Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Benjamin Winograd is active.

Publication


Featured researches published by Benjamin Winograd.


European Journal of Cancer and Clinical Oncology | 1991

Comparison of the sulforhodamine B protein and tetrazolium (MTT) assays for in vitro chemosensitivity testing

Yvonne P. Keepers; Paulo E. Pizao; Godefridus J. Peters; Jannette van Ark-Otte; Benjamin Winograd

The sulforhodamine B (SRB) protein stain assay was compared with the tetrazolium (MTT) colorimetric assay for in vitro chemosensitivity testing of various human tumour cell lines. The SRB assay provided a better linearity with cell number and a higher sensitivity, and its staining was not cell-line dependent. In contrast to the MTT assay, the SRB assay stained recently lysed cells. Cell debris, however, was not stained by SRB and therefore the drug sensitivity data were not affected.


Cancer Chemotherapy and Pharmacology | 1999

Promising new developments in cancer chemotherapy.

Karen Ferrante; Benjamin Winograd; Renzo Canetta

Abstract The positive impact on survival of traditional chemotherapeutic agents has renewed interest in developing newer cytotoxic agents and orally active compounds with improved therapeutic indices. In addition, new insights into the pathways of human tumorigenesis have led to novel approaches aimed at specific mechanism-based targets. The taxane class, of which paclitaxel was the first member, has the unique ability to promote and stabilize microtubule function directly, thereby inhibiting mitotic progression and inducing apoptotic cell death. Paclitaxel provides treatment benefit in a broad range of solid tumors including breast, ovarian, and lung cancer. The success with paclitaxel stimulated interest in the microtubule as a new therapeutic target. Taxane analogues with improved preclinical efficacy have been identified and are entering clinical trials. The enthusiasm for oral anticancer agents and the therapeutic importance of platinum compounds has led to the development of JM216 (satraplatin), a novel platinum IV coordination complex with oral activity in cisplatin-resistant cell lines, which is now in phase III trials in prostate cancer. Another compound in late development is DPPE, a chemopotentiator that enhances the in vivo antitumor effects of cytotoxic agents such as doxorubicin, cyclophosphamide, and cisplatin. Agents that inhibit topoisomerase I and II have also been of interest. TAS-103 is a dual topoisomerase I and II inhibitor with preclinical efficacy in a broad spectrum of tumors and in multidrug-resistant tumor cell lines. Vaccination strategies represent a rational therapeutic approach in the minimal residual disease or high-risk adjuvant therapy setting. The GMK and MGV vaccines utilizing ganglioside antigens overexpressed on human tumors such as melanoma and small cell lung cancer appear to induce antibody production reliably at tolerable doses and are under further clinical investigation. Inhibition of matrix metalloproteinases (MMPs) is another attractive target for intervention in several aspects of tumor progression. Local production of MMPs with subsequent degradation of the extracellular matrix is implicated in supporting tumor growth, invasion, and angiogenesis. The development of orally active, nontoxic MMP inhibitors is critical since these compounds will likely require chronic administration in conjunction with other therapies. Oncogenes and tumor suppressor genes are appealing targets for therapy since they are thought to be responsible for a significant number of cancers. Mutations in the Ras oncogene occur with great frequency in a number of human cancers including lung, pancreas, and colon cancer. Clinical development of potent and selective inhibitors of farnesyltransferase, the Ras-processing enzyme, is ongoing. These compounds uncouple Ras activity, affect tumor growth, and have demonstrated significant antitumor activity against experimental models of human cancer. The exciting compounds and novel therapeutic approaches currently under investigation by Bristol-Myers Squibb Pharmaceutical Research Institute offer great potential as effective cancer chemotherapy agents for the near future.


Cancer Chemotherapy and Pharmacology | 1990

Phase I study of Brequinar sodium (NSC 368390) in patients with solid malignancies

Gilberto Schwartsmann; Pierre Dodion; Jan B. Vermorken; Wim W. ten Bokkel Huinink; Jean Joggi; Benjamin Winograd; Helen Gall; Godeleine Simonetti; Wim J. F. van der Vijgh; Maike B. van Hennik; Nadine Crespeigne

SummaryBrequinar sodium (DUP 785, NSC 368390) is a novel quinoline-carboxylic acid derivative that has been selected for clinical evaluation because of its broad spectrum of antitumor activity in animal models and its novel chemical structure. This compound inhibits the mitochondrial enzyme dihydroorotate dehydrogenase (DHO-DH), which catalyzes the conversion of dihydroorotate to orotate, leading to a blockage in the pyrimidine de novo biosynthesis. A total of 43 patients received 110 courses of Brequinar sodium by short-term intravenous (i. v.) infusion, which was repeated every 3 weeks. Dose escalation was initially based on a modified Fibonacci scheme. After pharmacokinetic data from mice and man became available, a pharmacologically guided dose escalation was used; at toxic levels, dose escalation was applied on the basis of clinical judgement. The dose-limiting toxicities were myelosuppression, mucositis, skin rash, nausea and vomiting. The maximum tolerable doses for poor- and good-risk patients were 1,500 and 2,250 mg/m2, respectively. One mixed response was observed in a patient with papilary carcinoma of the thyroid. The recommended doses for phase II studies are 1,200 and 1,800 mg/m2 Brequinar sodium, given by a 1-h i.v. infusion every 3 weeks to poor- and good-risk patients, respectively.


Journal of Pharmacokinetics and Biopharmaceutics | 1996

Bioequivalence assessment of etoposide phosphate and etoposide using pharmacodynamic and traditional pharmacokinetic parameters

Vanaja Mummaneni; Sanjeev Kaul; Linus N. Igwemezie; David R. Newell; David Porter; Huw D. Thomas; A. Hilary Calvert; Benjamin Winograd; Rashmi H. Barbhaiya

The bioequivalence of etoposide phosphate, a prodrug of etoposide, to etoposide was assessed in a randomized, crossover study in 29 patients with histologically established solid tumors that had failed conventional treatment. Cohorts of patients received one treatment course each of etoposide and etoposide phosphate which consisted of a 100 mg/m2 per day etoposide equivalent dose infused iv over 1 hr on a Day 1 to 5 schedule of treatment. The second course was administered 21 days later or on recovery of blood cell counts. Plasma and urine samples were collected over 24 hr on Day 1 of each course and assayed for etoposide content by a validated HPLC/UV method. Resulting data were subjected to noncompartmental pharmacokinetic analysis. Hematology profiles were obtained by collecting blood samples prior to the first course and twice a week after each course. The pharmacodynamics and pharmacokinetics of etoposide were virtually identical after the two treatments. The point estimates (90% confidence intervals) for nadir WBC, granulocytes, hemoglobin, and platelets expressed as % decrease from the baseline, and for the pharmacokinetic parameters, Cmax, and AUC0-∞, after intravenous etoposide phosphate relative to etoposide were 100% (96%, 105%), 97% (91%, 103%), 95% (82%, 109%), 95% (84%, 106%), 107% (101%, 113%), and 113% (107%, 119%), respectively. Therefore, etoposide phosphate is bioequivalent to etoposide based on pharmacokinetic and pharmacodynamic assessments.


European Journal of Cancer and Clinical Oncology | 1989

Pharmacokinetics of Brequinar sodium (NSC 368390) in patients with solid tumors during a phase I study

G. Schwartsmann; W.J.F. van der Vijgh; M.B. van Hennik; I. Klein; Jan B. Vermorken; P. Dodion; W.W. ten Bokkel Huinink; G. Joggi; Helen Gall; Nadine Crespeigne; G. Simonetti; Benjamin Winograd; H.M. Pinedo

The pharmacokinetics of the novel antipyrimidine agent Brequinar sodium (NSC 368390; DUP 785) was studied in 23 patients with solid tumors during the phase I study of this compound. The drug was administered by short-term (10-60 min) intravenous infusion every 3 weeks. The doses ranged from 15 to 2250 mg/m2. At doses higher than 1500 mg/m2 the areas under the plasma concentration vs. time curve (AUC) increased non-proportionally, while the total body clearance (Clt) dropped substantially, indicating non-linear pharmacokinetics of the drug. Brequinar sodium showed a triphasic decay of plasma concentrations with half-life ranges of 11.1-36.6 min, 1.7-6.9 h and 12.5-25.0 h, respectively. The volume of distribution (Vdss) ranged from 4.4 to 10.6 l/m2. The total body clearance (Clt) ranged from 6.9 to 22.1 ml/min with a small contribution of the renal clearance (0.04-0.4 ml/min). Up to 7 days, the cumulative urinary excretion (CUE) and the cumulative fecal excretion (CFE) ranged from 0.4 to 8.3% and from 7.7 to 18.3% of the dose, respectively. There was evidence for the presence of drug metabolites in urine and feces. There was no drug accumulation with repeated administration of Brequinar sodium by the above mentioned drug schedule. The ratio between the plasma AUC at the maximum tolerable dose (MTD) in man and that at the mouse LD10 was 0.8, while the ratio between the respective doses was 5.7. The ratios between the AUC in patients and that at the mouse LD10 were applied to guide dose escalation in the phase I study. The results of the above mentioned pharmacokinetic studies were useful for the choice of an optimal schedule for phase II trials of Brequinar sodium.


Cancer Chemotherapy and Pharmacology | 1987

Pharmacokinetics and toxicology of sparsomycin in beagle dogs

Zbigniew Zylicz; D. J. Theo Wagener; Pilar Fernandez del Moral; Helga van Rennes; J. M. C. Wessels; Benjamin Winograd; Eppo van der Kleijn; T. B. Vree; Urbanus van Haelst; Leon A. G. M. van den Broek; Harry C. J. Ottenheijm

SummarySparsomycin is a cytotoxic drug exhibiting a broad spectrum of in vitro activity against murine tumors and many tumor cell lines. It also appears to be a potent stimulator of the antitumor activity of cisplatin against L1210 leukemia in vivo. However, because of its toxicity, the antitumor activity of sparsomycin on murine tumors in vivo has been disappointing. The purpose of our study was to investigate the pharmacokinetics of this drug as well as the possible mechanisms that produce sparsomycin toxicity. Tests on beagle dogs revealed that about 60% of the drug is eliminated by metabolic clearance, while 40% is eliminated by the kidneys. After a single bolus injection of 0.1 mg/kg sparsomycin without narcosis, sparsomycin was eliminated with a tβ1/2 of 0.6–0.7 h, the AUC being 0.32–0.38 mg·h·1-1, and the volume of distribution (Vd) 0.26 l/kg. In addition to being subject to glomerular filtration, sparsomycin is probably also actively excreted and actively reabsorbed by the renal tubuli. Sparsomycin itself may inhibit its active tubular excretion, thus resulting in a decrease in the drugs renal clearance and its accumulation in the plasma. Sparsomycin appeared to be toxic primarily in the liver, disturbing its function and the synthesis of plasma proteins. Two out of five dogs developed hemorrhagic diathesis due to hypofibrinogenemia and deficiency of other blood-coagulation factors. Sparsomycin was not toxic to the bone marrow.


British Journal of Cancer | 1998

Phase I study of the mitomycin C analogue BMS-181174

Valentine M. Macaulay; Kenneth J. O'Byrne; John Green; P. A. Philip; L. McKinley; F. P. LaCreta; Benjamin Winograd; Trivadi S. Ganesan; Adrian L. Harris; Denis C. Talbot

BMS-181174 is an aminodisulphide derivative of Mitomycin C (MMC) with activity against a range of tumour cell lines and xenografts, including MMC-resistant tumours. In a phase I study of 82 patients with confirmed malignancy, we administered BMS-181174 at doses of 0.8-75 mg m(-2) by intravenous injection every 28 days. At least three patients were evaluated at each dose level, and 174 courses were administered. The pharmacokinetics were dose linear at BMS-181174 doses of 11.5-75 mg m(-2) and the drug appeared to undergo wide distribution. The maximum-tolerated dose was 65 mg m(-2) in previously treated patients and 75 mg m(-2) in chemotherapy-naive cases. The dose-limiting toxicity was myelosuppression, particularly thrombocytopenia, which was prolonged and cumulative. Three patients treated at 65-75 mg m(-2) died suddenly with evidence of pneumonia/pneumonitis, thought to be drug-related. Other toxicities included thrombophlebitis, possible cardiotoxicity (asymptomatic, reversible decline in left ventricular function) and renal impairment. The partial response rate was 5% (4 out of 82) overall, and 9% (3 out of 32) in patients treated at 65-75 mg m(-2). Responses occurred in treated and previously-untreated patients, including cases of colorectal cancer, non-small-cell lung cancer, ovarian cancer and adenocarcinoma of unknown primary site. BMS-181174 has anti-cancer activity but, because of its toxicity, particularly pneumonitis and thrombophlebitis, no phase II studies are planned.


Cancer Research | 1987

Comparative Pharmacokinetics of Cisplatin and Three Analogues in Mice and Humans

M.B. van Hennik; W.J.F. van der Vijgh; I. Klein; F. Elferink; J.B. Vermorken; Benjamin Winograd; H.M. Pinedo


Seminars in Oncology | 1995

High dose Paclitaxel with Granulocyte Colony-stimulating factor in patients with advanced breast cancer Refractory to Anthracycline Therapy: A European Cancer Centre Trial

J.B. Vermorken; W.W. ten Bokkel Huinink; I. A. M. Mandjes; T.J. Postma; M. T. Huizing; J.J. Heimans; Jos H. Beijnen; F. Bierhorst; H.M. Pinedo; Benjamin Winograd


British Journal of Cancer | 1992

Growth, morphology and chemosensitivity studies on postconfluent cells cultured in 'V'-bottomed microtiter plates.

Paulo E. Pizao; Donacian M. Lyaruu; Godefridus J. Peters; J. Van Ark-Otte; Benjamin Winograd; Guiseppe Giaccone

Collaboration


Dive into the Benjamin Winograd's collaboration.

Top Co-Authors

Avatar

H.M. Pinedo

VU University Amsterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Helen Gall

VU University Amsterdam

View shared research outputs
Top Co-Authors

Avatar

I. Klein

University of Amsterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

F. Elferink

VU University Amsterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

G. Simonetti

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Godefridus J. Peters

VU University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge