Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
ESMO Open ; 7(1): 100339, 2022 02.
Article in English | MEDLINE | ID: mdl-34953404

ABSTRACT

The coronavirus disease-19 (COVID-19) pandemic promises to have lasting impacts on cancer clinical trials that could lead to faster patient access to new treatments. In this article, an international panel of oncology experts discusses the lasting impacts of the pandemic on oncology clinical trials and proposes solutions for clinical trial stakeholders, with the support of recent data on worldwide clinical trials collected by IQVIA. These lasting impacts and proposed solutions encompass three topic areas. Firstly, acceleration and implementation of new operational approaches to oncology trials with patient-centric, fully decentralized virtual approaches that include remote assessments via telemedicine and remote devices. Geographical differences in the uptake of remote technology, including telemedicine, are discussed in the article, focusing on the impact of the local adoption of new operational approaches. Secondly, innovative clinical trials. The pandemic has highlighted the need for new trial designs that accelerate research and limit risks and burden for patients while driving optimization of clinical trial objectives and endpoints, while testing is being minimized. Areas of considerations for clinical trial stakeholders are discussed in detail. In addition, the COVID-19 pandemic has exposed the underrepresentation of minority groups in clinical trials; the approach for oncology clinical trials to improve generalizability of efficacy and outcomes data is discussed. Thirdly, a new problem-focused collaborative framework between oncology trial stakeholders, including decision makers, to leverage and further accelerate the innovative approaches in clinical research developed during the COVID-19 pandemic. This could shorten timelines for patient access to new treatments by addressing the cultural and technological barriers to adopting new operational approaches and innovative clinical trials. The role of the different stakeholders is described, with the aim of making COVID-19 a catalyst for positive change in oncology clinical research and eventually in cancer care.


Subject(s)
COVID-19 , Neoplasms , Telemedicine , Clinical Trials as Topic , Humans , Neoplasms/epidemiology , Neoplasms/therapy , Pandemics , SARS-CoV-2
2.
Ann Oncol ; 31(5): 590-598, 2020 05.
Article in English | MEDLINE | ID: mdl-32245699

ABSTRACT

BACKGROUND: Little is known about mechanisms of resistance to poly(adenosine diphosphate-ribose) polymerase inhibitors (PARPi) and platinum chemotherapy in patients with metastatic breast cancer and BRCA1/2 mutations. Further investigation of resistance in clinical cohorts may point to strategies to prevent or overcome treatment failure. PATIENTS AND METHODS: We obtained tumor biopsies from metastatic breast cancer patients with BRCA1/2 deficiency before and after acquired resistance to PARPi or platinum chemotherapy. Whole exome sequencing was carried out on each tumor, germline DNA, and circulating tumor DNA. Tumors underwent RNA sequencing, and immunohistochemical staining for RAD51 foci on tumor sections was carried out for functional assessment of intact homologous recombination (HR). RESULTS: Pre- and post-resistance tumor samples were sequenced from eight patients (four with BRCA1 and four with BRCA2 mutation; four treated with PARPi and four with platinum). Following disease progression on DNA-damaging therapy, four patients (50%) acquired at least one somatic reversion alteration likely to result in functional BRCA1/2 protein detected by tumor or circulating tumor DNA sequencing. Two patients with germline BRCA1 deficiency acquired genomic alterations anticipated to restore HR through increased DNA end resection: loss of TP53BP1 in one patient and amplification of MRE11A in another. RAD51 foci were acquired post-resistance in all patients with genomic reversion, consistent with reconstitution of HR. All patients whose tumors demonstrated RAD51 foci post-resistance were intrinsically resistant to subsequent lines of DNA-damaging therapy. CONCLUSIONS: Genomic reversion in BRCA1/2 was the most commonly observed mechanism of resistance, occurring in four of eight patients. Novel sequence alterations leading to increased DNA end resection were seen in two patients, and may be targetable for therapeutic benefit. The presence of RAD51 foci by immunohistochemistry was consistent with BRCA1/2 protein functional status from genomic data and predicted response to later DNA-damaging therapy, supporting RAD51 focus formation as a clinically useful biomarker.


Subject(s)
Breast Neoplasms , Ovarian Neoplasms , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Female , Humans , Ovarian Neoplasms/drug therapy , Platinum/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use
3.
Ann Oncol ; 29(5): 1304-1311, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29788155

ABSTRACT

BACKGROUND: Checkpoint kinase 1 (Chk1) inhibition following chemotherapy-elicited DNA damage overrides cell cycle arrest and induces mitotic catastrophe and cell death. GDC-0575 is a highly-selective oral small-molecule Chk1 inhibitor that results in tumor shrinkage and growth delay in xenograft models. We evaluated the safety, tolerability, and pharmacokinetic properties of GDC-0575 alone and in combination with gemcitabine. Antitumor activity and Chk1 pathway modulation were assessed. PATIENTS AND METHODS: In this phase I open-label study, in the dose escalation stage, patients were enrolled in a GDC-0575 monotherapy Arm (1) or GDC-0575 combination with gemcitabine Arm (2) to determine the maximum tolerated dose. Patients in arm 2 received either i.v. gemcitabine 1000 mg/m2 (arm 2a) or 500 mg/m2 (arm 2b), followed by GDC-0575 (45 or 80 mg, respectively, as RP2D). Stage II enrolled disease-specific cohorts. RESULTS: Of 102 patients treated, 70% were female, the median age was 59 years (range 27-85), and 47% were Eastern Cooperative Oncology Group PS 0. The most common tumor type was breast (37%). The most frequent adverse events (all grades) related to GDC-0575 and/or gemcitabine were neutropenia (68%), anemia (48%), nausea (43%), fatigue (42%), and thrombocytopenia (35%). Maximum concentrations of GDC-0575 were achieved within 2 hours of dosing, and half-life was ∼23 hours. No pharmacokinetic drug-drug interaction was observed between GDC-0575 and gemcitabine. Among patients treated with GDC-0575 and gemcitabine, there were four confirmed partial responses, three occurring in patients with tumors harboring TP53 mutation. Pharmacodynamic data were consistent with GDC-0575 inhibition of gemcitabine-induced expression of pCDK1/2. CONCLUSION: GDC-0575 can be safely administered as a monotherapy and in combination with gemcitabine; however, overall tolerability with gemcitabine was modest. Hematological toxicities were frequent but manageable. Preliminary antitumor activity was observed but limited to a small number of patients with a variety of refractory solid tumors treated with GDC-0575 and gemcitabine. CLINICAL TRIAL NUMBER: NCT01564251.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Deoxycytidine/analogs & derivatives , Neoplasms/drug therapy , Piperidines/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Pyridines/administration & dosage , Pyrroles/administration & dosage , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Checkpoint Kinase 1/antagonists & inhibitors , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Deoxycytidine/pharmacokinetics , Dose-Response Relationship, Drug , Drug Interactions , Fatigue , Female , Half-Life , Humans , Male , Maximum Tolerated Dose , Middle Aged , Nausea , Neutropenia/chemically induced , Neutropenia/epidemiology , Piperidines/adverse effects , Piperidines/pharmacokinetics , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Pyridines/adverse effects , Pyridines/pharmacokinetics , Pyrroles/adverse effects , Pyrroles/pharmacokinetics , Thrombocytopenia , Treatment Outcome , Gemcitabine
4.
Ann Oncol ; 29(7): 1561-1568, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29726923

ABSTRACT

Background: Brontictuzumab is a monoclonal antibody that targets Notch1 and inhibits pathway activation. The purpose of this first-in-human study was to determine the maximum tolerated dose (MTD), safety, pharmacokinetics, immunogenicity and preliminary efficacy of brontictuzumab in patients with solid tumors. Patients and methods: Subjects with selected refractory solid tumors were eligible. Brontictuzumab was administered intravenously at various dose levels and schedule during dose escalation, and at 1.5 mg/kg every 3 weeks (Q3W) during expansion. Evidence of Notch1 pathway activation as determined by an immunohistochemistry assay was required for entry in the expansion cohort. Adverse events were graded according to the NCI-CTCAE v 4.03. Efficacy was assessed by RECIST 1.1. Results: Forty-eight subjects enrolled (33 in dose escalation and 15 in the expansion phase). The MTD was 1.5 mg/kg Q3W. Dose-limiting toxicities were grade 3 diarrhea in two subjects and grade 3 fatigue in one subject. The most common drug-related adverse events of any grade were diarrhea (71%), fatigue (44%), nausea (40%), vomiting (21%), and AST increase (21%). Brontictuzumab exhibited nonlinear pharmacokinetics with dose-dependent terminal half-life ranging 1-4 days. Clinical benefit was seen in 6 of 36 (17%) assessable subjects: 2 had unconfirmed partial response (PR) and 4 subjects had prolonged (≥ 6 months) disease stabilization (SD). Both PRs and three prolonged SD occurred in adenoid cystic carcinoma (ACC) subjects with evidence of Notch1 pathway activation. Pharmacodynamic effects of brontictuzumab were seen in patients' blood and tumor. Conclusion: Brontictuzumab was well tolerated at the MTD. The main toxicity was diarrhea, an on-target effect of Notch1 inhibition. An efficacy signal was noted in subjects with ACC and Notch1 pathway activation. ClinicalTrials.gov identifier: NCT01778439.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Drug Resistance, Neoplasm/drug effects , Neoplasm Recurrence, Local/drug therapy , Neoplasms/drug therapy , Receptor, Notch1/antagonists & inhibitors , Salvage Therapy , Adult , Aged , Antibodies, Monoclonal/pharmacokinetics , Antineoplastic Agents, Immunological/pharmacokinetics , Cohort Studies , Dose-Response Relationship, Drug , Female , Follow-Up Studies , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Recurrence, Local/pathology , Neoplasms/pathology , Prognosis , Receptor, Notch1/immunology , Survival Rate , Tissue Distribution
5.
Br J Cancer ; 117(11): 1592-1599, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-28972963

ABSTRACT

BACKGROUND: ARQ 087 is an orally administered pan-FGFR inhibitor with multi-kinase activity. This Phase 1 study evaluated safety, pharmacokinetics, and pharmacodynamics of ARQ 087 and defined the recommended Phase 2 dose (RP2D). METHODS: Patients with advanced solid tumours received ARQ 087 administered initially at 25 mg every other day and dose-escalated from 25 to 425 mg daily (QD) continuous dosing. FGF19, 21, 23, and serum phosphate were assessed as potential biomarkers of target engagement. RESULTS: 80 patients were enrolled, 61 in dose-escalation/food-effect cohorts and 19 with pre-defined tumour types in the expansion cohort. The most common ARQ 087-related adverse events were fatigue (49%), nausea (46%), aspartate aminotransferase (AST) increase (30%), and diarrhoea (23%). Four patients (5%) experienced grade 1 treatment-related hyperphosphataemia. Dose-limiting toxicity was reversible grade 3 AST increase. The RP2D was 300 mg QD. Pharmacokinetics were linear and dose-proportional from 25 to 325 mg QD, and were unaffected by food. Statistically significant changes (P-value<0.05) suggest phosphate and FGF19 levels as markers of target engagement. In 18 evaluable patients with FGFR genetic alterations, 3 confirmed partial responses (two intrahepatic cholangiocarcinomas (iCCA) with FGFR2 fusions and one urothelial cancer with FGFR2 and FGF19 amplification) and two durable stable disease at ⩾16 weeks with tumour reduction (FGFR2 fusion-positive iCCA and adrenocortical carcinoma with FGFR1 amplification) were observed. CONCLUSIONS: ARQ 087 had manageable toxicity at the RP2D of 300 mg QD, showed pharmacodynamics effects, and achieved objective responses, notably in patients with FGFR2 genetic alterations.


Subject(s)
Aniline Compounds/therapeutic use , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Quinazolines/therapeutic use , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Administration, Oral , Adult , Aged , Aniline Compounds/adverse effects , Aniline Compounds/pharmacokinetics , Female , Humans , Male , Middle Aged , Quinazolines/adverse effects , Quinazolines/pharmacokinetics , Receptors, Fibroblast Growth Factor/genetics
6.
Ann Oncol ; 28(8): 1776-1787, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28838216

ABSTRACT

Epigenetics has been defined as 'the structural adaptation of chromosomal regions so as to register, signal or perpetuate altered activity states.' Currently, several classes of anticancer drugs function at the epigenetic level, including inhibitors of DNA methyltransferase, histone deacetylase (HDAC), lysine-specific demethylase 1, zeste homolog 2, and bromodomain and extra-terminal motif (BET) proteins.BET proteins have multiple functions, including the initiation and elongation of transcription and cell cycle regulation. In recent years, inhibitors of BET proteins have been developed as anticancer agents. These inhibitors exhibit selectivity for tumor cells by preferentially binding to superenhancers, noncoding regions of DNA critical for the transcription of genes that determine a cell's identity. Preclinical research on BET inhibitors has identified them as a potential means of targeting MYC.Early clinical trials with BET inhibitors have had mixed results, with few responses in both hematologic and solid tumors that tend to be short-lived. Toxicities have included severe, thrombocytopenia, fatigue, nausea, vomiting, and diarrhea; GI side-effects, fatigue, and low-grade dysgeusia have limited compliance. However, preclinical data suggest that BET inhibitors may have a promising future in combination with other agents. They appear to be able to overcome resistance to targeted agents and have strong synergy with immune checkpoint inhibitors as well as with multiple epigenetic agents, particularly HDAC inhibitors. In many instances, BET and HDAC inhibitors were synergistic at reduced doses, suggesting a potential means of avoiding the overlapping toxicities of the two drug classes.BET inhibitors provide a novel approach to epigenetic anticancer therapy. However, to date they appear to have limited efficacy as single agents. A focus on BET inhibitors in combination with other drugs such as targeted and/or as other epigenetic agents is warranted, due to limited monotherapy activity, including pharmacodynamic correlatives differential activity amongst select drug combinations.


Subject(s)
Epigenesis, Genetic , Proteins/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Drug Synergism , Genes, myc , Humans , Neoplasms/drug therapy , Neoplasms/genetics
7.
Eur J Paediatr Dent ; 18(1): 32-36, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28494600

ABSTRACT

AIM: The present study aimed to evaluate the cephalometric effects of a headgear anchored to the deciduous second molars in the early mixed dentition. MATERIALS AND METHODS: Study design: The study followed a retrospective longitudinal design and enrolled 31 consecutive patients (17 females and 14 males) treated with high pull (HP) headgear anchored to the deciduous second molars, average age 8y 5m± 5m at pre-treatment time (T0) and 9y 8m± 6m at post-treatment time (T1). All the patients wore the headgear for approximately 8-10 hours at night, with a force of 250 g per side. The active phase of treatment ended once patients obtained a distal step on permanent molars of at least 2 mm. Lateral cephalograms at T0 and T1 were taken; 10 angular measurements were chosen as variables of the study. The paired sample t-test was employed to assess the significance of the differences of each variable between T0 and T1. RESULTS AND CONCLUSION: In this group of Class II patients, HP headgear anchored to the deciduous second molars in the early mixed dentition produced: significant reduction of SNA angle, significant increase of SN/NL angle with no significant change in SN/ML angle, significant labial flaring of upper incisors. As clinically evaluated, the correction of the Class II occlusal relationship and the anterior crowding of maxillary arch were also accomplished.


Subject(s)
Cephalometry , Extraoral Traction Appliances , Malocclusion, Angle Class II/diagnostic imaging , Malocclusion, Angle Class II/therapy , Molar , Child , Dentition, Mixed , Female , Humans , Longitudinal Studies , Male , Retrospective Studies , Tooth, Deciduous , Treatment Outcome
8.
Eur J Paediatr Dent ; 17(1): 47-52, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26949239

ABSTRACT

AIM: The present study aimed to evaluate the relationship between masseter size, maxillary intermolar width and craniofacial vertical skeletal pattern. STUDY DESIGN: The study followed a prospective longitudinal design and enrolled 61 subjects (30 males and 31 females), 9-14 years of age (mean age 11.5) at 2-3 CVM stage. The participants were divided into three groups based on their vertical skeletal pattern which was estimated using the Frankfurt-mandibular plane angle: low-angle group (L-A), normal-angle group (N-A), high-angle group (H-A). An additional gender-based distinction was made. Maxillary intermolar width was measured on the maxillary cast of each patient by means of an electronic caliper; masseter volume was estimated by using magnetic resonance (MR) and masseter thickness was measured by means of ultrasonography (US). The US registrations were performed during the relaxation state (RS) and the maximum voluntary contraction (MVC) of the muscle. The indipendent samples T- test was used for sex comparisons; the analysis of variance test (ANOVA) was used to evaluate the differences between the three groups in males and females, and the Pearson r correlation coefficient was employed to assess the correlation between maxillary intermolar width and masseter volume. RESULTS AND CONCLUSION: Maxillary intermolar width, masseter volume and thickness showed significant gender differences; all the tested variables decreased significantly according to the facial vertical pattern, with greater values in females, especially in low- and normal-angle subjects; maxillary intermolar width and masseter volume showed significant correlations, higher in females.


Subject(s)
Dental Arch/anatomy & histology , Masseter Muscle/anatomy & histology , Maxilla/anatomy & histology , Vertical Dimension , Adolescent , Cephalometry/methods , Child , Female , Humans , Longitudinal Studies , Magnetic Resonance Imaging/methods , Male , Masseter Muscle/diagnostic imaging , Models, Dental , Molar/anatomy & histology , Muscle Contraction/physiology , Muscle Relaxation/physiology , Organ Size , Prospective Studies , Sex Factors , Ultrasonography
9.
Ann Oncol ; 27(3): 526-32, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26646757

ABSTRACT

BACKGROUND: Fibroblast growth factors (FGFs) play important roles in multiple cancers by supporting tumor growth and angiogenesis. FP-1039 (GSK3052230) is a FGF ligand trap consisting of the extracellular domain of FGF receptor 1 (FGFR1) fused with the Fc region of IgG1. FP-1039 binds and neutralizes multiple FGFs that normally bind FGFR1. The primary objective of this phase I study was to evaluate the safety and tolerability of FP-1039. PATIENTS AND METHODS: Eligible patients with metastatic or locally advanced solid tumors for which standard treatments were ineffective were treated with weekly doses of FP-1039 for 4 weeks, followed by 2 weeks observation. RESULTS: Thirty-nine subjects received a mean of 6 infusions of FP-1039 at doses ranging from 0.5 to 16 mg/kg weekly, with no maximally tolerated dose identified. Grade 3 or greater treatment emergent adverse events were uncommon. Four dose-limiting toxicities were reported at doses of 0.75 mg/kg (urticaria), 1 mg/kg (intestinal perforation and neutropenia), and 16 mg/kg (muscular weakness). Drug exposure was dose proportional, and the terminal elimination half-life was 2.6-3.9 days following a single dose. Target engagement as measured by low free plasma FGF2 levels was achieved. FGF pathway dysregulation was uncommon. No objective responses were observed. CONCLUSION: In nonselected cancer patients with advanced disease, treatment with FP-1039 was well tolerated and toxicities associated with small molecule drugs that inhibit FGFR tyrosine kinases, including hyperphosphatemia, were not observed. Further studies of FP-1039 in patients selected for FGF pathway dysregulation, who are most likely to benefit, are now underway.


Subject(s)
Antineoplastic Agents/therapeutic use , Fibroblast Growth Factors/antagonists & inhibitors , Immunoglobulin G/therapeutic use , Neoplasms/drug therapy , Oncogene Proteins, Fusion/therapeutic use , Receptor, Fibroblast Growth Factor, Type 1/therapeutic use , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Fibroblast Growth Factors/metabolism , Humans , Immunoglobulin G/adverse effects , Male , Maximum Tolerated Dose , Middle Aged , Oncogene Proteins, Fusion/adverse effects , Oncogene Proteins, Fusion/pharmacokinetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Recombinant Fusion Proteins
10.
Invest New Drugs ; 33(2): 450-62, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25749986

ABSTRACT

BACKGROUND: Type 1 insulin-like growth factor receptor (IGF-IR) signaling is often dysregulated in cancer. Cixutumumab, a fully human IgG1 monoclonal antibody, blocks IGF-IR and inhibits downstream signaling. The current study determined the recommended dose, safety, and pharmacokinetic (PK) profile of weekly or every-2-week dosing of cixutumumab. PATIENTS AND METHODS: Two open-label, multicenter phase I studies evaluated weekly (3-15 mg/kg) or every-2-weeks (6-15 mg/kg) dosing of cixutumumab in patients with advanced solid tumors. Serial blood samples for PK were collected up to 168-336 h (day 8-15) following the first administration of cixutumumab. Efficacy was evaluated as best overall tumor response. RESULTS: A total of 24 and 16 patients were enrolled in the weekly and every-2-week dosing studies, respectively. Treatment-emergent adverse events (≥10%) included hyperglycemia, fatigue, anemia, nausea, and vomiting. Severe adverse events (AE) were infrequent; one serious AE (grade 3 electrocardiogram QT prolongation) was deemed possibly cixutumumab-related (10 mg/kg every-2-weeks). One death occurred due to disease progression (6 mg/kg weekly cohort). Maximum serum concentrations increased with dose. A maximum tolerated dose was not identified; pre-determined target serum minimum concentrations (60 µg/mL) were achieved with ≥6 mg/kg weekly and ≥10 mg/kg every-2-week dosing. Cixutumumab terminal elimination half-life is approximately a week (individual range, t1/2 = 4.58-9.33 days based upon 10 mg/kg every 2 weeks). Overall, stable disease was achieved in 25% of all patients. CONCLUSIONS: Cixutumumab was associated with favorable safety and PK profiles. A dosing regimen of 10 mg/kg every 2 weeks was recommended for subsequent disease-focused clinical trials.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Neoplasms/drug therapy , Receptor, IGF Type 1/antagonists & inhibitors , Adult , Aged , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Area Under Curve , Female , Half-Life , Humans , Male , Maximum Tolerated Dose , Metabolic Clearance Rate , Middle Aged
11.
Invest New Drugs ; 32(3): 436-44, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24258465

ABSTRACT

The aim of this study was to determine the maximum tolerated dose, dose-limiting toxicities, and pharmacokinetic profile of E7107 in patients with advanced solid tumors. Patients in this phase I, open-label, single-arm, dose-escalation study had metastatic or locally advanced solid tumors and received E7107 as a 30-minute intravenous infusion at doses of 0.6, 1.2, 1.8, 2.4, 3.2, 4.3, and 5.7 mg/m(2). Twenty-six patients were enrolled in the study. At 5.7 mg/m(2), two patients experienced dose-limiting toxicities including diarrhea, vomiting, dehydration, and myocardial infarction on Days 1-3 following E7107 administration. Three additional patients were recruited at the lower dose and all six patients tolerated E7107 4.3 mg/m(2) with no dose-limiting toxicities. The maximum tolerated dose of E7107 was therefore 4.3 mg/m(2). The most common drug-related adverse events were nausea, vomiting, and diarrhea. Vision loss was experienced by two patients at Cycles 2 and 7, each patient receiving 3.2 mg/m(2) and 4.3 mg/m(2), respectively. This resulted in the study being put on clinical hold. Pharmacokinetic analysis showed that E7107 was rapidly distributed with a moderate elimination half-life (6-13 h) and high clearance. Exposure to E7107 was dose-related. The best tumor response was stable disease in eight patients. E7107 is a unique first-in-class molecule. The incidence of two cases of vision loss probably related to E7107 led to study discontinuation.


Subject(s)
Antineoplastic Agents/administration & dosage , Epoxy Compounds/administration & dosage , Macrolides/administration & dosage , Neoplasms/drug therapy , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Diarrhea/chemically induced , Epoxy Compounds/adverse effects , Epoxy Compounds/pharmacokinetics , Female , Humans , Infusions, Intravenous , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Macrolides/adverse effects , Macrolides/pharmacokinetics , Male , Maximum Tolerated Dose , Middle Aged , Nausea/chemically induced , Neoplasms/metabolism , RNA Splicing/drug effects , RNA, Messenger/metabolism , Spliceosomes , Vision Disorders/chemically induced , Vomiting/chemically induced
12.
Br J Cancer ; 108(4): 826-30, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23412108

ABSTRACT

BACKGROUND: Adrenocortical carcinoma (ACC) is a rare and aggressive endocrine malignancy without an available effective systemic chemotherapy. Insulin growth factor 2 (IGF-2) overexpression leading to the activation of the IGF-1 receptor (IGF-1R)/mammalian target of rapamycin (mTOR) pathway is well described in ACC. Cixutumumab, a fully human IgG1 monoclonal antibody directed at IGF-1R was combined with temsirolimus on the basis of preclinical data. METHODS: Patients received cixutumumab, 3-6 mg kg(-1) intravenously (IV) weekly, and temsirolimus, 25-37.5 mg IV weekly (4-week cycles), with restaging after 8 weeks. RESULTS: Twenty-six patients were enrolled (13 (50%) men); median age, 47 years; median number of prior therapies, 4. Five patients previously received an IGF-1R inhibitor and one, temsirolimus. The most frequent toxicities, at least possibly drug related, were grade 1-2 thrombocytopenia (38%), mucositis (58%), hypercholesterolaemia (31%), hypertriglyceridemia (35%), and hyperglycaemia (31%). In all, 11 of 26 patients (42%) achieved stable disease (SD) >6 months (duration range=6-21 months) with 3 of the 11 having received a prior IGF-1R inhibitor. CONCLUSION: Cixutumumab combined with temsirolimus was well tolerated and >40% of patients achieved prolonged SD.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Sirolimus/analogs & derivatives , Adrenocortical Carcinoma/drug therapy , Adult , Aged , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Disease-Free Survival , Female , Humans , Male , Middle Aged , Sirolimus/administration & dosage , Young Adult
13.
Ann Oncol ; 24(3): 784-91, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23104723

ABSTRACT

BACKGROUND: Type 1 insulin-like growth factor receptor (IGF-1R) mediates resistance to chemotherapy and targeted agents. This study assessed the safety, pharmacokinetics (PK), and tolerability of humanized IGF-1R antibody AVE1642 with other cancer treatments. PATIENTS: Patients with advanced solid tumors received three weekly AVE1642 dosed at 6 mg/kg, chosen following previous study, with 75 (cohort A) or 100 mg/m(2) (B) docetaxel, 1250 mg/m(2) gemcitabine/100 mg erlotinib (C1), or 60 mg/m(2) doxorubicin (D1). Blood samples were assayed for PK, IGFs, and IGF-BP3. RESULTS: Fifty-eight patients received 317 AVE1642 infusions. The commonest adverse events were diarrhea (37/58 patients), asthenia (34/58), nausea (30/58), and stomatitis (21/58). Dose-limiting toxic effects in cohorts C1 (diarrhea) and D1 (neutropenia) prompted addition of cohorts C2 (1000 mg/m(2) gemcitabine/75 mg erlotinib) and D2 (50 mg/m(2) doxorubicin). Grade 3-4 hyperglycemia (three cases) accompanied steroid premedication for docetaxel administration. No PK interactions were detected. There were three partial responses in cohorts B (melanoma) and C (leiomyosarcoma, two cases) and 22 stabilizations ≥12 weeks, giving a control rate of 25/57 (44%). On treatment IGF-II rose by 68 ± 25 ng/ml in patients discontinuing treatment <12 weeks, and fell by 55.5 ± 21 ng/ml with disease control (P < 0.001). CONCLUSION: AVE1642 was tolerable with 75-100 mg/m(2) docetaxel and 1000 mg/m(2) gemcitabine/75 mg erlotinib, achieving durable disease control in 44%, with an association between IGF-II and response.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leiomyosarcoma/drug therapy , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Soft Tissue Neoplasms/drug therapy , Adult , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Diarrhea/chemically induced , Docetaxel , Doxorubicin/administration & dosage , Erlotinib Hydrochloride , Female , Humans , Male , Middle Aged , Quinazolines/administration & dosage , Receptor, IGF Type 1/immunology , Taxoids/administration & dosage , Treatment Outcome , Gemcitabine
14.
Br J Cancer ; 105(1): 44-52, 2011 Jun 28.
Article in English | MEDLINE | ID: mdl-21629245

ABSTRACT

BACKGROUND: The objectives of this phase I study were to determine the safety, pharmacokinetics (PK), pharmacodynamics and efficacy of brivanib combined with full-dose cetuximab in patients with advanced gastrointestinal malignancies. METHODS: Patients with advanced gastrointestinal malignancies who had failed prior therapies received brivanib (320, 600 or 800 mg daily) plus cetuximab (400 mg m(-2) loading dose then 250 mg m(-2) weekly). Assessments included adverse events, PK, tumour response, 2[18F]fluoro-2-deoxyglucose positron-emitting tomography and K-Ras mutation analyses. RESULTS: Toxicities observed were manageable; the most common treatment-related toxicities (>10% of patients) were fatigue, diarrhoea, anorexia, increase in aspartate aminotransferase and alanine aminotransferase, acneiform dermatitis, headache, mucosal inflammation, nausea, dry skin, vomiting, hypertension, pruritus, proteinuria and weight loss. Of 62 patients, 6 (9.7%) had objective radiographic partial responses, with an overall response rate of 10%. Median duration of response was 9.2 months; median progression-free survival was 3.9 months. CONCLUSIONS: The acceptable toxicity profile and efficacy of brivanib observed in this study were promising. These findings are being further evaluated in a phase III study of brivanib plus cetuximab vs cetuximab alone in patients previously treated with combination chemotherapy for K-Ras wild-type advanced metastatic colorectal cancer.


Subject(s)
Alanine/analogs & derivatives , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Gastrointestinal Neoplasms/drug therapy , Salvage Therapy , Triazines/pharmacokinetics , Triazines/therapeutic use , Adult , Aged , Alanine/pharmacokinetics , Alanine/therapeutic use , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Cetuximab , Drug Therapy, Combination , Female , Gastrointestinal Neoplasms/pathology , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Survival Rate , Tissue Distribution , Treatment Outcome , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
15.
Br J Cancer ; 104(12): 1862-8, 2011 Jun 07.
Article in English | MEDLINE | ID: mdl-21610706

ABSTRACT

BACKGROUND: This phase I, open-label, first-in-human study determined dose-limiting toxicities (DLTs) and maximum tolerated dose (MTD) of PD 0332991, an oral cyclin-dependent kinase 4/6 inhibitor with potent anti-proliferative activity in vitro/vivo. METHODS: A total of 33 patients with retinoblastoma protein-positive advanced solid tumours or non-Hodgkin's lymphoma refractory to standard therapy or for which no therapy was available received PD 0332991 once daily (QD) for 14 days followed by 7 days off treatment (21-day cycles; Schedule 2/1). RESULTS: Six patients had DLTs (18%; four receiving 200 mg QD; two receiving 225 mg QD); the MTD was 200 mg QD. Treatment-related, non-haematological adverse events occurred in 29 patients (88%) during cycle 1 and 27 patients (82%) thereafter. Adverse events were generally mild-moderate. Of 31 evaluable patients, one with testicular cancer achieved a partial response; nine had stable disease (≥10 cycles in three cases). PD 0332991 was slowly absorbed (mean T(max) 4.2 h) and eliminated (mean half-life 26.7 h). Volume of distribution was large (mean 3241 l) with dose-proportional exposure. Using a maximum effective concentration model, neutropenia was proportional to exposure. CONCLUSION: PD 0332991 was generally well tolerated, with DLTs related mainly to myelosuppression. The MTD, 200 mg QD, is recommended for phase II study.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , Neoplasms/drug therapy , Piperazines/administration & dosage , Pyridines/administration & dosage , Adult , Aged , Drug Administration Schedule , Female , Humans , Lymphoma, Non-Hodgkin/drug therapy , Male , Maximum Tolerated Dose , Middle Aged , Piperazines/adverse effects , Piperazines/pharmacokinetics , Pyridines/adverse effects , Pyridines/pharmacokinetics , Retinoblastoma Protein/analysis
16.
Clin Pharmacol Ther ; 88(6): 818-23, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20980999

ABSTRACT

Pazopanib is an oral angiogenesis inhibitor of vascular endothelial growth factor (VEGF) receptor, platelet-derived growth factor receptor, and cytokine receptor. This open-label, randomized, crossover, phase I study evaluated the effect of low- and high-fat meals on the pharmacokinetics (PK) of pazopanib in patients with advanced solid tumors. Patients participated in either the lead-in cohort or randomized food-effect cohort. Patients in the lead-in cohort were administered a single dose of pazopanib 400 mg with a high-fat meal. Patients in the food-effect cohort were randomized to receive single doses of pazopanib 800 mg in fed condition (high- or low-fat meal) or fasting condition, in random sequence 14 days apart. After completion of the study, patients were given the opportunity to continue treatment with daily pazopanib 800 mg. Administration of pazopanib with both low- and high-fat meals increased maximum observed plasma concentration (C(max)) and area under the plasma concentration-time curve (AUC) by approximately twofold as compared with the corresponding values when administered to patients in the fasted condition. Therefore, pazopanib should be administered to patients in the fasted state so as to minimize within- and between-day variability in the systemic exposure to pazopanib in patients with cancer.


Subject(s)
Dietary Fats/metabolism , Food-Drug Interactions/physiology , Neoplasms/metabolism , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Sulfonamides/adverse effects , Sulfonamides/pharmacokinetics , Administration, Oral , Adult , Aged , Cohort Studies , Cross-Over Studies , Fatigue/chemically induced , Female , Humans , Hypertension/chemically induced , Indazoles , Male , Middle Aged , Neoplasms/drug therapy , Pyrimidines/administration & dosage , Sulfonamides/administration & dosage
17.
J Cell Biochem ; 99(4): 1021-39, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16927308

ABSTRACT

It has been well established that a functioning vascular supply is essential for solid tumor growth and metastases. In the absence of a viable vascular network, tumors are unable to grow beyond a few millimeters and therefore remain dormant. Initiation of angiogenesis allows for continued tumor growth and progression. Targeting tumor vasculature, either by inhibiting growth of new tumor blood vessels (antiangiogenic agents) or by destroying the already present tumor vessels (vascular disrupting agents; VDA), is an area of extensive research in the development of new antitumor agents. These two groups differ in their direct physiological target, the type or extent of disease that is likely to be susceptible, and the treatment schedule. VDAs target the established tumor blood vessels, resulting in tumor ischemia and necrosis. These agents show more immediate effects compared to antiangiogenic agents and may have more efficacy against advanced bulky disease. VDAs can be divided into two groups--ligand-bound and small molecule agents. Both VDA groups have demonstrated antitumor effects and tumor core necrosis, but consistently leave a thin rim of viable tumor cells at the periphery of the tumor. More evidence suggests VDAs will have their greatest effect in combination with conventional chemotherapy or other modes of treatment that attack this outer rim of cells.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Neoplasms/blood supply , Animals , Flavonoids/pharmacology , Ligands , Tubulin/metabolism , Xanthones/pharmacology
18.
Br J Cancer ; 91(3): 418-24, 2004 Aug 02.
Article in English | MEDLINE | ID: mdl-15238978

ABSTRACT

The epidermal growth factor receptor (EGFR) is a member of the erbB family of tyrosine kinase receptors (RTK). The EGFR is involved in cell proliferation, metastasis and angiogenesis, and is expressed in a large proportion of epithelial tumours. The two main classes of EGFR inhibitors in clinical trials are the RTK inhibitors and the monoclonal antibodies. The clinical development of EGFR inhibitors has introduced new challenges to the design of phase I, II, and III trials. Both classes of agents can be safely administered at doses sufficient to inhibit the EGFR system. Receptor tyrosine kinase inhibitors have been extensively evaluated in non-small-cell lung cancer. In this setting, gefitinib has demonstrated activity in patients who fail initial chemotherapy. Monoclonal antibodies have been developed in combination with cytotoxic chemotherapy in several tumour types, most notably colorectal and head and neck cancer. The preliminary results suggest an increase in response rate and time to progression with the combination of cetuximab and chemotherapy in both disease models. Future issues in the development of EGFR inhibitors include the identification of biologic predictors of response, combination with other targeted agents, and their utilisation in earlier stage malignancies.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , ErbB Receptors/physiology , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinazolines/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Cetuximab , Clinical Trials as Topic , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride , Gefitinib , Humans , Neoplasms/drug therapy , Neoplasms/physiopathology
19.
Invest New Drugs ; 21(1): 63-74, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12795531

ABSTRACT

The spicamycin derivative KRN5500 was considered as a potential anti-cancer agent based on in vitro and preclinical studies. A Phase I study involving 24 cancer patients in whom tumors were refractory to all other conventional therapies was conducted to determine the dose limiting toxicity, maximum tolerated dose, effectiveness, and pharmacokinetic parameters of this drug administered by 1-h IV infusion daily for five consecutive days every 3 weeks. Using an accelerated dose titration strategy, 8.4 mg/m2/d x 5 days was the maximum administered dose. Severe gastrointestinal and hepatic toxicities were observed at doses at or above 4.3 mg/m2/d x 5. The recommended Phase II dose i s 4.3mg/m2/d x 5. The distribution of KRN5500 followed a two-compartment model, and clearance did not decrease significantly over the dose range 0.8-8.4 mg/m2/d x 5. No significant correlation was observed between plasma levels and toxicity. No tumor responses were observed among the 14 patients evaluable for response.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Neoplasms/drug therapy , Purine Nucleosides/administration & dosage , Purine Nucleosides/pharmacokinetics , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Purine Nucleosides/adverse effects , Purine Nucleosides/pharmacology , Time Factors , Treatment Outcome
20.
Clin Cancer Res ; 7(11): 3336-42, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11705845

ABSTRACT

PURPOSE: XK469 (2-[4-(7-chloro-2-quinoxalinyloxy) phenoxy]propionic acid), a synthetic quinoxaline phenoxypropionic acid derivative, has broad activity against murine tumors and is entering Phase I clinical development as a topoisomerase IIbeta inhibitor. This study investigated the underlying molecular mechanism of XK469's effects on the cell cycle. EXPERIMENTAL DESIGN: Growth inhibition, cell cycle arrest, induction of p53 and p21 mRNA and protein, and cdc2 phosphorylation and kinase activity were studied in treated cells from the H460 lung cancer line and p21 and p53 knockout cells of the HCT 116 colon cancer line. RESULTS: XK469 arrested H460 cells at G(2)-M, which was associated with cdc2 phosphorylation and decreased cdc2 kinase activity. Moreover, XK469 stabilized p53 and subsequently increased p21(WAF1/CIP1). Furthermore, HCT116 p21(-/-) cells were less sensitive than wild-type cells to XK469-induced growth inhibition, but p53(+/+) and p53(-/-) cells were equally sensitive despite the absence of p21 induction in the p53(-/-) cells. CONCLUSIONS: When considered with published data, our study suggests a complex mechanism of XK469-mediated anticancer activity involving multiple pathways, including p53-dependent and -independent G(2)-M arrest via inactivation of cdc2-cyclin B1 kinase activity.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Quinoxalines/pharmacology , Tumor Suppressor Protein p53/physiology , Cell Cycle/physiology , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Cyclins/physiology , Dose-Response Relationship, Drug , G2 Phase/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mitosis/drug effects , Mutation , Neoplasms/pathology , Neoplasms/prevention & control , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...