J Natl Cancer Inst ; 86(3): 222-7, 1994 Feb 02.
Article in English | MEDLINE | ID: mdl-7506794

ABSTRACT

BACKGROUND: The best treatment for patients with "hormone-refractory" metastatic prostate cancer is unclear, particularly in patients for whom suramin and hydrocortisone have failed. PURPOSE: We investigated a combination of flutamide withdrawal and aminoglutethimide in suramin- and hydrocortisone-pretreated patients with "hormone-refractory" prostate cancer. METHODS: Twenty-nine patients with metastatic prostate cancer were treated with simultaneous flutamide withdrawal and aminoglutethimide (250 mg given orally four times daily). All patients were taking flutamide at the time of entry, and previous treatments with medical or surgical castration, flutamide, suramin, and hydrocortisone had failed in all of these patients. Because of suramin-induced adrenal insufficiency, all patients had previously received, and continued to receive, physiological doses of hydrocortisone. Treatment of all non-surgically castrated patients had previously failed; however, these patients continued to receive depot leuprolide. RESULTS: In 14 (48%) of 29 patients, the prostate-specific antigen (PSA) decreased by more than 80% for 4 or more weeks. Improvements in anemia, thrombocytopenia, soft-tissue masses, bone scans, and symptoms were also noted. Factors associated with response included prolonged flutamide pretreatment, a markedly elevated pretreatment PSA, and the absence of soft-tissue disease. CONCLUSIONS: Flutamide withdrawal, when combined with the simultaneous administration of aminoglutethimide, is a therapeutically active approach in patients with "hormone-refractory" prostate cancer. IMPLICATIONS: On the basis of these and additional data, we hypothesize that prolonged exposure to flutamide results in the selective proliferation of cancer cells containing a mutant androgen receptor that aberrantly recognizes flutamide metabolites and nonandrogenic steroids as androgenic stimuli.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Flutamide/administration & dosage , Prostatic Neoplasms/drug therapy , Aged , Aged, 80 and over , Aminoglutethimide/administration & dosage , Humans , Male , Middle Aged , Neoplasm Metastasis , Prostate-Specific Antigen/blood , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Time Factors , Treatment Outcome
11.
Cancer Res ; 60(20): 5630-4, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11059752

ABSTRACT

Hypoxia limits tumor growth but selects for higher metastatic potential. We tested the functional activity of hypoxia-inducible factor-1 (HIF-1) in prostate cell lines ranging from normal epithelial cells (PrEC), hormone-dependent LNCaP, hormone-independent DU145, PC-3 to highly metastatic PC-3M cancer cell lines. We found that HIF-1-stimulated transcription was the lowest in PrEC and LNCaP cells and the highest in PC-3M cells. The induction by hypoxia of the HIF-1 dependent genes Cap43 and GAPDH was the highest in the most aggressive PC-3M cancer cells. Because these advanced prostate cancer cell lines have lost p53 function, this further shifts a balance from p53 to HIF-1 transcriptional regulation, and a high ratio of HIF-1-dependent:p53-dependent transcription was a marker of the advanced malignant phenotype. Transient transfection of HIF-1alpha expression vector induced transcription from p21 promoter construct in prostate cancer cell lines. Furthermore, hypoxia slightly induced p21 mRNA in these cells. However, neither expression of p21 nor hypoxia caused growth arrest in PC-3M cells. Therefore, high inducibility of HIF-1-dependent genes, loss of p53 functions with high ratio of HIF-1-dependent:p53-dependent transcription, and loss of sensitivity to p21 inhibition is a part of hypoxic phenotype associated with aggressive cancer behavior.


Subject(s)
Cyclins/physiology , DNA-Binding Proteins/genetics , Nuclear Proteins/genetics , Prostatic Neoplasms/genetics , Transcription Factors/genetics , Tumor Suppressor Protein p53/physiology , Cell Cycle/genetics , Cell Cycle Proteins , Cell Hypoxia/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/physiology , Flow Cytometry , Gene Expression Regulation, Neoplastic , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Intracellular Signaling Peptides and Proteins , Male , Neoplasm Metastasis , Nuclear Proteins/biosynthesis , Nuclear Proteins/physiology , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Biosynthesis , Proteins/genetics , Transcription Factors/biosynthesis , Transcription Factors/physiology , Transcription, Genetic/physiology , Transcriptional Activation/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
12.
Cancer Res ; 56(3): 569-73, 1996 Feb 01.
Article in English | MEDLINE | ID: mdl-8564973

ABSTRACT

Carboxyamido-triazole (CAI) is a synthetic inhibitor of non-excitable calcium channels that reversibly inhibits angiogenesis, tumor cell proliferation, and metastatic potential. Inhibition of calcium influx and calcium-dependent events is a potential common mechanism underlying these effects of CAI. The cytostatic and antiangiogenic properties of CAI led to its development for clinical investigation. In a Phase I clinical trial open to patients with refractory solid tumors, 49 patients received p.o. administered CAI daily or every other day. Two oral formulations, PEG-400 CAI solution and a gelatin capsule containing CAI in PEG-400, were tested. All administered dosages of CAI yielded plasma concentration at or above the range demonstrated to be effective in inhibiting signaling and cancer progression in vitro and in preclinical models (1 microgram/ml, 2.3 microM). Toxicity of p.o. administered CAI most commonly consisted of dose-related grade 1-2 nausea, vomiting, and occasional anorexia. CAI administration at bedtime ameliorated gastrointestinal complaints in many patients; others required addition of simple antiemetic regimens, usually consisting of metoclopropamide or prochlorperazine. Gastrointestinal complaints were the cause for compliance-limiting toxicity at 175 mg/m2/day of the liquid formulation and 125 mg/m2/day of the gelatin capsule formation. Reversible and rare sensory axonal neuropathy (grade 3, 1 patient) and neutropenia (grade 4, 1 patient) were dose-limiting toxicities observed at the 330 mg/m2 every-other-day liquid CAI dose level. No evidence of cumulative end organ damage or central nervous system injury was observed. Disease stabilization and improvement in performance status was observed. Disease stabilization and improvement in performance status was observed in 49% of evaluable patients who had disease progression before CAI. Disease stabilization and associated improvement in performance status was seen in patients with renal cell carcinoma (7 months), pancreaticobiliary carcinomas (3, 5, and 5 months), melanoma (7 months), ovarian cancer (7 months), and non-small cell lung cancer (3 months). The recommended Phase II doses from this trial are 150 mg/m2/day in the liquid formation and 100 mg/m2/day in the gelatin capsule formation.


Subject(s)
Antineoplastic Agents/therapeutic use , Calcium Channel Blockers/therapeutic use , Neoplasms/drug therapy , Triazoles/therapeutic use , Administration, Oral , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Calcium Channel Blockers/adverse effects , Calcium Channel Blockers/pharmacokinetics , Female , Humans , Male , Middle Aged , Neoplasms/metabolism , Triazoles/adverse effects , Triazoles/pharmacokinetics
13.
Cancer Res ; 54(7): 1690-4, 1994 Apr 01.
Article in English | MEDLINE | ID: mdl-8137283

ABSTRACT

Phenylacetate has recently been shown to suppress tumor growth and promote differentiation in experimental models. A phase I trial of phenylacetate was conducted in 17 patients with advanced solid tumors. Each patient received a single i.v. bolus dose followed by a 14-day continuous i.v. infusion of the drug. Twenty-one cycles of therapy were administered at four dose levels, achieved by increasing the rate of the continuous i.v. infusion. Phenylacetate displayed nonlinear pharmacokinetics [Km = 105.1 +/- 44.5 (SD) microgram/ml, Vmax = 24.1 +/- 5.2 mg/kg/h and Vd = 19.2 +/- 3.3 L]. There was also evidence for induction of drug clearance. Ninety-nine % of phenylacetate elimination was accounted for by conversion to phenylacetylglutamine, which was excreted in the urine. Continuous i.v. infusion rates resulting in serum phenylacetate concentrations exceeding Km often resulted in rapid drug accumulation and dose-limiting toxicity, which consisted of reversible central nervous system depression, preceded by emesis. Three of nine patients with metastatic, hormone-refractory prostate cancer maintained stable prostatic specific antigen levels for more than 2 months; another had less bone pain. One of six patients with glioblastoma multiforme, whose steroid dosage has remained unchanged for the duration of therapy, has sustained functional improvement for more than 9 months. The use of adaptive control with feedback for the dosing of each patient enabled us to safely maintain stable phenylacetate concentrations up to the range of 200-300 micrograms/ml, which resulted in clinical improvement in some patients with advanced disease.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Neoplasms/drug therapy , Phenylacetates/pharmacokinetics , Phenylacetates/toxicity , Adult , Aged , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Drug Administration Schedule , Female , Glioblastoma/drug therapy , Glioblastoma/pathology , Glutamine/blood , Humans , Infusions, Intravenous , Injections, Intravenous , Male , Middle Aged , Neoplasms/blood , Neoplasms/pathology , Phenylacetates/blood
14.
Cancer Res ; 58(15): 3248-53, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9699650

ABSTRACT

The pharmacokinetics of UCN-01 after administration as a 72- or 3-h infusion to cancer patients in initial Phase I trials displayed distinctive features that could not have been predicted from preclinical data. The distribution volumes (0.0796-0.158 liters/kg) and the systemic clearance (0.0407-0.252 ml/h/kg) were extremely low, in contrast to large distribution volume and rapid systemic clearance in experimental animals. The elimination half-lives (253-1660 h) were unusually long. In vitro protein binding experiments demonstrated that UCN-01 was strongly bound to human alpha1-acid glycoprotein. The results suggest that unusual pharmacokinetics of UCN-01 in humans could be due, at least in part, to its specifically high binding to alpha1-acid glycoprotein.


Subject(s)
Alkaloids/pharmacokinetics , Alkaloids/therapeutic use , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Orosomucoid/metabolism , Alkaloids/blood , Animals , Antineoplastic Agents/blood , Dogs , Drug Administration Schedule , Humans , Infusions, Intravenous , Male , Mice , Mice, Inbred BALB C , Protein Binding , Rats , Serum Albumin/metabolism , Staurosporine/analogs & derivatives , Substrate Specificity
15.
Clin Transl Oncol ; 18(7): 653-9, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26661112

ABSTRACT

Historically, patients diagnosed with metastatic pancreatic cancer have faced a grim prognosis. The survival benefit seen with systemic chemotherapies and even combinations thereof have been disappointing. However, growing data suggest that the microenvironment of pancreatic cancer may be contributing to this poor prognosis. This microenvironment has a dense fibrotic stroma, and is hypoxic and highly immunosuppressive, all of which pose barriers to treatment. Newer strategies looking to disrupt the fibrotic stroma, target hypoxic areas, and improve local immune responses in the tumor microenvironment are currently undergoing clinical evaluation and seem to offer great promise. In addition to these therapies, preclinical work evaluating novel cytotoxic agents including nanoparticles has also been encouraging. While much research still needs to be done, these strategies offer new hope for patients with pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/immunology , Pancreatic Neoplasms/immunology , Tumor Microenvironment/immunology , Carcinoma, Pancreatic Ductal/pathology , Humans , Pancreatic Neoplasms/pathology
16.
J Clin Oncol ; 19(2): 584-92, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11208854

ABSTRACT

PURPOSE: This phase I clinical trial was designed to determine the maximum-tolerated dose and dose-limiting toxicities of the matrix metalloproteinase (MMP) inhibitor COL-3 in patients with refractory solid tumors. PATIENTS AND METHODS: Thirty-five patients with different cancer types were enrolled. COL-3 doses were escalated from 36 mg/m2/d in successive cohorts of at least three patients. Circulating levels of MMP-2, MMP-9, vascular endothelial growth factor, and basic fibroblast growth factor were assessed during treatment. Pharmacokinetic parameters were assessed for single and multiple doses of drug. RESULTS: Cutaneous phototoxicity was dose-limiting at 98 mg/m2/d. With the use of prophylactic sunblock, COL-3 was well tolerated at 70 mg/m2/d. The dose of 36 mg/m2/d was well tolerated without the use of sunblock. Other toxicities that did not seem to be related to dose or pharmacokinetics included anemia, anorexia, constipation, dizziness, elevated liver function test results, fever, headache, heartburn, nausea, vomiting, peripheral and central neurotoxicities, fatigue, and three cases of drug-induced lupus. Disease stabilization for periods of 26+ months, 8 months, and 6 months were seen in hemangioendothelioma, Sertoli-Leydig cell tumor, and fibrosarcoma, respectively. There was a potentially statistically significant relationship between changes in plasma MMP-2 levels and cumulative doses of drug when progressive disease patients were compared with those with stable disease or toxicity (P = .042). CONCLUSION: COL-3 induced disease stabilization in several patients who had a nonepithelial type of malignancy. Phototoxicity was dose-limiting. We recommend the dose of 36 mg/m2/d for phase II trials.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Matrix Metalloproteinase Inhibitors , Neoplasms/drug therapy , Tetracyclines/therapeutic use , Adult , Aged , Antineoplastic Agents/pharmacokinetics , Endothelial Growth Factors/blood , Enzyme Inhibitors/pharmacokinetics , Female , Fibroblast Growth Factor 2/blood , Humans , Lymphokines/blood , Male , Matrix Metalloproteinase 2/blood , Matrix Metalloproteinase 9/blood , Matrix Metalloproteinases/blood , Middle Aged , Statistics, Nonparametric , Tetracyclines/pharmacokinetics , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
17.
J Clin Oncol ; 15(5): 1985-93, 1997 May.
Article in English | MEDLINE | ID: mdl-9164210

ABSTRACT

PURPOSE: Cytostatic agents targeted against angiogenesis and tumor cell invasive potential form a new class of investigational drugs. Orally administered carboxyamidotriazole (CAI) (NSC609974) is both antiangiogenic and antimetastatic. An encapsulated micronized powder formulation has been developed to optimize CAI administration. A phase I dose escalation trial with pharmacokinetic analysis has been performed. PATIENTS AND METHODS: Twenty-one patients with refractory solid tumors and good end organ function and performance status were enrolled onto the study. Patients received a test dose followed 1 week later by daily administration of CAI in the encapsulated micronized formulation at doses of 100 to 350 mg/m2. Patients remained on CAI until disease progression or dose-limiting toxicity. Plasma samples were taken to characterize the pharmacokinetic parameters of this formulation of CAI. RESULTS: All patients were assessable for toxicity and 18 were assessable for pharmacokinetics and response analysis. Grade 1 and 2 gastrointestinal side effects were observed in up to 50% of patients. Dose-limiting toxicity was observed in both patients treated at 350 mg/m2/d, consisting of reversible grade 2 to 3 cerebellar ataxia (n = 1) and confusion (n = 1). One minor response (MR) was observed in a patient with renal cell carcinoma and another nine patients had disease stabilization (MR + SD = 47%). Pharmacokinetic analysis demonstrated reduced bioavailability (58% reduction) compared with the PEG-400 liquid formulation previously reported. CONCLUSION: The better toxicity profile of encapsulated micronized CAI with similar frequency of disease stabilization and ease of administration compared with the liquid or gelatin capsule, suggests that the micronized formulation is a preferable formulation for subsequent studies. A dose of 300 mg/m2/d is proposed for phase II investigations.


Subject(s)
Antineoplastic Agents/administration & dosage , Neoplasms/drug therapy , Triazoles/administration & dosage , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Drug Administration Schedule , Drug Compounding , Female , Humans , Male , Middle Aged , Neoplasms/metabolism , Neoplasms/pathology , Triazoles/adverse effects , Triazoles/pharmacokinetics
18.
J Clin Oncol ; 13(9): 2223-9, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7666080

ABSTRACT

PURPOSE: To describe pharmacologic variables correlated with the development of neurologic toxicity in patients treated with suramin. METHODS: Eighty-one patients were treated with suramin in a phase I study. The rate of drug infusion was continuously adjusted to maintain a preassigned plasma suramin concentration (175, 215, or 275 micrograms/mL) for a fixed duration (2 to 8 weeks). RESULTS: Eight patients developed grade III/IV neurologic motor impairment (predominantly motor axonal polyneuropathy). All were treated at the 275-micrograms/mL concentration. One patient treated at the 215-micrograms/mL concentration developed grade II motor dysfunction. In addition, seven of nine patients had sensory symptoms. Pharmacologic variables associated with the development of polyneuropathy included total cumulative suramin dose, duration of exposure to plasma concentrations greater than 200 micrograms/mL, and area under the curve (AUC) greater than 200 micrograms/mL. CONCLUSION: Significant neurologic toxicity can result from therapy with suramin, even when dosing is designed to avoid exposure to plasma concentrations greater than 350 micrograms/mL. Future clinical trials of suramin should be designed in such a way as to limit the total cumulative dose to < or = 157 mg/kg given over a period of > or = 8 weeks, limit the period of exposure to plasma suramin concentrations greater than 200 micrograms/mL to < or = 25 days, and limit the AUC greater than 200 micrograms/mL to < or = 48,000 mg.h/AL.


Subject(s)
Nervous System Diseases/chemically induced , Suramin/adverse effects , Adult , Aged , Aged, 80 and over , Humans , Logistic Models , Male , Melanoma/blood , Melanoma/drug therapy , Middle Aged , Motor Neuron Disease/chemically induced , Multivariate Analysis , Prostatic Neoplasms/blood , Prostatic Neoplasms/drug therapy , Sarcoma/blood , Sarcoma/drug therapy , Suramin/administration & dosage , Suramin/pharmacokinetics
19.
J Clin Oncol ; 19(3): 832-42, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11157037

ABSTRACT

PURPOSE: PSC 833 (valspodar) is a second-generation P-glycoprotein (Pgp) antagonist developed to reverse multidrug resistance. We conducted a phase I study of a 7-day oral administration of PSC 833 in combination with paclitaxel, administered as a 96-hour continuous infusion. PATIENTS AND METHODS: Fifty patients with advanced cancer were enrolled onto the trial. PSC 833 was administered orally for 7 days, beginning 72 hours before the start of the paclitaxel infusion. Paclitaxel dose reductions were planned because of the pharmacokinetic interactions known to occur with PSC 833. RESULTS: In combination with PSC 833, maximum-tolerated doses were defined as paclitaxel 13.1 mg/m(2)/d continuous intravenous infusion (CIVI) for 4 days without filgrastim, and paclitaxel 17.5 mg/m(2)/d CIVI for 4 days with filgrastim support. Dose-limiting toxicity for the combination was neutropenia. Statistical analysis of cohorts revealed similar mean steady-state concentrations (C(pss)) and areas under the concentration-versus-time curve (AUCs) when patients received paclitaxel doses of 13.1 or 17.5 mg/m(2)/d for 4 days with PSC 833, as when they received a paclitaxel dose of 35 mg/m(2)/d for 4 days without PSC 833. However, the effect of PSC 833 on paclitaxel pharmacokinetics varied greatly among individual patients, although a surrogate assay using CD56+ cells suggested inhibition of Pgp was complete or nearly complete at low concentrations of PSC 833. Responses occurred in three of four patients with non-small-cell lung cancer, and clinical benefit occurred in five of 10 patients with ovarian carcinoma. CONCLUSION: PSC 833 in combination with paclitaxel can be administered safely to patients provided the paclitaxel dose is reduced to compensate for the pharmacokinetic interaction. Surrogate studies with CD56+ cells indicate that the maximum-tolerated dose for PSC 833 gives serum levels much higher than those required to block Pgp. The variability in paclitaxel pharmacokinetics, despite complete inhibition of Pgp in the surrogate assay, suggests that other mechanisms, most likely related to P450, contribute to the pharmacokinetic interaction. Future development of combinations such as this should include strategies to predict pharmacokinetics of the chemotherapeutic agent. This in turn will facilitate dosing to achieve comparable CPss and AUCs.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Neoplasms/drug therapy , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Administration, Oral , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , CD56 Antigen/biosynthesis , Cyclosporins/administration & dosage , Cyclosporins/adverse effects , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Fluorescent Dyes/pharmacokinetics , Humans , Infusions, Intravenous , Male , Middle Aged , Neoplasms/metabolism , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Paclitaxel/pharmacokinetics , Rhodamines/pharmacokinetics , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
20.
J Clin Oncol ; 15(4): 1470-7, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9193342

ABSTRACT

PURPOSE: To assess the efficacy and toxicity of suramin, hydrocortisone, leuprolide, and flutamide in previously untreated metastatic prostate cancer. PATIENTS AND METHODS: Patients with stage D2 and poor-prognosis stage D1 prostate cancer were given suramin on a pharmacokinetically derived dosing schedule to maintain suramin concentrations between 175 and 300 micrograms/mL. Additionally, all patients received flutamide 250 mg orally three times daily, initiated on day 1 and continued until disease progression; depot leuprolide 7.5 mg intramuscularly begun on day 5 and repeated every 4 weeks indefinitely; and replacement doses of hydrocortisone. RESULTS: Fifty patients were entered onto the study: 48 with stage D2 and two with stage D1 disease. The median age was 59 years (range, 42 to 79) and 31 patients had a Karnofsky performance status (KPS) of 100%. Forty-five patients had bone metastases and 25 had measurable soft tissue disease. Forty-one (82%) had severe disease. The overall response rate in 49 assessable patients was three complete responses (CRs) and 30 partial responses (PRs) for an overall response rate of 67%. Eighteen patients have died. The median survival time has not been reached, with a median potential follow-up duration of 44 months. Grade 3 to 4 toxicity was seen in 38% of patients and was predominantly hematologic and reversible. CONCLUSION: The high response rate and prolonged survival in a poor-prognosis group of patients with metastatic prostate cancer warrant a phase III randomized comparison of this regimen versus hormonal therapy alone. Toxicity was moderate and reversible.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Adult , Aged , Antineoplastic Agents, Hormonal/administration & dosage , Disease Progression , Drug Administration Schedule , Flutamide/administration & dosage , Humans , Leuprolide/administration & dosage , Male , Middle Aged , Prostate-Specific Antigen/blood , Prostatic Neoplasms/immunology , Suramin/administration & dosage , Survival Analysis , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL