Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 8 de 8
1.
Invest New Drugs ; 42(2): 221-228, 2024 Apr.
Article En | MEDLINE | ID: mdl-38441850

AbGn-107 is an antibody-drug conjugate directed against AG-7 antigen, a Lewis A-like glycol-epitope expressed in a variety of gastrointestinal (GI) malignancies. Based on promising antitumor activity of AbGn-107 in both in vitro and in vivo preclinical studies, we performed a GI cancer-specific Phase I trial. Standard 3 + 3 dose escalation was used evaluating intravenous doses ranging from 0.1 mg/kg every 4 weeks to 1.0 mg/kg every 2 weeks. Key eligibility included chemo-refractory locally advanced, recurrent, or metastatic gastric, colorectal, pancreatic, or biliary cancer, with ECOG PS 0-1; positive AG-7 expression was not required during dose escalation phase. Patients were treated until disease progression or unacceptable toxicity, with tumor assessments every 8 weeks. Primary objectives included safety and determination of maximum tolerated dose; secondary objectives included efficacy defined by objective response rate. Thirty-nine patients were enrolled across seven dose levels during dose escalation phase. Based on safety profile and pharmacokinetic data, 1.0 mg/kg Q2W was selected as the dose schedule for cohort expansion phase, in which an additional seven patients were enrolled. Median number of lines of prior therapy was 3 (range 1-7). AbGn-107 was generally well-tolerated, with infections, cytopenias, hyponatremia, fatigue, abdominal pain, and diarrhea representing the most common grade 3 or higher treatment-emergent adverse events. One subject achieved a partial response, while 18 (46.2%) achieved a best response of stable disease. Disease control lasting > 6 months was observed in 6 subjects (13.0%), including 4 of 15 (26.7%) treated at the highest dose level. AbGn-107 showed a reasonable safety profile and modest clinical activity in this highly pretreated patient population. Further evaluation is required to assess the clinical validity of AG-7 as a suitable antigen for therapeutic targeting. Clinical Trial information: NCT02908451.


Gastrointestinal Neoplasms , Immunoconjugates , Humans , Immunoconjugates/adverse effects , Gastrointestinal Neoplasms/drug therapy , Maximum Tolerated Dose
2.
Neoplasia ; 17(9): 716-722, 2015 Sep.
Article En | MEDLINE | ID: mdl-26476079

Overall survival (OS) has emerged as the definitive regulatory "be-all, end-all" for the demonstration of benefit in cancer clinical trials. The reason and the rationale for why this is so are easily appreciated: literally a "test of time," OS is a seemingly unambiguous, agenda-free end point, independent of bias-prone variables such as the frequency and methods of assessment, clinical evaluation, and the definition of progression. However, by general consensus, OS is an imperfect end point for several reasons: First, it may often be impractical because of the length, cost, and the size of clinical trials. Second, OS captures the impact of subsequent therapies, both beneficial (i.e., active) and detrimental, on survival but it does not take into account the contribution of subsequent therapies by treatment arm; the postprogression period is treated as an unknown black box (no information about the potential influence of next-line therapies on the outcome) under the implicit assumption that the clinical trial treatment is the only clinical variable that matters: what OS explicitly measures is the destination, that is, the elapsed time between the date of randomization (or intention to treat) and the date of death, not the journey, that is, what transpires in-between. In long-term maintenance strategies, patients receive treatment in temporally separated but mutually interdependent and causally linked sequences that exert a "field of influence" akin to action-at-a-distance forces like gravity, electricity, and magnetism on both the tumor and each other. Hence, in this setting, a new end point, PFS2, is required to measure this field of influence. This article reviews the definition and use in clinical trials of PFS2 and makes the case for its potential applicability as a preferred end point to measure the mutual influence of individual regimens in long-term maintenance strategies with resensitizing agents in particular.


Antineoplastic Agents/therapeutic use , Clinical Trials as Topic/methods , Neoplasms/drug therapy , Neoplasms/mortality , Animals , Humans , Neoplasms/diagnosis , Survival Rate/trends
3.
Redox Biol ; 6: 1-8, 2015 Dec.
Article En | MEDLINE | ID: mdl-26164533

The endogenous mediator of vasodilation, nitric oxide (NO), has been shown to be a potent radiosensitizer. However, the underlying mode of action for its role as a radiosensitizer - while not entirely understood - is believed to arise from increased tumor blood flow, effects on cellular respiration, on cell signaling, and on the production of reactive oxygen and nitrogen species (RONS), that can act as radiosensitizers in their own right. NO activity is surprisingly long-lived and more potent in comparison to oxygen. Reports of the effects of NO with radiation have often been contradictory leading to confusion about the true radiosensitizing nature of NO. Whether increasing or decreasing tumor blood flow, acting as radiosensitizer or radioprotector, the effects of NO have been controversial. Key to understanding the role of NO as a radiosensitizer is to recognize the importance of biological context. With a very short half-life and potent activity, the local effects of NO need to be carefully considered and understood when using NO as a radiosensitizer. The systemic effects of NO donors can cause extensive side effects, and also affect the local tumor microenvironment, both directly and indirectly. To minimize systemic effects and maximize effects on tumors, agents that deliver NO on demand selectively to tumors using hypoxia as a trigger may be of greater interest as radiosensitizers. Herein we discuss the multiple effects of NO and focus on the clinical molecule RRx-001, a hypoxia-activated NO donor currently being investigated as a radiosensitizer in the clinic.


Antineoplastic Agents/pharmacology , Azetidines/pharmacology , Neoplasms/therapy , Nitric Oxide Donors/pharmacology , Nitric Oxide/pharmacology , Nitro Compounds/pharmacology , Radiation-Sensitizing Agents/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Azetidines/chemistry , Azetidines/metabolism , Epigenesis, Genetic , Gamma Rays/therapeutic use , Hemoglobins/chemistry , Hemoglobins/metabolism , Humans , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia/pathology , Hypoxia/therapy , Mice , Models, Molecular , Neoplasms/blood supply , Neoplasms/genetics , Neoplasms/metabolism , Nitric Oxide/metabolism , Nitric Oxide Donors/chemistry , Nitric Oxide Donors/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Nitro Compounds/chemistry , Nitro Compounds/metabolism , Oxidation-Reduction , Oxygen/metabolism , Protein Binding , Radiation-Sensitizing Agents/chemistry , Radiation-Sensitizing Agents/metabolism
4.
Surgery ; 152(6): 1078-87, 2012 Dec.
Article En | MEDLINE | ID: mdl-23158178

BACKGROUND: Anaplastic thyroid cancer (ATC) is an aggressive neoplasm for which a paucity of data exist about the relative role of operative procedures in disease management. METHODS: The FACT trial was a randomized, controlled phase 2/3 trial assessing the safety and efficacy of carboplatin/paclitaxel with CA4P (experimental arm) or without CA4P (control arm) in ATC, 2007-11. Patients were permitted to have had an operation before enrollment, which was stratified on the basis of exposure to operation. A subpopulation of patients who had a cancer-related operation (thyroidectomy) was compared with those who did not, and 1-year and median survival were estimated. RESULTS: A total of 80 patients were enrolled; 55% had undergone a cancer-related operation, of whom 70% had near-total/total thyroidectomy. Baseline characteristics for operative and nonoperative patients were not substantially different. Median survival for patients who had cancer-related operation was 8.2 months in the CA4P arm versus 4.0 months in the control arm, resulting in a hazard ratio of 0.66 (P = .25) and a suggested associated reduction in risk of death of 35%. 1-year survival was 33.3% in the CA4P arm versus 7.7% in the control arm. CONCLUSION: In this largest prospective study ever conducted in ATC, thyroidectomy followed by CA4P combination regimen showed a nonsignificant trend toward improvement in patient survival.


Antineoplastic Agents, Phytogenic/therapeutic use , Stilbenes/therapeutic use , Thyroid Neoplasms/therapy , Thyroidectomy , Aged , Combined Modality Therapy , Female , Humans , Male , Middle Aged , Survival Rate , Thyroid Carcinoma, Anaplastic , Thyroid Neoplasms/mortality
5.
J Clin Oncol ; 25(31): 4961-6, 2007 Nov 01.
Article En | MEDLINE | ID: mdl-17971594

PURPOSE: To compare time to progression (TTP) with a steroidal aromatase inhibitor (AI) atamestane (ATA) combined with toremifene (TOR; complete estrogen blockade) versus letrozole (LET) in receptor-positive advanced breast cancer (ABC). PATIENTS AND METHODS: Eligibility included postmenopausal receptor-positive ABC and adjuvant hormonal therapy completed more than 12 months prior to study entry. Participants received daily ATA 500 mg with TOR 60 mg (ATA + TOR), or letrozole 2.5 mg (LET). The primary end point was TTP, whereas secondary objectives included objective response (OR), overall survival (OS), and time to treatment failure (TTF). The study had 80% power to detect a 25% increase in TTP assuming a TTP of 9.4 months in the LET population. RESULTS: A total of 865 patients were randomly assigned (434 to ATA + TOR and 431 to LET) in 60 centers in the United States, Canada, Russia, and Ukraine. Baseline characteristics were balanced. Median TTP was identical in the two arms at 11.2 months (P < .92). Median TTF was similar at 9.24 months (ATA + TOR) versus 10.44 months (LET). The hazard ratios (LET/ATA + TOR) were 1.00 (95% CI, 0.92 to 1.08) for TTP, 0.99 (95% CI, 0.92 to 1.06) for TTF, and 0.98 (95% CI, 0.87 to 1.11) for OS. OR occurred in 30% of patients receiving ATA + TOR and in 36% of patients receiving LET (P < .1). Adverse events (AEs) were similar for patients receiving ATA + TOR versus LET, and serious AEs were 10% v 11%, respectively. CONCLUSION: TTP for patients receiving ATA + TOR was identical to that for patients receiving LET, representing the first endocrine therapy comparable to LET in ABC. Unlike in the Anastrozole, Tamoxifen, and Combined trial, addition of an antiestrogen did not decrease efficacy of the AI. Future studies of AIs in combination with more effective selective estrogen receptor modulators or selective receptor downregulators is warranted.


Androstenedione/analogs & derivatives , Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Nitriles/administration & dosage , Toremifene/administration & dosage , Triazoles/administration & dosage , Aged , Androstenedione/administration & dosage , Breast Neoplasms/metabolism , Disease Progression , Double-Blind Method , Female , Humans , Letrozole , Middle Aged , Postmenopause , Receptors, Steroid/metabolism
6.
Steroids ; 72(8): 666-71, 2007 Jul.
Article En | MEDLINE | ID: mdl-17588628

Currently available radioimmunoassay methods for estradiol in serum lack sufficient sensitivity and precision to monitor estradiol levels in patients placed on third generation aromatase inhibitors. We recently validated a gas chromatography/tandem mass spectrometry assay (GC/MS/MS) for estradiol and determined estrogen levels in normal post-menopausal women and in women with breast cancer before and during administration of aromatase inhibitors. Validation of the GC/MS/MS assay in human plasma and human serum included determination of assay sensitivity (<0.63 pg/ml), precision (all CVs less than 17.8%), recovery (98-103%), and linearity of recovery (R=0.998). Levels of estradiol were lower when assayed by GC/MS/MS compared to RIA under all conditions (7.26+/-4.82 pg/ml versus 11.9+12.0 pg/ml in normal post-menopausal women; 5.88+/-3.43 pg/ml versus 13.8+/-7.5 pg/ml in breast cancer patients prior to treatment; and<0.63 pg/ml versus 5.8+/-4.1 pg/ml during aromatase inhibitor therapy). Fifty-five women treated either with atamestane/toremiphene or letrozole/placebo were monitored for estradiol levels at 4, 8 and 12 weeks of therapy. The mean levels of estradiol during aromatase inhibitor therapy was 5.8+/-4.1 pg/ml as measured by RIA and <0.63 pg/ml by GC/MS/MS. The degree of suppression with the aromatase inhibitors as detected by RIA was 58% versus >89% by GC/MS. These results suggest that most RIA methods detect cross-reacting estrogen metabolites and yield higher measured levels than GC/MS/MS. Several pharmacological and clinical considerations suggest that GC/MS/MS should become the preferred method for monitoring aromatase inhibitor therapy.


Aromatase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Estradiol/blood , Gas Chromatography-Mass Spectrometry , Tandem Mass Spectrometry , Androstenedione/analogs & derivatives , Androstenedione/therapeutic use , Biological Assay , Breast Neoplasms/blood , Female , Humans , Letrozole , Nitriles/therapeutic use , Placebos , Postmenopause , Radioimmunoassay , Sensitivity and Specificity , Triazoles/therapeutic use
7.
Antiviral Res ; 73(2): 118-25, 2007 Feb.
Article En | MEDLINE | ID: mdl-16987555

The fully glycosylated human omega interferon produced from CHO-SS cells (glycosylated IFN-omega) has been shown to be well-tolerated in man and to induce a sustained virologic response in patients infected with hepatitis C virus (HCV). We examined the antiviral activity of glycosylated IFN-omega and various human IFNs (IFN-alpha, -beta, -gamma and non-glycosylated bacterial (Escherichia coli) recombinant IFN-omega (non-glycosylated IFN-omega)) against HCV RNA replicons and several viruses related to HCV. Since none of the IFNs displayed cytotoxicity we compared their activities based on the effective concentration of the IFN that inhibited virus growth by 50% (EC50). Glycosylated IFN-omega was found to be the most potent antiviral agent of all the IFNs tested against bovine viral diarrhea virus (BVDV), yellow fever virus and West Nile virus. With HCV RNA replicons, non-glycosylated IFN-omega was comparable in activity to IFN-alpha while glycosylated IFN-omega was markedly more potent, indicating that glycosylation has an important effect on its activity. Drug combination analysis of glycosylated IFN-omega+ribavirin (RBV) in BVDV showed a synergy of antiviral effects similar to IFN-alpha+RBV, as well as a unique antagonism of RBV cytotoxic effects by glycosylated IFN-omega. Transcription factor (TF) profiling indicated that IFN-alpha or glycosylated IFN-omega treatment upregulated the same 17 TFs. IFN-alpha and glycosylated IFN-omega also upregulated 9 and 40 additional unique TFs, respectively. The differences in the expression of these TFs were modest, but statistically significantly different for eight of the TFs that were upregulated exclusively by glycosylated IFN-omega. The activation of these additional TFs by glycosylated IFN-omega might contribute to its high potency.


Antiviral Agents/pharmacology , Hepacivirus/genetics , Interferon Type I/pharmacology , Interferons/pharmacology , Replicon/drug effects , Animals , Antiviral Agents/immunology , CHO Cells , Cattle , Chlorocebus aethiops , Cricetinae , Cricetulus , Drug Combinations , Humans , Interferon Type I/immunology , Interferons/immunology , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Replicon/genetics , Transcription Factors/genetics , Vero Cells
8.
Basic Clin Pharmacol Toxicol ; 99(1): 62-70, 2006 Jul.
Article En | MEDLINE | ID: mdl-16867173

Human omega-interferon (IFN-omega) has been shown to be well-tolerated in man and to induce reductions of hepatitis C virus RNA levels in a series of human clinical trials. Here we provide an overview of our preclinical safety evaluation of the fully-glycosylated human IFN-omega produced from CHO-SS cells that is currently being evaluated clinically. IFN-omega was not associated with any biologically-relevant adverse effects in a series of 10 safety pharmacology experiments, in the Ames mutagenicity test, in the micronucleus test, or in intraarterial, intravenous, paravenous or subcutaneous local tolerance studies. Acute, subacute, subchronic and reproductive toxicity studies performed in cynomolgus monkeys and rats showed a toxicity profile similar to that of human alpha interferon (IFN-alpha). Except for the acute (single-dose) toxicology study, all of the other toxicity studies showed evidence for the formation of anti-IFN-omega antibodies over time in the animals. These antibodies were found to neutralize IFN-omega antiviral activity in vitro in a dose-dependent manner. The average pharmacokinetic parameters following a single subcutaneous dose of IFN-omega in rabbits, rats and monkeys were determined and found to be similar to that of human IFN-alpha. These findings demonstrate that IFN-omega has a safety profile consistent with that required for its use in man. IFN-omega might be beneficial for the treatment of patients infected with hepatitis C virus who fail to respond to IFN-alpha or as a first-line treatment option.


Antiviral Agents/pharmacology , Interferon Type I/pharmacology , Animals , Antiviral Agents/pharmacokinetics , Antiviral Agents/toxicity , CHO Cells , Cardiovascular Physiological Phenomena/drug effects , Cricetinae , Enzyme-Linked Immunosorbent Assay , Exploratory Behavior/drug effects , Female , Hemolysis/drug effects , Hepatitis C/drug therapy , Hepatitis C/virology , Humans , Injections, Intra-Arterial , Injections, Intravenous , Injections, Subcutaneous , Interferon Type I/pharmacokinetics , Interferon Type I/toxicity , Kidney Function Tests , Liver Function Tests , Macaca fascicularis , Mice , Motor Activity/drug effects , Mutagenicity Tests , Mutagens/toxicity , Neutralization Tests , Pregnancy , Rabbits , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Recombinant Proteins/toxicity , Reproduction , Respiratory Function Tests
...