29 results on '"Rubin E"'
Search Results
2. Synthetic lethality-mediated precision oncology via the tumor transcriptome.
- Author
-
Lee JS, Nair NU, Dinstag G, Chapman L, Chung Y, Wang K, Sinha S, Cha H, Kim D, Schperberg AV, Srinivasan A, Lazar V, Rubin E, Hwang S, Berger R, Beker T, Ronai Z, Hannenhalli S, Gilbert MR, Kurzrock R, Lee SH, Aldape K, and Ruppin E
- Subjects
- Aged, Biomarkers, Tumor antagonists & inhibitors, Biomarkers, Tumor immunology, Clinical Trials as Topic, Female, Follow-Up Studies, Humans, Immunotherapy, Male, Neoplasms genetics, Neoplasms pathology, Prognosis, Prospective Studies, Retrospective Studies, Survival Rate, Biomarkers, Tumor genetics, Gene Expression Regulation, Neoplastic drug effects, Molecular Targeted Therapy, Neoplasms drug therapy, Precision Medicine, Synthetic Lethal Mutations, Transcriptome drug effects
- Abstract
Precision oncology has made significant advances, mainly by targeting actionable mutations in cancer driver genes. Aiming to expand treatment opportunities, recent studies have begun to explore the utility of tumor transcriptome to guide patient treatment. Here, we introduce SELECT (synthetic lethality and rescue-mediated precision oncology via the transcriptome), a precision oncology framework harnessing genetic interactions to predict patient response to cancer therapy from the tumor transcriptome. SELECT is tested on a broad collection of 35 published targeted and immunotherapy clinical trials from 10 different cancer types. It is predictive of patients' response in 80% of these clinical trials and in the recent multi-arm WINTHER trial. The predictive signatures and the code are made publicly available for academic use, laying a basis for future prospective clinical studies., Competing Interests: Declaration of interests E.R. is a co-founder of Medaware, Metabomed, and Pangea Therapeutics (divested from the latter). E.R. serves as a non-paid scientific consultant to Pangea Therapeutics, a company developing a precision oncology SL-based multi-omics approach. J.S.L. is a scientific consultant; T.B. is chief executive officer and chief technical officer; G.D. is head of research and development; R.B. is a member of the Scientific Advisory Board; and Z.R. is a co-founder and a scientific advisor at Pangea Therapeutics. R.K. receives research funding from Genentech, Merck Serono, Pfizer, Boehringer Ingelheim, TopAlliance, Takeda, Incyte, Debiopharm, Medimmune, Sequenom, Foundation Medicine, Konica Minolta, Grifols, Omniseq, and Guardant; received consultant, speaker, and/or advisory board fees for X-Biotech, Neomed, Pfizer, Actuate Therapeutics, Roche, Turning Point Therapeutics, TD2/Volastra, and Bicara Therapeutics; has an equity interest in IDbyDNA and CureMatch; serves on the board of CureMatch and CureMetrix; and is a co-founder of CureMatch. A patent application associated with this manuscript is in process., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
3. Genomic and transcriptomic profiling expands precision cancer medicine: the WINTHER trial.
- Author
-
Rodon J, Soria JC, Berger R, Miller WH, Rubin E, Kugel A, Tsimberidou A, Saintigny P, Ackerstein A, Braña I, Loriot Y, Afshar M, Miller V, Wunder F, Bresson C, Martini JF, Raynaud J, Mendelsohn J, Batist G, Onn A, Tabernero J, Schilsky RL, Lazar V, Lee JJ, and Kurzrock R
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biomarkers, Tumor genetics, Combined Modality Therapy, Female, Gene Expression Profiling, Genomics, High-Throughput Nucleotide Sequencing, Humans, Kaplan-Meier Estimate, Lung Neoplasms genetics, Lung Neoplasms therapy, Male, Middle Aged, Molecular Targeted Therapy, Precision Medicine, Progression-Free Survival, Sequence Analysis, DNA, Neoplasms genetics, Neoplasms therapy
- Abstract
Precision medicine focuses on DNA abnormalities, but not all tumors have tractable genomic alterations. The WINTHER trial ( NCT01856296 ) navigated patients to therapy on the basis of fresh biopsy-derived DNA sequencing (arm A; 236 gene panel) or RNA expression (arm B; comparing tumor to normal). The clinical management committee (investigators from five countries) recommended therapies, prioritizing genomic matches; physicians determined the therapy given. Matching scores were calculated post-hoc for each patient, according to drugs received: for DNA, the number of alterations matched divided by the total alteration number; for RNA, expression-matched drug ranks. Overall, 303 patients consented; 107 (35%; 69 in arm A and 38 in arm B) were evaluable for therapy. The median number of previous therapies was three. The most common diagnoses were colon, head and neck, and lung cancers. Among the 107 patients, the rate of stable disease ≥6 months and partial or complete response was 26.2% (arm A: 23.2%; arm B: 31.6% (P = 0.37)). The patient proportion with WINTHER versus previous therapy progression-free survival ratio of >1.5 was 22.4%, which did not meet the pre-specified primary end point. Fewer previous therapies, better performance status and higher matching score correlated with longer progression-free survival (all P < 0.05, multivariate). Our study shows that genomic and transcriptomic profiling are both useful for improving therapy recommendations and patient outcome, and expands personalized cancer treatment.
- Published
- 2019
- Full Text
- View/download PDF
4. Molecular Profiling of Cohorts of Tumor Samples to Guide Clinical Development of Pembrolizumab as Monotherapy.
- Author
-
Ayers M, Nebozhyn M, Cristescu R, McClanahan TK, Perini R, Rubin E, Cheng JD, Kaufman DR, and Loboda A
- Subjects
- Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized adverse effects, Antineoplastic Agents, Immunological administration & dosage, Antineoplastic Agents, Immunological adverse effects, B7-H1 Antigen genetics, B7-H1 Antigen metabolism, Clinical Decision-Making, Databases, Genetic, Disease Management, Gene Expression Regulation, Neoplastic drug effects, Humans, Mutation, Neoplasms pathology, Research Design, Transcriptome, Treatment Outcome, United States, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Agents, Immunological therapeutic use, Biomarkers, Tumor, Gene Expression Profiling, Neoplasms drug therapy, Neoplasms genetics
- Abstract
Purpose: Molecular profiling of large databases of human tumor gene expression profiles offers novel opportunities for informing decisions in clinical development programs., Experimental Design: Gene expression profile of programmed death ligand 1 (PD-L1) was explored in a dataset of 16,000 samples, including approximately 4,000 metastatic tumors, across >25 tumor types prevalent in the United States, looking for new indications for the programmed death 1 (PD-1) inhibitor pembrolizumab. PD-L1 expression was highly concordant with several genomic signatures indicative of immune-inflamed tumor microenvironment. Prevalence of activated immune-inflamed tumors across all tumor types was explored and used to rank tumor types for potential response to pembrolizumab monotherapy., Results: The analysis yielded 3 tiers of indications in which high levels of PD-L1 and immune-inflamed signatures were found in up to 40% to 60%, 20% to 40%, and 0% to 20% of tumors. Tier 1 contained novel indications known at the time of analysis to be responsive to PD-1 checkpoint blockade in the clinic (such as melanoma and non-small cell lung cancer), as well as indications not studied in the clinic previously, including microsatellite instability-high colorectal, head and neck, bladder, and triple-negative breast cancers. Complementary analysis of an Asian/Pacific cancer dataset (gastric cancer) revealed high prevalence of immune-inflamed tumors in gastric cancer. These data contributed to prioritization of these indications for clinical development of pembrolizumab as monotherapy., Conclusions: Data highlight the value of molecular profiling in identifying populations with high unmet needs with potentially favorable response characteristics and accelerating development of novel therapies for these patients. See related commentary by Mansfield and Jen, p. 1443 ., (©2018 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
5. NKp44 and NKp30 splice variant profiles in decidua and tumor tissues: a comparative viewpoint.
- Author
-
Shemesh A, Kugel A, Steiner N, Yezersky M, Tirosh D, Edri A, Teltsh O, Rosental B, Sheiner E, Rubin E, Campbell KS, and Porgador A
- Subjects
- Abortion, Spontaneous immunology, Abortion, Spontaneous pathology, Decidua immunology, Decidua pathology, Female, Flow Cytometry, Humans, Immune Privilege, Interleukin-15 metabolism, Interleukin-18 metabolism, Killer Cells, Natural immunology, Killer Cells, Natural metabolism, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Natural Cytotoxicity Triggering Receptor 2 immunology, Neoplasms genetics, Neoplasms immunology, Neoplasms pathology, Pre-Eclampsia immunology, Pre-Eclampsia pathology, Pregnancy, Transforming Growth Factor beta metabolism, Tumor Microenvironment immunology, Decidua metabolism, Natural Cytotoxicity Triggering Receptor 2 genetics, Natural Cytotoxicity Triggering Receptor 3 genetics, Neoplasms metabolism, RNA Splicing
- Abstract
NKp44 and NKp30 splice variant profiles have been shown to promote diverse cellular functions. Moreover, microenvironment factors such as TGF-β, IL-15 and IL-18 are able to influence both NKp44 and NKp30 splice variant profiles, leading to cytokine-associated profiles. Placenta and cancerous tissues have many similarities; both are immunologically privileged sites and both share immune tolerance mechanisms to support tissue development. Therefore, we studied the profiles of NKp44 and NKp30 splice variants in these states by comparing (i) decidua from pregnancy disorder and healthy gestation and (ii) matched normal and cancer tissue. Decidua samples had high incidence of both NKp44 and NKp30. In cancerous state it was different; while NKp30 expression was evident in most cancerous and matched normal tissues, NKp44 incidence was lower and was mostly associated with the cancerous tissues. A NKp44-1dominant inhibitory profile predominated in healthy pregnancy gestation. Interestingly, the NKp44-2/3 activation profile becomes the leading profile in spontaneous abortions, whereas balanced NKp44 profiles were observed in preeclampsia. In contrast, a clear preference for the NKp30a/b profile was evident in the 1st trimester decidua, yet no significant differences were observed for NKp30 profiles between healthy gestation and spontaneous abortions/preeclampsia. Both cancerous and matched normal tissues manifested balanced NKp30c inhibitory and NKp30a/b activation profiles with a NKp44-1dominant profile. However, a shift in NKp30 profiles between matched normal and cancer tissue was observed in half of the cases. To summarize, NKp44 and NKp30 splice variants profiles are tissue/condition specific and demonstrate similarity between placenta and cancerous tissues.
- Published
- 2016
- Full Text
- View/download PDF
6. Challenges in initiating and conducting personalized cancer therapy trials: perspectives from WINTHER, a Worldwide Innovative Network (WIN) Consortium trial.
- Author
-
Rodon J, Soria JC, Berger R, Batist G, Tsimberidou A, Bresson C, Lee JJ, Rubin E, Onn A, Schilsky RL, Miller WH, Eggermont AM, Mendelsohn J, Lazar V, and Kurzrock R
- Subjects
- Antineoplastic Agents economics, Antineoplastic Agents supply & distribution, Canada, Clinical Trials as Topic economics, Clinical Trials as Topic legislation & jurisprudence, France, Gene Expression Profiling, Genomics, Humans, Israel, Neoplasms metabolism, Prospective Studies, Spain, United States, Antineoplastic Agents therapeutic use, Clinical Trials as Topic methods, Molecular Targeted Therapy methods, Neoplasms drug therapy, Precision Medicine methods
- Abstract
Advances in 'omics' technology and targeted therapeutic molecules are together driving the incorporation of molecular-based diagnostics into the care of patients with cancer. There is an urgent need to assess the efficacy of therapy determined by molecular matching of patients with particular targeted therapies. WINTHER is a clinical trial that uses cutting edge genomic and transcriptomic assays to guide treatment decisions. Through the lens of this ambitious multinational trial (five countries, six sites) coordinated by the Worldwide Innovative Networking Consortium for personalized cancer therapy, we discovered key challenges in initiation and conduct of a prospective, omically driven study. To date, the time from study concept to activation has varied between 19 months at Gustave Roussy Cancer Campus in France to 30 months at the Segal Cancer Center, McGill University (Canada). It took 3+ years to be able to activate US sites due to national regulatory hurdles. Access to medications proposed by the molecular analysis remains a major challenge, since their availability through active clinical trials is highly variable over time within sites and across the network. Rules regarding the off-label use of drugs, or drugs not yet approved at all in some countries, pose a further challenge, and many biopharmaceutical companies lack a simple internal mechanism to supply the drugs even if they wish to do so. These various obstacles should be addressed to test and then implement precision medicine in cancer., (© The Author 2015. Published by Oxford University Press on behalf of the European Society for Medical Oncology. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2015
- Full Text
- View/download PDF
7. Phase I/IIa trial of the mammalian target of rapamycin inhibitor ridaforolimus (AP23573; MK-8669) administered orally in patients with refractory or advanced malignancies and sarcoma.
- Author
-
Mita MM, Poplin E, Britten CD, Tap WD, Rubin EH, Scott BB, Berk L, Rivera VM, Loewy JW, Dodion P, Haluska F, Sarantopoulos J, Mita A, and Tolcher A
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Drug Administration Schedule, Female, Humans, Male, Maximum Tolerated Dose, Middle Aged, Neoplasm Staging, Neoplasms pathology, Sarcoma pathology, Sirolimus administration & dosage, Sirolimus adverse effects, Sirolimus antagonists & inhibitors, Sirolimus pharmacokinetics, TOR Serine-Threonine Kinases antagonists & inhibitors, Treatment Outcome, Neoplasms drug therapy, Sarcoma drug therapy, Sirolimus analogs & derivatives, TOR Serine-Threonine Kinases metabolism
- Abstract
Background: Ridaforolimus is an inhibitor of mTOR with evidence of antitumor activity in an I.V. formulation. This multicenter, open-label, 3 + 3 design nonrandomized, dose-escalation, phase I/IIa trial was conducted to determine the safety, pharmacokinetic (PK) and pharmacodynamic parameters, maximum tolerated dose, and antitumor activity of oral ridaforolimus., Patients and Methods: Patients with metastatic or unresectable solid tumors refractory to therapy were eligible. Seven different continuous and intermittent dosing regimens were examined., Results: One hundred and forty-seven patients were enrolled in this study among which 85 were patients with sarcoma. Stomatitis was the most common DLT observed. The dosing regimen, 40 mg QD × 5 days/week, provided the best combination of cumulative dose, dose density, and cumulative exposure, and was the recommended dosing regimen for subsequent clinical development. PK was nonlinear, with less than proportional increases in day-1 blood AUC0-∞ and Cmax, particularly with doses >40 mg. The terminal half-life estimate of ridaforolimus (QD × 5 40 mg) was 42.0 h, and the mean half-life ∼30-60 h. The clinical benefit rate, (complete response, partial response, or stable disease for ≥4 months was 24.5% for all patients and 27.1% for patients with sarcoma., Conclusion: Oral ridaforolimus had an acceptable safety profile and exhibited antitumor activity in patients with sarcoma and other malignancies. ClinicalTrials.gov Identifier NCT00112372.
- Published
- 2013
- Full Text
- View/download PDF
8. A novel "reactomics" approach for cancer diagnostics.
- Author
-
Kolusheva S, Yossef R, Kugel A, Hanin-Avraham N, Cohen M, Rubin E, and Porgador A
- Subjects
- Humans, Neoplasms blood, Neoplasms metabolism, Spectrometry, Fluorescence, Neoplasms diagnosis
- Abstract
Non-invasive detection and monitoring of lethal diseases, such as cancer, are considered as effective factors in treatment and survival. We describe a new disease diagnostic approach, denoted "reactomics", based upon reactions between blood sera and an array of vesicles comprising different lipids and polydiacetylene (PDA), a chromatic polymer. We show that reactions between sera and such a lipid/PDA vesicle array produce chromatic patterns which depend both upon the sera composition as well as the specific lipid constituents within the vesicles. The chromatic patterns were processed through machine-learning algorithms, and the bioinformatics analysis could distinguish both between cancer-bearing and healthy patients, respectively, as well between two types of cancers. Size-separation and enzymatic digestion experiments indicate that lipoproteins are the primary components in sera which react with the chromatic biomimetic vesicles. This colorimetric reactomics concept is highly generic, robust, and does not require a priori knowledge upon specific disease markers in sera. Therefore, it could be employed as complementary or alternative approach for disease diagnostics.
- Published
- 2012
- Full Text
- View/download PDF
9. Deficiency of the dual ubiquitin/SUMO ligase Topors results in genetic instability and an increased rate of malignancy in mice.
- Author
-
Marshall H, Bhaumik M, Aviv H, Moore D, Yao M, Dutta J, Rahim H, Gounder M, Ganesan S, Saleem A, and Rubin E
- Subjects
- Animals, Chromobox Protein Homolog 5, Fibroblasts, Genomic Instability, Heterozygote, Histone Deacetylase Inhibitors, Humans, Hydroxamic Acids, Mice, Small Ubiquitin-Related Modifier Proteins metabolism, Ubiquitin-Protein Ligases genetics, Neoplasms genetics, Tumor Suppressor Proteins metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
Background: Topors is a nuclear protein that co-localizes with promyelocytic leukemia bodies and has both ubiquitin and SUMO E3 ligase activity. Expression studies implicated Topors as a tumor suppressor in various malignancies. To gain insight into the function of Topors, we generated a Topors-deficient mouse strain., Results: Mice homozygous for a mutant Topors allele exhibited a high rate of perinatal mortality and decreased lifespan. In addition, heterozygotes were found to have an increased incidence of malignancy, involving a variety of tissues. Consistent with this finding, primary embryonic fibroblasts lacking Topors exhibited an increased rate of malignant transformation, associated with aneuploidy and defective chromosomal segregation. While loss of Topors did not alter sensitivity to DNA-damaging or microtubule-targeting agents, cells lacking Topors exhibited altered pericentric heterochromatin, manifested by mislocalization of HP1alpha and an increase in transcription from pericentric major satellite DNA. Topors-deficient cells exhibited a transcriptional profile similar to that of cells treated with histone deacetylase inhibitors, and were resistant to the anti-proliferative effects of the histone deacetylase inhibitor trichostatin A., Conclusion: These results indicate a unique role for Topors in the maintenance of genomic stability and pericentric heterochromatin, as well as in cellular sensitivity to histone deacetylase inhibitors.
- Published
- 2010
- Full Text
- View/download PDF
10. MtDNA mutation pattern in tumors and human evolution are shaped by similar selective constraints.
- Author
-
Zhidkov I, Livneh EA, Rubin E, and Mishmar D
- Subjects
- Haplotypes genetics, Humans, Biological Evolution, DNA, Mitochondrial genetics, DNA, Neoplasm genetics, Mutation genetics, Neoplasms genetics, Selection, Genetic
- Abstract
Multiple human mutational landscapes of normal and cancer conditions are currently available. However, while the unique mutational patterns of tumors have been extensively studied, little attention has been paid to similarities between malignant and normal conditions. Here we compared the pattern of mutations in the mitochondrial genomes (mtDNAs) of cancer (98 sequences) and natural populations (2400 sequences). De novo mtDNA mutations in cancer preferentially colocalized with ancient variants in human phylogeny. A significant portion of the cancer mutations was organized in recurrent combinations (COMs), reaching a length of seven mutations, which also colocalized with ancient variants. Thus, by analyzing similarities rather than differences in patterns of mtDNA mutations in tumor and human evolution, we discovered evidence for similar selective constraints, suggesting a functional potential for these mutations.
- Published
- 2009
- Full Text
- View/download PDF
11. Optimal modeling for phase I design of a two drug combination-results of a phase I study of cisplatin with 9-nitrocamptothecin.
- Author
-
Lee SJ, Gounder M, Rubin EH, Li JM, Gu Z, Thalasila A, Loyer E, Kudelka AP, and Verschraegen CF
- Subjects
- Adolescent, Adult, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols pharmacokinetics, Bayes Theorem, Camptothecin administration & dosage, Camptothecin analogs & derivatives, Cisplatin administration & dosage, Cross-Over Studies, Dose-Response Relationship, Drug, Female, Humans, Male, Maximum Tolerated Dose, Remission Induction methods, Young Adult, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Neoplasms drug therapy
- Abstract
Combination of platinum with topoisomerase-I inhibitors are synergistic. The objectives of this study were to determine MTD range and toxicity profile of combinations of oral 9-nitrocamptothecin (9NC) and intravenous cisplatin in patients with refractory solid tumors. Each course was 28 days starting on day 1 with cisplatin, and then 9NC daily for 5 days/week for three weeks. A new two arm crossing design was created: patients in arm 1 were treated with at the single agent recommended dose of cisplatin (50 mg/m(2)), and increasing doses of 9NC and in arm 2 with the single agent recommended dose of 9NC (1.5 mg/day) and increasing dose of cisplatin. Once a dose limiting toxicity was observed, the dose of the escalated drug was decreased by one level, and the fixed-dose drug was then escalated. A 3 + 3 design was used. Eligibility criteria were standard for a phase I trial. Pharmacokinetics was performed. Eighteen patients were treated on Arm 1, 3 at the crossing level, and 33 on Arm 2. Dose limiting toxicities were gastrointestinal at the crossing dose level. After crossing, prolonged grade 3 thrombocytopenia was the DLT in arm 1, and grade 4 neutropenia in Arm 2. Only one patient with ovarian cancer had a partial remission, and 12 patients had disease stabilization (24% of clinical benefit). A Bayesian optimal dose finding was tested post-facto. The recommended doses for phase II studies by the 3 + 3 design are cisplatin 60 mg/m(2) and 9NC 1.25 mg/day and cisplatin 40 mg/m(2) and 9NC 2.0 mg/day. The Bayesian optimal dose finding suggested a different solution, closest to that of the latter dosing which may be less toxic.
- Published
- 2008
- Full Text
- View/download PDF
12. A phase I study of the dolastatin-15 analogue tasidotin (ILX651) administered intravenously daily for 5 consecutive days every 3 weeks in patients with advanced solid tumors.
- Author
-
Ebbinghaus S, Rubin E, Hersh E, Cranmer LD, Bonate PL, Fram RJ, Jekunen A, Weitman S, and Hammond LA
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Agents chemistry, Area Under Curve, Depsipeptides chemistry, Dose-Response Relationship, Drug, Female, Humans, Infusions, Intravenous, Male, Maximum Tolerated Dose, Melanoma drug therapy, Middle Aged, Neoplasm Metastasis, Oligopeptides chemistry, Time Factors, Tomography, X-Ray Computed, Antineoplastic Agents administration & dosage, Depsipeptides administration & dosage, Neoplasms drug therapy, Oligopeptides administration & dosage
- Abstract
Purpose: To determine the maximum tolerated dose, dose-limiting toxicity, and pharmacokinetics of the dolastatin-15 analogue, tasidotin (ILX651), when administered i.v. daily for 5 days every 3 weeks., Experimental Design: Thirty-six patients with advanced solid tumors received a total of 114 courses through eight dose levels ranging from 2.3 to 36.3 mg/m(2). Pharmacokinetic samples were collected in cycle 1., Results: Neutropenia was the principal dose-limiting toxicity at 36.3 mg/m(2)/d along with grade 3 ileus and elevated aspartate amino transaminase/alanine amino transaminase (n = 1). At the maximum tolerated dose, 27.3 mg/m(2), 4 of 14 patients experienced dose-limiting grade 4 neutropenia. The other principal toxicities consisted of mild-to-moderate elevated transaminases, alopecia, fatigue, and nausea. One patient with melanoma metastatic to liver and bone treated at 15.4 mg/m(2)/d experienced a complete response and received 20 courses of tasidotin. Two other patients with melanoma had mixed responses of cutaneous metastases at 27.3 mg/m(2)/d associated with either stable or progressive visceral disease. In addition, nine patients had stable disease. There was no accumulation of tasidotin following repeated daily dosing. Tasidotin decayed from plasma in a biphasic fashion with a half-life of <45 minutes in most cases., Conclusion: The maximum tolerated dose and recommended phase II dose for tasidotin when administered on this schedule was 27.3 mg/m(2)/d. The favorable toxicity profile of tasidotin compared with other antitubulin agents (particularly the lack of severe cumulative neuropathy, peripheral edema, and fatigue), the observed antitumor activity of tasidotin, and its novel mechanism of action support further disease-directed evaluations of this agent on this 5-day schedule every 3 weeks.
- Published
- 2005
- Full Text
- View/download PDF
13. Tubulin-targeting agents.
- Author
-
Hait WN, Rubin E, and Goodin S
- Subjects
- Drug Design, Humans, Vinblastine analogs & derivatives, Vinblastine therapeutic use, Vinorelbine, Antineoplastic Agents therapeutic use, Neoplasms drug therapy, Tubulin Modulators
- Published
- 2005
- Full Text
- View/download PDF
14. A phase I trial of weekly paclitaxel, 13- cis-retinoic acid, and interferon alpha in patients with prostate cancer and other advanced malignancies.
- Author
-
Thalasila A, Poplin E, Shih J, Dvorzhinski D, Capanna T, Doyle-Lindrud S, Beers S, Goodin S, Rubin E, and DiPaola RS
- Subjects
- Aged, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols adverse effects, Dose-Response Relationship, Drug, Down-Regulation, Female, Humans, Interferon-alpha administration & dosage, Isotretinoin administration & dosage, Leukocytes, Mononuclear metabolism, Male, Middle Aged, Neoplasms metabolism, Paclitaxel administration & dosage, Prostatic Neoplasms drug therapy, Prostatic Neoplasms metabolism, Proto-Oncogene Proteins c-bcl-2 biosynthesis, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Neoplasms drug therapy
- Abstract
Purpose: Based on prior studies demonstrating the effect of 13- cis-retinoic acid and interferon alpha (CRA/IFN) in decreasing the expression of the antiapoptotic protein bcl-2, our prior clinical study of CRA/IFN with paclitaxel (TAX) administered every 3 weeks, and data demonstrating increased activity of weekly TAX against prostate cancer, we designed a phase I study of weekly TAX in combination with CRA/IFN in patients with prostate cancer and other advanced malignancies. To develop a marker of drug effect, we assessed bcl-2 downregulation in patient peripheral blood mononuclear cells (PBMC)., Methods: Enrolled in the study were 14 patients with prostate cancer or other advanced malignancies, and 13 were treated with 1 mg/kg CRA on days 1 and 2, 6 MU/m(2) IFN subcutaneously on days 1 and 2, and TAX at increasing doses on day 2 each week for 6 weeks out of an 8-week cycle. The effect of CRA/IFN on bcl-2 expression was assessed in PBMCs by immunoblotting., Results: The combination of CRA/IFN and TAX was well tolerated. Dose-limiting toxicities (DLT) in the first cycle of therapy included one patient with fever and neutropenia, and one patient with grade 4 hypertriglyceridemia. The recommended phase II dose of TAX in this combination was 80 mg/m(2). Of 13 patients assessable by tumor markers or scans, 5 had stable disease and 2 had a biochemical partial response including a patient with a decrease in PSA of >50% while on study. The assessment of patient PBMC bcl-2 was feasible in ten patients., Conclusions: This is the first study in which the safety and clinical activity of weekly TAX combined with CRA/IFN has been demonstrated. The assessment of PBMC bcl-2 is feasible in this weekly chemotherapy schedule
- Published
- 2003
- Full Text
- View/download PDF
15. A phase I and pharmacologic study of the combination of marimastat and paclitaxel in patients with advanced malignancy.
- Author
-
Toppmeyer DL, Gounder M, Much J, Musanti R, Vyas V, Medina M, Orlando T, Pennick M, Lin Y, Shih W, Goodin S, and Rubin E
- Subjects
- Adult, Aged, Antineoplastic Agents, Phytogenic chemistry, Antineoplastic Agents, Phytogenic pharmacokinetics, Dose-Response Relationship, Drug, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacokinetics, Female, Humans, Hydroxamic Acids chemistry, Hydroxamic Acids pharmacokinetics, Male, Middle Aged, Molecular Structure, Paclitaxel chemistry, Paclitaxel pharmacokinetics, Antineoplastic Agents, Phytogenic therapeutic use, Antineoplastic Combined Chemotherapy Protocols, Enzyme Inhibitors therapeutic use, Hydroxamic Acids therapeutic use, Neoplasms drug therapy, Paclitaxel therapeutic use
- Abstract
Background: Marimastat is a potent inhibitor of matrix metalloproteinases and in preclinical studies enhances the anti-tumor activity of certain chemotherapeutics. We performed a phase I clinical evaluation of the combination of oral marimastat and intravenous paclitaxel, to determine if these drugs could be co-administered safely, and to determine whether marimastat alters paclitaxel pharmacokinetics., Material/methods: Marimastat was administered twice daily and paclitaxel as a three hour infusion every three weeks. Doses of both marimastat and paclitaxel were escalated in cohorts of patients up to maximal doses of 10 mg for marimastat and 175 mg/m2 for paclitaxel. Paclitaxel plasma pharmacokinetic parameters were assessed in the absence (cycle 1) and presence (cycle 2) of marimastat. Trough marimastat plasma levels were evaluated during cycle 2., Results: A total of 19 patients were treated at three different dose levels. There were no dose-limiting toxicities during the first cycle of therapy, resulting in dose escalation up to the planned maximal dose for each drug. Neutropenia was the most common significant toxicity at the highest dose level, with grade 3 or higher neutropenia occurring in 38% of patients. There were no complete or partial responses. Pharmacokinetic analyses indicate that marimastat does not alter paclitaxel clearance. At the 10 mg dose, the mean trough marimastat level was 14.8 Kg/L., Conclusions: Marimastat and paclitaxel can be co-administered safely at doses equivalent to those recommended for single-agent administration. Additional studies are necessary to determine whether this combination is more effective in controlling tumor progression than paclitaxel alone.
- Published
- 2003
16. Gemcitabine combined with sequential paclitaxel and carboplatin in patients with urothelial cancers and other advanced malignancies.
- Author
-
DiPaola RS, Rubin E, Toppmeyer D, Eid J, Butzbach D, Dvorzhinski D, Capanna T, Cairdella M, Shih JW, Goodin S, and Todd MB
- Subjects
- Adult, Aged, Carboplatin administration & dosage, Carcinoma, Transitional Cell drug therapy, Carcinoma, Transitional Cell pathology, Cell Survival, Cohort Studies, Drug Administration Schedule, Female, Humans, Male, Middle Aged, Paclitaxel administration & dosage, Tumor Cells, Cultured, Urinary Bladder Neoplasms pathology, Urothelium pathology, Gemcitabine, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Carboplatin therapeutic use, Deoxycytidine analogs & derivatives, Deoxycytidine therapeutic use, Neoplasms drug therapy, Paclitaxel therapeutic use, Urinary Bladder Neoplasms drug therapy
- Abstract
Background: To investigate a novel schedule of gemcitabine (G), paclitaxel (P), and carboplatin (C), based on preclinical studies demonstrating greater activity and decreased toxicity of administering P prior to C., Material/methods: The effect of the P and C drug sequence on tumor cell viability was assessed with a tetrazolium assay on T24 bladder and DU145 prostate cancer cells. Patients with transitional cell cancer (TCC) and other advanced malignancies were treated with G and P on days 1, 8, and 15 of each 28 day cycle. C was administered on day 2 at an AUC of 5. Doses of G and P were varied among cohorts of three patients., Results: Preclinical studies demonstrated that the sequence of P followed by C induced greater cytotoxicity than the reverse sequence. The recommended phase II dose (RPTD) was defined as 70 mg/m2 P, 300 mg/m2 G, and C with AUC of 5. Therapy was well tolerated; fever and neutropenia occurred in only one patient at the RPTD. Grade 3 thrombocytopenia occurred in 5 of 21 patients treated at the RPTD. Out of all 37 patients treated on study, 9 achieved a partial tumor response (PR) and two patients achieved a complete response (CR). Out of the 15 patients with TCC, six had a PR and two had a CR., Conclusions: Preclinical studies demonstrated that the sequence of paclitaxel followed by carboplatin was more effective than the opposite sequence. Administration of gemcitabine and paclitaxel followed by carboplatin was well tolerated and clinically active. GCP should be compared to other combination regimens under investigation for the treatment of TCC.
- Published
- 2003
17. Tubulin-targeting agents.
- Author
-
Hait WN, Rubin E, and Goodin S
- Subjects
- Animals, Antineoplastic Agents chemistry, Antineoplastic Agents therapeutic use, Antineoplastic Agents toxicity, Humans, Paclitaxel chemistry, Paclitaxel pharmacology, Paclitaxel therapeutic use, Paclitaxel toxicity, Taxoids chemistry, Taxoids pharmacology, Taxoids therapeutic use, Taxoids toxicity, Tubulin physiology, Antineoplastic Agents pharmacology, Neoplasms drug therapy, Tubulin drug effects
- Published
- 2003
- Full Text
- View/download PDF
18. Pilot study of gemcitabine (10 mg/m(2) per min) and cisplatin.
- Author
-
Poplin E, Benson A 3rd, Musanti R, Rubin E, and Mulcahy M
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols adverse effects, Cisplatin administration & dosage, Cisplatin adverse effects, Deoxycytidine administration & dosage, Deoxycytidine adverse effects, Female, Humans, Male, Middle Aged, Pilot Projects, Gemcitabine, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Deoxycytidine analogs & derivatives, Neoplasms drug therapy
- Abstract
Purpose: Gemcitabine and cisplatin are routinely used in combination. In this combination, gemcitabine at conventional doses of 1000-1500 mg/m(2) is delivered weekly as a 30-minute bolus. Laboratory data suggest that the synthesis of gemcitabine triphosphate is saturable, and that gemcitabine infused at a rate of 10 mg/m(2) per min optimizes accumulation of the triphosphate. Early clinical experience suggests that gemcitabine delivered by a more prolonged infusion is more myelosuppressive. In this pilot study, we wished to assess if full-dose gemcitabine when given with cisplatin could be delivered by this more prolonged infusion., Methods: In this study, all patients received cisplatin 75 mg/m(2). All gemcitabine doses were delivered at 10 mg/m(2) per min. For the initial cohort (level 1) the gemcitabine dose was 800 mg/m(2) per min. Subsequent escalations were 1000 mg/m(2) per min (level 2), and 1250 mg/m(2) per min (levels 3 and 4). For the first three cohorts, patients received gemcitabine on days 1, 8, and 15 and cisplatin on day 15 on a 28-day cycle. Patients on level 4 received gemcitabine on days 1 and 8 and cisplatin on day 8 on a 21-day cycle. Dose omissions or delays (holds) were mandated for NCI CTC grade 3 or 4 granulocytopenia or grade 2-4 thrombocytopenia., Results: Entered onto this dose-finding study were 23 patients (12 male, 11 female) with advanced solid tumors. Seven patients were treatment-naive. Six patients were treated on level 1, five each on levels 2 and 3 and seven on level 4. Patients received one to seven cycles of treatment. Myelosuppression-related dose holds occurred at all levels. First-cycle dose holds occurred in three of six, four of five, three of five and two of seven patients on successive levels. First-cycle grade 3 or 4 granulocytopenia/thrombocytopenia occurred in three patients on level 1, one patient on level 2, two patients on level 3 and three patients on level 4. There were no partial or complete responses. Four patients were removed for toxicity (three myelosuppression, one nephrotoxicity), one at physician discretion, and 15 with disease progression. Three patients stopped therapy with stable disease after 5-6 months. On level 3, three of five patients remained on therapy for 4 months or more, compared to only one patient on each of the other three levels. CONCLUSIONS Weekly gemcitabine 1250 mg/m(2), utilizing a 10 mg/m(2) per min infusion rate, can be delivered with cisplatin 75 mg/m(2) with tolerable toxicity. When used in combination with cisplatin, however, the benefit of this fixed dose rate infusion gemcitabine compared to standard bolus gemcitabine remains to be determined.
- Published
- 2002
- Full Text
- View/download PDF
19. Tubulin-targeting agents.
- Author
-
Hait WN, Rubin E, and Goodin S
- Subjects
- Animals, Binding Sites, Clinical Trials as Topic, Humans, Neoplasms metabolism, Antineoplastic Agents pharmacology, Neoplasms drug therapy, Tubulin drug effects
- Published
- 2002
20. Tubulin targeting agents.
- Author
-
Hait WN, Rubin E, and Goodin S
- Subjects
- Animals, Binding Sites, Humans, Neoplasms metabolism, Antineoplastic Agents pharmacology, Neoplasms drug therapy, Tubulin drug effects
- Published
- 2001
21. DNA topoisomerase expression in tumors--a novel target for chemotherapy.
- Author
-
Rubin EH
- Subjects
- DNA Topoisomerases, Type I genetics, Gene Expression, Humans, Enzyme Inhibitors therapeutic use, Neoplasms drug therapy, Neoplasms enzymology, Topoisomerase I Inhibitors
- Published
- 2000
- Full Text
- View/download PDF
22. Phase I study of 9-nitro-20(S)-camptothecin in combination with cisplatin for patients with advanced malignancies.
- Author
-
Verschraegen CF, Vincent M, Abbruzzese JL, Siegler D, Kavanagh JJ, Loyer E, Kudelka AP, and Rubin E
- Subjects
- Antineoplastic Combined Chemotherapy Protocols therapeutic use, Camptothecin administration & dosage, Camptothecin adverse effects, Camptothecin analogs & derivatives, Cisplatin administration & dosage, Cisplatin adverse effects, Dose-Response Relationship, Drug, Drug Administration Schedule, Drug Synergism, Humans, Antineoplastic Combined Chemotherapy Protocols adverse effects, Neoplasms drug therapy
- Published
- 2000
- Full Text
- View/download PDF
23. Leucovorin, 5-fluorouracil, and gemcitabine: a phase I study.
- Author
-
Poplin E, Roberts J, Tombs M, Grant S, and Rubin E
- Subjects
- Adult, Aged, Deoxycytidine administration & dosage, Deoxycytidine analogs & derivatives, Female, Fluorouracil administration & dosage, Humans, Leucovorin administration & dosage, Male, Middle Aged, Time Factors, Treatment Outcome, Gemcitabine, Antimetabolites, Antineoplastic therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Neoplasms drug therapy
- Abstract
Gemcitabine is a chemotherapy agent with efficacy in the treatment of lung, pancreas, bladder and breast cancer. It inhibits DNA synthesis by interfering with cytidine triphosphate production and also inhibits the activity of ribonucleotide reductase. Gemcitabine may potentiate fluorouracil's inhibition of thymidylate synthase. This inhibition would be expected to be sequence dependent, occurring only if gemcitabine were administered following fluorouracil (5FU). The combination of leucovorin, 5-FU, and gemcitabine was assessed in this phase I trial. Eligibility requirements included refractory solid tumor malignancy; adequate hematologic, renal and hepatic reserve; no prior therapy with the combination of leucovorin and 5FU, or with gemcitabine; ECOG performance status 0-2, and signed informed consent. Eleven men and nine women were eligible. The median age was 52.5 years and the median performance status was 1. All but three patients had prior chemotherapy. The starting doses were leucovorin 20 mg/m2, 5FU 255 mg/m2 and gemcitabine 600 mg/m2. 5FU and gemcitabine were escalated in tandem to 340 mg/m2 and 800 mg/m2 and thereafter to 425 mg/m2 and 1000 mg/m2, respectively. Gemcitabine administration always followed that of 5FU by 30 minutes. The median number of cycles was 2 (range 1-32). Two patients at the starting dose had disease progression within the first cycle with one death on day 28. One patient with cholangiocarcinoma had a partial response and remained on study for 40 months. There were no other responses. The maximum tolerated dose is leucovorin 20 mg/m2, 5FU 340 mg/m2, and gemcitabine 800 mg/m2. The impact of drug sequence remains undetermined.
- Published
- 1999
- Full Text
- View/download PDF
24. Trials of 9-amino-20(S)-camptothecin in Boston.
- Author
-
Eder JP, Rubin E, Stone R, Bryant M, Xu G, Supko J, Kinchla N, Lynch T, Hurwitz S, Rodriguez D, Shapiro C, Toppmeyer D, Grossbard M, Vosburg E, Huberman M, Schnipper L, Shulman L, and Kufe DW
- Subjects
- Adult, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, Boston, Camptothecin adverse effects, Camptothecin pharmacokinetics, Camptothecin therapeutic use, DNA Topoisomerases, Type I blood, Female, Humans, Male, Middle Aged, Antineoplastic Agents therapeutic use, Breast Neoplasms drug therapy, Camptothecin analogs & derivatives, Leukemia drug therapy, Neoplasms drug therapy
- Abstract
9-Amino-20(S)-camptothecin (9-AC) is an analog of camptothecin with limited water solubility which has shown significant preclinical activity in a variety of human solid tumor xenografts. A Phase I trial using a soluble formulation of 9-AC, given as a 72-hour continuous infusion, has been completed. Thirty-one patients with resistant cancers received 5-60 micrograms/M2/h at three week intervals. The Maximum Tolerated Dose (MTD) was 45 micrograms/M2/hour. Neutropenia was the dose limiting toxicity, with few significant non-myelosuppressive toxicities. Minor responses were seen in 3/31 patients. Pharmacokinetic studies of 9-AC lactone (closed ring) showed substantial interpatient variability with a predicted half-life of 36 hours. A phase I/II trial of the same formulation of 9-AC is ongoing in refractory leukemia. Stomatitis and diarrhea are the non-myelosuppressive dose limiting toxicities. Evidence of antineoplastic activity has been seen in 3/15 patients. A Phase II trial in previously untreated metastatic breast cancer is also underway. A Phase I trial of a colloidal dispersion formulation, not yet completed, is better tolerated with a MTD > 45 micrograms/M2/h as a 72-hour continuous infusion. Evidence of antineoplastic activity has also been demonstrated.
- Published
- 1996
- Full Text
- View/download PDF
25. The questionable link between alcohol intake and cancer.
- Author
-
Rubin E
- Subjects
- Alcohol Drinking epidemiology, Humans, Neoplasms epidemiology, Risk Assessment, Smoking adverse effects, Alcohol Drinking adverse effects, Neoplasms chemically induced
- Published
- 1996
- Full Text
- View/download PDF
26. A phase I and pharmacokinetic study of a new camptothecin derivative, 9-aminocamptothecin.
- Author
-
Rubin E, Wood V, Bharti A, Trites D, Lynch C, Hurwitz S, Bartel S, Levy S, Rosowsky A, and Toppmeyer D
- Subjects
- Adult, Aged, Anemia chemically induced, Antineoplastic Agents administration & dosage, Camptothecin administration & dosage, Camptothecin adverse effects, Camptothecin pharmacokinetics, DNA Topoisomerases, Type I blood, DNA Topoisomerases, Type II blood, Drug Administration Schedule, Female, Half-Life, Humans, Infusions, Intravenous, Leukocytes, Mononuclear enzymology, Male, Metabolic Clearance Rate, Middle Aged, Neoplasms blood, Neutropenia chemically induced, Thrombocytopenia chemically induced, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, Camptothecin analogs & derivatives, Neoplasms drug therapy
- Abstract
Camptothecins are the only available antitumor agents which target the nuclear enzyme topoisomerase I. 9-Aminocamptothecin (9-AC) is a water-insoluble derivative of camptothecin which has demonstrated impressive antitumor activity in preclinical models. While two other water-soluble derivatives, CPT-11 and topotecan, have successfully completed Phase I and Phase II testing, biochemical and tissue culture studies suggest that camptothecin analogues differ in characteristics which may be important in determining antitumor activity. We performed a Phase I trial of 9-AC to determine the pharmacokinetics, dose-limiting toxicity, and maximum tolerated dose of this agent when administered as a 72-h continuous i.v. infusion. Thirty-one patients with resistant solid cancers received 5-60 microgram/m2/h 9-AC for 72 h, repeated at 3-week intervals. The drug was administered in a vehicle containing dimethylacetamide, polyethylene glycol, and phosphoric acid. Blood samples were collected and the lactone (closed ring) form of 9-AC was quantitated. The maximum tolerated dose of 9-AC was determined to be 45 microgram/m2/h. Dose-limiting toxicity consisted of neutropenia. Thrombocytopenia was also prominent. There were no significant nonhematological toxicities. Minimal responses were seen in patients with gastric, colon, and non-small cell lung cancer. Although significant interpatient variation in plasma 9-AC lactone levels was observed, pooled data were fit to a two-compartment model, with a terminal half-life of 36 h. Analyses of topoisomerase protein levels in peripheral blood cells indicated decreases in topoisomerase I accompanied by increases in topoisomerase II in two of three patients. 9-AC is an active antitumor agent and may be administered safely as a 72-h infusion in patients with cancer. Although Phase II trials with a 72-h infusion of 9-AC are warranted, alternate schedules should be evaluated given the dramatic preclinical activity seen with more prolonged administrations.
- Published
- 1995
27. A minimal toxicity approach to cancer therapy: possible role of beta-glucuronidase.
- Author
-
Rubin DM and Rubin EJ
- Subjects
- Animals, Cell Survival drug effects, Cytoplasm enzymology, Drug Evaluation, Preclinical, Glucuronates pharmacology, Humans, Hydrogen-Ion Concentration, Models, Biological, Neoplasm Metastasis, Neoplasms enzymology, Neoplasms pathology, Neoplasms ultrastructure, Antineoplastic Agents pharmacology, Glucuronates therapeutic use, Glucuronidase metabolism, Neoplasms drug therapy
- Abstract
Most cancer cells differ from normal cells in that they show higher beta-glucuronidase activity and lower pH of their cytoplasm. Anti-cancer drugs can be designed which take advantage of these gradients to deliver maximal toxicity to tumors and minimal toxicity to normal tissue. Many design criteria are suggested here, the most basic of which is the use of the glucuronide structure, in which glucuronic acid acts as a protective carrier of a toxic fragment which becomes active when split off by the beta-glucuronidase at the tumor site. The high beta-glucuronidase activity in cancer cells is also discussed here as a possible explanation for some of the pathognomonic features of a malignant growth: the automatic proliferation of tumor tissue, the invasion of tumors into adjacent tissue, the metastases to remote sites, and the weak response of the immune system.
- Published
- 1980
- Full Text
- View/download PDF
28. ROLE OF CELL PROLIFERATION IN HEPATIC CARCINOGENESIS.
- Author
-
RUBIN E, MASUKO K, GOLDFARB S, and ZAK FG
- Subjects
- Rats, Autoradiography, Carcinogenesis, Carcinogens, Cell Proliferation, DNA, DNA, Neoplasm, Liver Neoplasms, Neoplasms, Neoplasms, Experimental, Nitrosamines, Research
- Published
- 1964
- Full Text
- View/download PDF
29. LUNG BIOPSY FOR DIFFUSE PULMONARY LESIONS: VALUE AND LIMITATIONS.
- Author
-
RUBIN EH and RUBIN M
- Subjects
- Adolescent, Child, Humans, Biopsy, Bronchoscopy, Clinical Laboratory Techniques, Geriatrics, Hodgkin Disease, Lung, Lung Diseases, Lung Neoplasms, Lymph Nodes, Neoplasms diagnosis, Pathology, Pneumoconiosis, Pulmonary Fibrosis, Sarcoidosis, Tuberculosis, Tuberculosis, Miliary
- Published
- 1964
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.