Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Oncologist ; 2024 May 30.
Article in English | MEDLINE | ID: mdl-38815166

ABSTRACT

The prognosis of patients with glioblastoma (GBM) remains poor despite current treatments. Targeted therapy in GBM has been the subject of intense investigation but has not been successful in clinical trials. The reasons for the failure of targeted therapy in GBM are multifold and include a lack of patient selection in trials, the failure to identify driver mutations, and poor blood-brain barrier penetration of investigational drugs. Here, we describe a case of a durable complete response in a newly diagnosed patient with GBM with leptomeningeal dissemination and PTPRZ1-MET fusion who was treated with tepotinib, a brain-penetrant MET inhibitor. This case of successful targeted therapy in a patient with GBM demonstrates that early molecular testing, identification of driver molecular alterations, and treatment with brain-penetrant small molecule inhibitors have the potential to change the outcome in select patients with GBM.

2.
Abdom Radiol (NY) ; 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38530430

ABSTRACT

BACKGROUND AND PURPOSE: The objective is to demonstrate feasibility of quantitative susceptibility mapping (QSM) in autosomal dominant polycystic kidney disease (ADPKD) patients and to compare imaging findings with traditional T1/T2w magnetic resonance imaging (MRI). METHODS: Thirty-three consecutive patients (11 male, 22 female) diagnosed with ADPKD were initially selected. QSM images were reconstructed from the multiecho gradient echo data and compared to co-registered T2w, T1w, and CT images. Complex cysts were identified and classified into distinct subclasses based on their imaging features. Prevalence of each subclass was estimated. RESULTS: QSM visualized two renal calcifications measuring 9 and 10 mm and three pelvic phleboliths measuring 2 mm but missed 24 calcifications measuring 1 mm or less and 1 larger calcification at the edge of the field of view. A total of 121 complex T1 hyperintense/T2 hypointense renal cysts were detected. 52 (43%) Cysts appeared hyperintense on QSM consistent with hemorrhage; 60 (49%) cysts were isointense with respect to simple cysts and normal kidney parenchyma, while the remaining 9 (7%) were hypointense. The presentation of the latter two complex cyst subtypes is likely indicative of proteinaceous composition without hemorrhage. CONCLUSION: Our results indicate that QSM of ADPKD kidneys is possible and uniquely suited to detect large renal calculi without ionizing radiation and able to identify properties of complex cysts unattainable with traditional approaches.

3.
Clin Cancer Res ; 28(6): 1117-1126, 2022 03 15.
Article in English | MEDLINE | ID: mdl-34789481

ABSTRACT

PURPOSE: Primary analysis of VISION showed tepotinib had durable clinical activity in patients with MET exon 14 (METex14) skipping non-small cell lung cancer (NSCLC). We present updated outcomes for clinically relevant subgroups. PATIENTS AND METHODS: This phase II, open-label, multi-cohort study of 500 mg (450 mg active moiety) tepotinib in patients with METex14 skipping NSCLC assessed efficacy and safety in predefined subgroups according to age, prior therapies (chemotherapy and immune checkpoint inhibitors), and brain metastases. An ad hoc retrospective analysis using Response Assessment in Neuro-Oncology Brain Metastases (RANO-BM) criteria assessed intracranial activity. RESULTS: 152 patients were evaluable for efficacy (median age: 73.1). Overall, objective response rate (ORR) was 44.7% [95% confidence interval (CI): 36.7-53.0]. Patients aged <75 (n = 84) and ≥75 (n = 68) had ORRs of 48.8% (95% CI: 37.7-60.0) and 39.7% (95% CI: 28.0-52.3), respectively. Treatment-naïve (n = 69) versus previously treated (n = 83) patients showed consistent efficacy [ORR (95% CI): 44.9% (32.9-57.4) vs. 44.6% (33.7-55.9); median duration of response (95% CI): 10.8 (6.9-not estimable) vs. 11.1 (9.5-18.5) months]. Of 15 patients analyzed by RANO-BM (12 received prior radiotherapy), 13 achieved intracranial disease control; 5 of 7 patients with measurable brain metastases had partial intracranial responses. Of 255 patients evaluable for safety, 64 (25.1%) experienced grade ≥3 treatment-related adverse events (TRAE), leading to discontinuation in 27 patients (10.6%). Rates of adverse events (AE) were broadly consistent irrespective of prior therapies. CONCLUSIONS: Tepotinib showed meaningful activity across subgroups by age, prior therapies, and brain metastases, with a manageable safety profile and few treatment discontinuations. See related commentary by Rosner and Spira, p. 1055.


Subject(s)
Brain Neoplasms , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Piperidines , Pyridazines , Pyrimidines , Aged , Antineoplastic Agents/adverse effects , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cohort Studies , Exons , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Piperidines/adverse effects , Pyridazines/adverse effects , Pyrimidines/adverse effects , Retrospective Studies
4.
Future Oncol ; 18(9): 1039-1054, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34918545

ABSTRACT

MET amplification (METamp), a mechanism of acquired resistance to EGFR tyrosine kinase inhibitors, occurs in up to 30% of patients with non-small-cell lung cancer (NSCLC) progressing on first-line osimertinib. Combining osimertinib with a MET inhibitor, such as tepotinib, an oral, highly selective, potent MET tyrosine kinase inhibitor, may overcome METamp-driven resistance. INSIGHT 2 (NCT03940703), an international, open-label, multicenter phase II trial, assesses tepotinib plus osimertinib in patients with advanced/metastatic EGFR-mutant NSCLC and acquired resistance to first-line osimertinib and METamp, determined centrally by fluorescence in situ hybridization (gene copy number ≥5 and/or MET/CEP7 ≥2) at time of progression. Patients will receive tepotinib 500 mg (450 mg active moiety) plus osimertinib 80 mg once-a-day. The primary end point is objective response, and secondary end points include duration of response, progression-free survival, overall survival and safety. Trial registration number: NCT03940703 (clinicaltrials.gov).


Osimertinib is used to treat a type of lung cancer that has specific changes (mutations) in a gene called EGFR. Although tumors will usually shrink (respond) during treatment with osimertinib, they can stop responding, or become resistant, to osimertinib. A common cause of resistance is 'MET amplification', which describes when extra copies of a gene called MET are present. Lung cancer that is resistant to osimertinib due to MET amplification could be treated by combining osimertinib with a treatment that blocks MET, such as tepotinib. INSIGHT 2 is an ongoing study that is designed to learn about the effects and safety of tepotinib combined with osimertinib, in patients with lung cancer that has stopped responding to osimertinib because of MET amplification. A plain language version of this article is available and is published alongside the paper online: www.futuremedicine.com/doi/suppl/10.2217/fon-2021-1406.


Subject(s)
Acrylamides/therapeutic use , Aniline Compounds/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm , Lung Neoplasms/drug therapy , Neoplasm Metastasis , Piperidines/therapeutic use , Pyridazines/therapeutic use , Pyrimidines/therapeutic use , Acrylamides/administration & dosage , Aniline Compounds/administration & dosage , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/secondary , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Piperidines/administration & dosage , Progression-Free Survival , Pyridazines/administration & dosage , Pyrimidines/administration & dosage
5.
Jpn J Clin Oncol ; 51(8): 1261-1268, 2021 Aug 01.
Article in English | MEDLINE | ID: mdl-34037224

ABSTRACT

BACKGROUND: MET exon 14 skipping is an oncogenic driver occurring in 3-4% of non-small cell lung cancer (NSCLC). The MET inhibitor tepotinib has demonstrated clinical efficacy in patients with MET exon 14 skipping NSCLC. Here, we present data from Japanese patients in the Phase II VISION study, evaluating the efficacy and safety of tepotinib. METHODS: In the open-label, single-arm, Phase II VISION study, patients with advanced/metastatic NSCLC with MET exon 14 skipping received oral tepotinib 500 mg once daily. The primary endpoint was objective response by independent review. Subgroup analyses of Japanese patients were preplanned. RESULTS: As of 1 January 2020, 19 Japanese patients received tepotinib and were evaluated for safety, 15 of whom had ≥9 months' follow-up and were also analysed for efficacy. By independent review, objective response rate (ORR) was 60.0% (95% confidence interval [CI]: 32.3, 83.7), median duration of response was not reached (95% CI: 6.9, not estimable [ne]), and progression-free survival was 11.0 months (95% CI: 1.4, ne). ORR in patients with MET exon 14 skipping identified by liquid biopsy (n = 8) was 87.5% (95% CI: 47.3, 99.7), and by tissue biopsy (n = 12) was 50.0% (95% CI: 21.1, 78.9). Patients' quality of life was maintained with tepotinib treatment. Among patients evaluated for safety, the most common treatment-related adverse events (any grade) were blood creatinine increase and peripheral oedema (12 and nine patients, respectively). CONCLUSIONS: Tepotinib demonstrated robust and durable clinical efficacy in Japanese patients with advanced NSCLC harbouring MET exon 14 skipping, identified by either liquid or tissue biopsy. The main adverse events, blood creatinine increase and peripheral oedema, were manageable.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Piperidines , Proto-Oncogene Proteins c-met , Pyridazines , Pyrimidines , Aged , Aged, 80 and over , Antineoplastic Agents/radiation effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Clinical Trials, Phase II as Topic , Exons/genetics , Female , Humans , Japan , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Male , Middle Aged , Mutation , Protein Kinase Inhibitors/adverse effects , Proto-Oncogene Mas , Proto-Oncogene Proteins c-met/genetics , Quality of Life , Retrospective Studies
6.
iScience ; 23(12): 101832, 2020 Dec 18.
Article in English | MEDLINE | ID: mdl-33305187

ABSTRACT

Tepotinib is an oral MET inhibitor approved for metastatic non-small cell lung cancer (NSCLC) harboring MET exon 14 (METex14) skipping mutations. Examining treatment-naive or tepotinib-resistant cells with MET amplification or METex14 skipping mutations identifies other receptor tyrosine kinases (RTKs) that co-exist in cells prior to tepotinib exposure and become more prominent upon tepotinib resistance. In a small cohort of patients with lung cancer with MET genetic alterations treated with tepotinib, gene copy number gains of other RTKs were found at baseline and affected treatment outcome. An Src homology 2 domain-containing phosphatase 2 (SHP2) inhibitor delayed the emergence of tepotinib resistance and synergized with tepotinib in treatment-naive and tepotinib-resistant cells as well as in xenograft models. Alternative signaling pathways potentially diminish the effect of tepotinib monotherapy, and the combination of tepotinib with an SHP2 inhibitor enables the control of tumor growth in cells with MET genetic alterations.

7.
JAMA Oncol ; 5(2): e184475, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30543347

ABSTRACT

Importance: The phosphatidylinositol 3-kinase (PI3K) pathway is frequently activated in patients with estrogen receptor-positive (ER+), endocrine therapy-resistant breast cancers. Objective: To assess the maximum tolerated dose (MTD), safety, and activity of alpelisib, an oral, PI3Kα-specific inhibitor, plus fulvestrant in patients with ER+ advanced breast cancer (ABC). Design, Setting, and Participants: An open-label, single-arm, phase 1b study of alpelisib plus fulvestrant was conducted at 10 centers in 5 countries. Participants were 87 postmenopausal women with PIK3CA-altered or PIK3CA-wild-type ER+ ABC, whose cancer progressed during or after antiestrogen therapy. The study began enrolling patients October 5, 2010, and the data cutoff was March 22, 2017. Interventions: Escalating doses of alpelisib were administered once daily, starting at 300 mg, plus fixed-dose fulvestrant, 500 mg, in the dose-escalation phase; alpelisib at the recommended phase 2 dose plus fulvestrant in the dose-expansion phase. Main Outcomes and Measures: The primary end point was determination of the MTD of once-daily alpelisib plus fulvestrant. Secondary end points included safety and preliminary activity. Results: From October 5, 2010, to March 22, 2017, 87 women (median age: 58 years [range, 37-79 years]; median of 5 prior lines of antineoplastic therapy) received escalating once-daily doses of alpelisib (300 mg, n = 9; 350 mg, n = 8; 400 mg, n = 70) plus fixed-dose fulvestrant (500 mg). During dose escalation, dose-limiting toxic effects were reported in 1 patient (alpelisib, 400 mg): diarrhea (grade 2), vomiting, fatigue, and decreased appetite (all grade 3). The MTD of alpelisib when combined with fulvestrant was 400 mg once daily, and the recommended phase 2 dose was 300 mg once daily. Overall, the most frequent grade 3/4 adverse events with alpelisib, 400 mg, once daily (≥10% of patients), regardless of causality, were hyperglycemia (19 [22%]) and maculopapular rash (11 [13%]); 9 patients permanently discontinued therapy owing to adverse events. Median progression-free survival at the MTD was 5.4 months (95% CI, 4.6-9.0 months). Median progression-free survival with alpelisib, 300 to 400 mg, once daily plus fulvestrant was longer in patients with PIK3CA-altered tumors (9.1 months; 95% CI, 6.6-14.6 months) vs wild-type tumors (4.7 months; 95% CI, 1.9-5.6 months). Overall response rate in the PIK3CA-altered group was 29% (95% CI, 17%-43%), with no objective tumor responses in the wild-type group. Conclusions and Relevance: Alpelisib plus fulvestrant has a manageable safety profile in patients with ER+ ABC, and data suggest that this combination may have greater clinical activity in PIK3CA-altered vs wild-type tumors. Trial Registration: ClinicalTrials.gov identifier: NCT01219699.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/genetics , Estrogen Receptor Antagonists/administration & dosage , Fulvestrant/administration & dosage , Mutation , Protein Kinase Inhibitors/administration & dosage , Receptors, Estrogen/analysis , Thiazoles/administration & dosage , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/chemistry , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Disease Progression , Estrogen Receptor Antagonists/adverse effects , Female , Fulvestrant/adverse effects , Humans , Maximum Tolerated Dose , Middle Aged , Progression-Free Survival , Protein Kinase Inhibitors/adverse effects , Thiazoles/adverse effects , Time Factors
8.
Arthritis Res Ther ; 20(1): 155, 2018 07 27.
Article in English | MEDLINE | ID: mdl-30053896

ABSTRACT

BACKGROUND: This double-blind, active-controlled, randomized, multinational study evaluated the efficacy, safety, pharmacokinetics (PK), and immunogenicity of PF-06438179/GP1111 (IxifiTM/Zessly®), an infliximab biosimilar, vs infliximab (Remicade®) reference product sourced from the European Union (infliximab-EU) in biologic-naïve patients with moderate to severe active rheumatoid arthritis (RA) despite methotrexate therapy. This paper reports results from the initial 30-week treatment period. METHODS: Patients (N = 650) were stratified by geographic region and randomized 1:1 to PF-06438179/GP1111 or infliximab-EU (3 mg/kg intravenous at weeks 0, 2, and 6, then every 8 weeks). Dose escalation to 5 mg/kg was allowed starting at week 14 for patients with inadequate RA response. The primary endpoint was American College of Rheumatology criteria for ≥ 20% clinical improvement (ACR20) response at week 14. Therapeutic equivalence was declared if the two-sided 95% CI for the treatment difference was within the symmetric equivalence margin of ± 13.5%. Statistical analysis was also performed with a two-sided 90% CI using an asymmetric equivalence margin (- 12.0%, 15.0%). RESULTS: Patients (80.3% female; 79.4% seropositive) had a mean RA duration of 6.9 years, and mean baseline Disease Activity Score in 28 joints, four components based on C-reactive protein was 6.0 in both arms. Week 14 ACR20 in the intention-to-treat population was 62.7% for PF-06438179/GP1111 and 64.1% for infliximab-EU. Week 14 ACR20 using nonresponder imputation was 61.1% for PF-06438179/GP1111 and 63.5% for infliximab-EU, and the 95% (- 9.92%, 5.11%) and 90% (- 8.75%, 4.02%) CIs for the treatment difference (- 2.39%) were entirely contained within the prespecified symmetric and asymmetric equivalence margins, respectively. No differences were observed between arms for secondary efficacy endpoints. Overall postdose antidrug antibody (ADA) rates through week 30 were 48.6% and 51.2% for PF-06438179/GP1111 and infliximab-EU, respectively. Efficacy and immunogenicity were similar between treatments for patients with dose escalation (at or after week 14), as well as between treatments for patients without dose escalation. Safety profiles of PF-06438179/GP1111 and infliximab-EU were similar, with no clinically meaningful differences observed between arms, including after ADA development. Serum drug concentrations were similar between arms at each time point during the initial 30-week treatment period. CONCLUSION: PF-06438179/GP1111 and infliximab-EU demonstrated similar efficacy, safety, immunogenicity, and PK with or without dose escalation in patients with moderate to severe active RA on background methotrexate. TRIAL REGISTRATION: ClinicalTrials.gov, NCT02222493 . Registered on 21 August 2014. EudraCT, 2013-004148-49 . Registered on 14 July 2014.


Subject(s)
Antirheumatic Agents/pharmacokinetics , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Biosimilar Pharmaceuticals/pharmacokinetics , Biosimilar Pharmaceuticals/therapeutic use , Infliximab/pharmacokinetics , Infliximab/therapeutic use , Adult , Aged , Double-Blind Method , Female , Humans , Male , Methotrexate/therapeutic use , Middle Aged , Therapeutic Equivalency
9.
Kidney Int ; 83(4): 593-603, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23389418

ABSTRACT

Renal cells are used in basic research, disease models, tissue engineering, drug screening, and in vitro toxicology. In order to provide a reliable source of human renal cells, we developed a protocol for the differentiation of human embryonic stem cells into renal epithelial cells. The differentiated stem cells expressed markers characteristic of renal proximal tubular cells and their precursors, whereas markers of other renal cell types were not expressed or expressed at low levels. Marker expression patterns of these differentiated stem cells and in vitro cultivated primary human renal proximal tubular cells were comparable. The differentiated stem cells showed morphological and functional characteristics of renal proximal tubular cells, and generated tubular structures in vitro and in vivo. In addition, the differentiated stem cells contributed in organ cultures for the formation of simple epithelia in the kidney cortex. Bioreactor experiments showed that these cells retained their functional characteristics under conditions as applied in bioartificial kidneys. Thus, our results show that human embryonic stem cells can differentiate into renal proximal tubular-like cells. Our approach would provide a source for human renal proximal tubular cells that are not affected by problems associated with immortalized cell lines or primary cells.


Subject(s)
Bioartificial Organs , Cell Differentiation , Embryonic Stem Cells/physiology , Epithelial Cells/physiology , Kidney Tubules, Proximal/physiology , Tissue Engineering , Activins/pharmacology , Animals , Biomarkers/metabolism , Bioreactors , Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein 7/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Shape , Cells, Cultured , Dose-Response Relationship, Drug , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/transplantation , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/transplantation , Gene Expression Regulation, Developmental , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/transplantation , Mice , Mice, SCID , Organ Culture Techniques , Time Factors , Tissue Engineering/methods , Tretinoin/pharmacology
10.
Lancet Oncol ; 13(1): 33-42, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22056021

ABSTRACT

BACKGROUND: Findings from the phase 3 First-Line ErbituX in lung cancer (FLEX) study showed that the addition of cetuximab to first-line chemotherapy significantly improved overall survival compared with chemotherapy alone (hazard ratio [HR] 0·871, 95% CI 0·762-0·996; p=0·044) in patients with advanced non-small-cell lung cancer (NSCLC). To define patients benefiting most from cetuximab, we studied the association of tumour EGFR expression level with clinical outcome in FLEX study patients. METHODS: We used prospectively collected tumour EGFR expression data to generate an immunohistochemistry score for FLEX study patients on a continuous scale of 0-300. We used response data to select an outcome-based discriminatory threshold immunohistochemistry score for EGFR expression of 200. Treatment outcome was analysed in patients with low (immunohistochemistry score <200) and high (≥200) tumour EGFR expression. The primary endpoint in the FLEX study was overall survival. We analysed patients from the FLEX intention-to-treat (ITT) population. The FLEX study is registered with ClinicalTrials.gov, number NCT00148798. FINDINGS: Tumour EGFR immunohistochemistry data were available for 1121 of 1125 (99·6%) patients from the FLEX study ITT population. High EGFR expression was scored for 345 (31%) evaluable patients and low for 776 (69%) patients. For patients in the high EGFR expression group, overall survival was longer in the chemotherapy plus cetuximab group than in the chemotherapy alone group (median 12·0 months [95% CI 10·2-15·2] vs 9·6 months [7·6-10·6]; HR 0·73, 0·58-0·93; p=0·011), with no meaningful increase in side-effects. We recorded no corresponding survival benefit for patients in the low EGFR expression group (median 9·8 months [8·9-12·2] vs 10·3 months [9·2-11·5]; HR 0·99, 0·84-1·16; p=0·88). A treatment interaction test assessing the difference in the HRs for overall survival between the EGFR expression groups suggested a predictive value for EGFR expression (p=0·044). INTERPRETATION: High EGFR expression is a tumour biomarker that can predict survival benefit from the addition of cetuximab to first-line chemotherapy in patients with advanced NSCLC. Assessment of EGFR expression could offer a personalised treatment approach in this setting. FUNDING: Merck KGaA.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/metabolism , Lung Neoplasms/drug therapy , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers, Tumor/antagonists & inhibitors , Brazil , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Cetuximab , Cisplatin/administration & dosage , Disease-Free Survival , ErbB Receptors/antagonists & inhibitors , Europe , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lung Neoplasms/enzymology , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Odds Ratio , Patient Selection , Proportional Hazards Models , Prospective Studies , Protein Kinase Inhibitors/administration & dosage , Republic of Korea , Risk Assessment , Risk Factors , Survival Rate , Time Factors , Treatment Outcome , Up-Regulation , Vinblastine/administration & dosage , Vinblastine/analogs & derivatives , Vinorelbine , Young Adult
11.
Lancet Oncol ; 12(1): 30-7, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21169060

ABSTRACT

BACKGROUND: The randomised phase 3 First-Line Erbitux in Lung Cancer (FLEX) study showed that the addition of cetuximab to cisplatin and vinorelbine significantly improved overall survival compared with chemotherapy alone in the first-line treatment of advanced non-small-cell lung cancer (NSCLC). The main cetuximab-related side-effect was acne-like rash. Here, we assessed the association of this acne-like rash with clinical benefit. METHODS: We did a subgroup analysis of patients in the FLEX study, which enrolled patients with advanced NSCLC whose tumours expressed epidermal growth factor receptor. Our landmark analysis assessed if the development of acne-like rash in the first 21 days of treatment (first-cycle rash) was associated with clinical outcome, on the basis of patients in the intention-to-treat population alive on day 21. The FLEX study is registered with ClinicalTrials.gov, number NCT00148798. FINDINGS: 518 patients in the chemotherapy plus cetuximab group-290 of whom had first-cycle rash-and 540 patients in the chemotherapy alone group were alive on day 21. Patients in the chemotherapy plus cetuximab group with first-cycle rash had significantly prolonged overall survival compared with patients in the same treatment group without first-cycle rash (median 15·0 months [95% CI 12·8-16·4] vs 8·8 months [7·6-11·1]; hazard ratio [HR] 0·631 [0·515-0·774]; p<0·0001). Corresponding significant associations were also noted for progression-free survival (median 5·4 months [5·2-5·7] vs 4·3 months [4·1-5·3]; HR 0·741 [0·607-0·905]; p=0·0031) and response (rate 44·8% [39·0-50·8] vs 32·0% [26·0-38·5]; odds ratio 1·703 [1·186-2·448]; p=0·0039). Overall survival for patients without first-cycle rash was similar to that of patients that received chemotherapy alone (median 8·8 months [7·6-11·1] vs 10·3 months [9·6-11·3]; HR 1·085 [0·910-1·293]; p=0·36). The significant overall survival benefit for patients with first-cycle rash versus without was seen in all histology subgroups: adenocarcinoma (median 16·9 months, [14·1-20·6] vs 9·3 months [7·7-13·2]; HR 0·614 [0·453-0·832]; p=0·0015), squamous-cell carcinoma (median 13·2 months [10·6-16·0] vs 8·1 months [6·7-12·6]; HR 0·659 [0·472-0·921]; p=0·014), and carcinomas of other histology (median 12·6 months [9·2-16·4] vs 6·9 months [5·2-11·0]; HR 0·616 [0·392-0·966]; p=0·033). INTERPRETATION: First-cycle rash was associated with a better outcome in patients with advanced NSCLC who received cisplatin and vinorelbine plus cetuximab as a first-line treatment. First-cycle rash might be a surrogate clinical marker that could be used to tailor cetuximab treatment for advanced NSCLC to those patients who would be most likely to derive a significant benefit.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Exanthema/chemically induced , Lung Neoplasms/drug therapy , Adult , Aged , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Carcinoma, Non-Small-Cell Lung/mortality , Cetuximab , Female , Humans , Lung Neoplasms/mortality , Male , Middle Aged
12.
Biomaterials ; 31(5): 863-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19878994

ABSTRACT

Hydrogel scaffolds are highly hydrated polymer networks that allow cells to adhere, proliferate and differentiate in the treatment of diseased or injured tissues and organs. Using hydrodynamic shaping and in situ cross-linking of hydrogel precursors, we have developed a highly efficient "hydrodynamic spinning" approach for synthesizing hydrogel fibers of different diameters in a multiphase coaxial flow. A triple-orifice spinneret has been created, and three different types of hydrogel precursors have been examined. Without changing the spinning head, hollow and solid hydrogel fibers with different diameters have been spun by simply manipulating the ratio of input flow rates. Together with the ability of simultaneous cell-seeding in the hydrogel matrix, hydrodynamic spinning can be broadly applied to many hydrogel materials, providing a powerful technique in the preparation of fiber-like and tubule-like hydrogel constructs for tissue engineering.


Subject(s)
Biocompatible Materials/chemistry , Hydrogels/chemistry , Kidney/cytology , Kidney/physiology , Microfluidics/methods , Tissue Engineering/methods , Absorption , Animals , Biomimetic Materials/chemistry , Cell Culture Techniques/methods , Cell Line , Cell Survival , Cells, Cultured , Crystallization/methods , Dogs , Extracellular Matrix/chemistry , Materials Testing , Particle Size , Porosity , Rotation , Surface Properties
13.
Biomaterials ; 30(21): 3523-31, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19328545

ABSTRACT

Gelatin-hydroxyphenylpropionic acid (Gtn-HPA) hydrogels are highly porous and biodegradable materials. Herein we report a fiber spinning method that can produce cell-seeded solid and hollow hydrogel fibers by enzymatically cross-linking Gtn-HPA in solutions flowing within a capillary tube. The cell-immobilized hydrogel fibers, with feature sizes down to 20 microm, are formed as a result of continuous cross-linking of cell-mixed hydrogel precursors in a multiphase laminar flow. This fiber formation process is mild enough to retain the cell viability. The continuous fiber formation, simultaneous cell encapsulation, as well as versatile combination of fiber structures provided by this approach make it a promising and effective technique for the preparation of cell-seeded hydrogel scaffolds and carriers for tissue engineering.


Subject(s)
Gelatin/chemistry , Hydrogels/chemistry , Phenylpropionates/chemistry , Microscopy, Electron, Scanning , Molecular Structure , Tissue Engineering/methods
14.
Langmuir ; 24(6): 2611-7, 2008 Mar 18.
Article in English | MEDLINE | ID: mdl-18260679

ABSTRACT

The presentation of bioactive ligands on biomaterial surfaces is often confounded by the adsorption of proteins present in the biological milieu, rendering any type of cellular response nonspecific. We have engineered a polyelectrolyte complex membrane that demonstrates specific adhesion of various cell types for both two-dimensional (2D) and three-dimensional (3D) cell culture systems. Specific cell adhesion is achieved by a three-tiered structure: a silica cross-linked polycation as the bottom (first) tier, a nonfouling polyanion-poly(ethylene glycol) (PEG) conjugate as the intermediate (second) tier, and the cell-adhesion ligand as the top (third) tier. Each tier of the membrane was characterized in terms of chemical composition and dimensions. Epithelial cells (primary human cortical renal cells and a hepatocellular carcinoma cell line) cultured on the membranes exhibited little cell attachment on the polyanion-PEG second tier and good cell adhesion on the RGD-modified third tier. Thus, the second tier allowed the effect of cell adhesion due to the ligand (third tier) to be isolated and distinguished from nonspecific cell attachment to the first tier. For the culturing of cells in three dimensions, the three-tiered membrane system was applied using a highly swellable chitosan membrane as the first tier. The resulting cell-membrane construct was uniformly dispersed and centrifuged to form a matrix that interacted intimately with cells in the form of a pellet. Presentation of RGD in the latter format enhanced the viability of human mesenchymal stem cells (hMSCs) over controls without RGD.


Subject(s)
Epithelial Cells/chemistry , Membranes, Artificial , Oligopeptides/chemistry , Polyethylene Glycols/chemistry , Cell Adhesion , Cells, Cultured , Electrolytes/chemistry , Humans , Ligands , Molecular Structure , Particle Size , Surface Properties , Time Factors
15.
Tissue Eng ; 13(1): 197-205, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17518593

ABSTRACT

Cultured precision-cut liver tissue slices are useful for studying the metabolism and toxicity of xenobiotics in liver. They may also be used to investigate the behavior of and interaction between different cell types in an intact histo-architecture. Because cultured liver tissues undergo a loss of function and morphology because of their separation from the blood supply, we investigated changes in key protein marker expressions in parenchymal and non-parenchymal cells, as well as in the extracellular matrix (ECM) at different time points. We also compared conventional culture methods such as static and dynamic cultures with perfusion culture, which allows a continuous exchange of the culture medium. In conventional culture methods, the expression of vimentin and collagen type IV decreased after 5 h in the non-parenchymal cells and the ECM, respectively, whereas the hepatocyte nuclear factor 4 alpha (HNF4alpha) staining in the hepatocytes remained constant. In perfusion culture, on the other hand, vimentin, collagen type IV, and HNF4alpha staining were clearly detectable after 5 h. The histo-architecture obtained from perfusion culture was also more compact than those obtained from conventional culture methods. After 24 h, only the perfusion cultured sample retained protein marker expression in all components of the liver tissue. Our results suggest that, to develop improved culture techniques for liver slices, changes at the early time-points should be taken into consideration. Our results also show that culture techniques that enable a continuous exchange of the culture medium seem to be superior to static or dynamic cultures in terms of maintaining the protein expression and the histo-architecture.


Subject(s)
Liver , Perfusion , Tissue Culture Techniques/methods , Animals , Biomarkers/metabolism , Gene Expression Regulation/physiology , Hepatocytes/cytology , Hepatocytes/metabolism , Liver/cytology , Liver/metabolism , Male , Perfusion/instrumentation , Perfusion/methods , Proteins/genetics , Proteins/metabolism , Rats , Rats, Wistar , Tissue Culture Techniques/instrumentation
16.
Cell Tissue Res ; 319(3): 491-500, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15668799

ABSTRACT

Growth of the kidney is a complex process piloted by the collecting duct (CD) ampullae. The dichotomous arborisation and consecutive elongation of this tubular element determines the exact site and time for the induction of nephrons in the overlaying mesenchymal cap condensates. The mechanism by which the CD ampullae find the correct orientation is currently unknown. Recently, we have demonstrated micro-fibres that originate from the basal aspect of the CD ampullae and extend through the mesenchyme to the organ capsule. The micro-fibres are assumed to be involved in the growth and arborisation process of the CD ampulla. Therefore, we have investigated the specific distribution of the micro-fibres during branching morphogenesis. We have also analysed whether the micro-fibres co-localise with extracellular matrix (ECM)-modulating enzymes and whether the co-localisation pattern changes during CD ampulla arborisation. Micro-fibres were detected in all stages of CD ampulla arborisation. Tissue transglutaminase (Tgase2) co-localised with soybean agglutinin (SBA)-positive micro-fibres, whose presence depended upon the degree of CD branching. Matrix metalloproteinase-9 (MMP-9) also co-localised with micro-fibres, but its expression pattern was different from that for Tgase2. Western blotting experiments demonstrated that Tgase2 and MMP-9 co-migrated with SBA-labelled proteins. Thus, the micro-fibres are developmentally modulated by enzymes of the ECM in embryonic kidney cortex. These findings illustrate the importance of micro-fibres in directing CD ampulla growth.


Subject(s)
GTP-Binding Proteins/metabolism , Kidney Cortex/enzymology , Kidney Tubules, Collecting/enzymology , Matrix Metalloproteinase 9/metabolism , Microfibrils/enzymology , Plant Lectins/metabolism , Soybean Proteins/metabolism , Transglutaminases/metabolism , Animals , Animals, Newborn , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Extracellular Matrix/enzymology , Fluorescent Antibody Technique, Indirect , Kidney Cortex/embryology , Kidney Tubules, Collecting/embryology , Organogenesis , Protein Glutamine gamma Glutamyltransferase 2 , Rabbits
17.
Biomed Mater Eng ; 15(1-2): 51-63, 2005.
Article in English | MEDLINE | ID: mdl-15623930

ABSTRACT

In the organism epithelia perform perfect barrier functions. Strong rheological and mechanical influences constitute the normal environment of this tissue throughout life. Most epithelia are exposed to different fluids at the luminal and basal sides. To obtain realistic information about tissue development in modern biomaterial testing and tissue engineering it is necessary to mimick the natural environment of epithelia. Cultured cells are brought in contact with an artificial extracellular matrix to determine whether proper development into a functional epithelium occurs. As under natural conditions the cultures have to withstand mechanical and fluid stress over a prolonged period of time in close contact to a selected biomaterial. However, development of tissue-specific features such as polarization, tightness and transport under in vitro conditions will only occur, if the biomaterial and the culture conditions support tissue development. Leakage, edge damage and pressure differences during culture have to be avoided so that the natural functions of the growing epithelium can develop. Our aim is to generate functional epithelia derived from renal explants containing stem cells, which are microsurgically isolated and placed into specific O-ring carriers for optimal handling. The cells develop in combination with a collagenous matrix from an embryonic into a functional collecting duct (rCD) epithelium. To achieve optimal culture conditions the tissue is placed in a gradient culture container. A typical environment can be simulated by superfusing different culture media at the luminal and basal sides. Within days epithelia growing inside the gradient container build up a physiological barrier, which is maintained during the whole culture period. The described method allows to investigate the influence of new biomaterials over prolonged periods of time.


Subject(s)
Cell Culture Techniques/methods , Epithelial Cells/cytology , Kidney/cytology , Materials Testing/methods , Stem Cells/cytology , Tissue Engineering/methods , Animals , Animals, Newborn , Cell Culture Techniques/instrumentation , Cell Differentiation/physiology , Cell Proliferation , Cell Survival/physiology , Cells, Cultured , Epithelial Cells/physiology , Equipment Design , Equipment Failure Analysis , Kidney/embryology , Kidney/physiology , Materials Testing/instrumentation , Perfusion , Rabbits , Stem Cells/physiology , Tissue Engineering/instrumentation
18.
Cell Physiol Biochem ; 14(4-6): 387-94, 2004.
Article in English | MEDLINE | ID: mdl-15319542

ABSTRACT

During kidney development a multitude of tubular portions is formed. Little knowledge is available by which cellbiological mechanism a cluster of embryonic cells is able to generate the three-dimensional structure of a tubule. However, this know-how is most important in tissue engineering approaches such as the generation of an artificial kidney module or for the therapy of renal diseases using stem cells. To obtain cellbiological insights in parenchyme development we elaborate a new technique to generate under in vitro conditions renal tubules derived from the embryonic cortex of neonatal rabbits. The aim of the experiments is to establish a specific extracellular environment allowing optimal three-dimensional development of renal tubules under serum-free culture conditions. In the present paper we demonstrate features of the renal stem cell niche and show their isolation as intact microcompartments for advanced tissue culture. Perfusion culture in containers exhibiting a big dead fluid volume results in the development of a flat collecting duct (CD) epithelium at the surface of the tissue explant. In contrast, by fine-tuning the dead fluid volume within a perfusion culture container by an artificial interstitium made of a polyester fleece shows the generation of tubules. It is an up to date unknown morphogenetic information which tells the cells to form tubular structures.


Subject(s)
Kidney Tubules, Collecting/cytology , Kidney Tubules/cytology , Stem Cells/cytology , Tissue Engineering/methods , Animals , Biocompatible Materials/chemistry , Embryo, Mammalian/cytology , Epithelium/immunology , Epithelium/metabolism , Extracellular Space/chemistry , Kidney Tubules/metabolism , Kidney Tubules, Collecting/embryology , Laminin/analysis , Laminin/metabolism , Membrane Proteins/analysis , Membrane Proteins/metabolism , Occludin , Organ Culture Techniques , Perfusion/instrumentation , Perfusion/methods , Rabbits , Stem Cells/metabolism
19.
J Am Soc Nephrol ; 14(11): 2758-66, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14569085

ABSTRACT

Peanut agglutinin (PNA) represents a commonly used marker for beta-type intercalated (IC) cells and their distribution in the corticomedullary course of the collecting duct (CD) in the mature rabbit kidney. It has been shown that aldosterone is able to generate >90% of PNA-binding cells in an embryonic CD epithelium in vitro. In adult kidney, a maximum of only 25% PNA-positive cells is found in the cortical segment of the CD, and PNA-binding completely disappears in the inner-medullary CD. Molecules that regulate the gradual development of CD-specific cells during organ growth are unknown. In the present experiments, it was found that addition of physiologic concentrations of urea to the culture medium is able to restrain the action of aldosterone in embryonic CD epithelia. Urea antagonizes in a concentration-dependent manner the action of aldosterone finally leading to only 10% of PNA-binding cells. The data point to a urea-specific effect, because osmolytes such as NaCl and mannitol did not affect PNA binding. In addition, urea did not influence expression of principal-cell typical markers such as AQP2 and 3. The findings may explain that a higher number of PNA-positive cells is found in the cortical region of the kidney correlated with a low concentration of urea as compared with only few PNA-binding cells in the medullary CD, where a high concentration of urea occurs. Thus, an increasing concentration of urea may trigger the number of PNA-positive cells in the cortical-medullary course of the CD during organ development.


Subject(s)
Aldosterone/metabolism , Epithelial Cells/metabolism , Kidney Tubules, Collecting/embryology , Kidney Tubules, Collecting/metabolism , Peanut Agglutinin/metabolism , Urea/metabolism , Animals , Cell Culture Techniques , Protein Binding , Rabbits , Time Factors
20.
Pflugers Arch ; 447(2): 214-22, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14504926

ABSTRACT

Based on the controversy about the relevance of cyclooxygenase-2 (Cox-2)-derived prostanoids from the macula densa for the control of the renin system, this study aimed to determine the interrelation between Cox-2 and renin expression in the mouse kidney. In control mice renin mRNA was readily detectable whilst renocortical Cox-2 mRNA abundance was at the detection limit of the RNase protection assay and no specific signals for Cox-2 were obtained by in situ hybridization or Western blot analysis. Experimental maneuvers such as low-salt diet, treatment with loop diuretics or angiotensin I converting enzyme inhibitors clearly increased renin mRNA abundance up to sevenfold, but under none of these conditions renocortical Cox-2 mRNA levels were significantly changed. Moreover, the strong stimulation of renin expression by angiotensin I-converting enzyme inhibition was not changed by the cyclooxygenase inhibitor ibuprofen, which in turn clearly lowered tissue prostanoid content. Our data suggest a marked divergence of renin and Cox-2 expression in the kidney cortex of C57Bl/6 mice with no clear evidence for a role of Cox-2-derived prostanoids from the macula densa in the regulation of renin expression.


Subject(s)
Isoenzymes/metabolism , Kidney Cortex/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Renin/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Bumetanide/pharmacology , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Diet, Sodium-Restricted , Diuretics/pharmacology , Ibuprofen/pharmacology , Isoenzymes/genetics , Kidney Medulla/metabolism , Male , Mice , Mice, Inbred C57BL , Nuclease Protection Assays , Prostaglandin-Endoperoxide Synthases/genetics , RNA, Messenger/metabolism , Ramipril/pharmacology , Renin/genetics , Ribonucleases
SELECTION OF CITATIONS
SEARCH DETAIL
...