Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
2.
J Neurooncol ; 168(1): 35-45, 2024 May.
Article in English | MEDLINE | ID: mdl-38561565

ABSTRACT

PURPOSE: Maximal cardiopulmonary exercise testing (max. CPET) provides the most accurate measurement of cardiorespiratory fitness. However, glioblastoma (GBM) patients often undergo less intensive tests, e.g., 6-min walk test or self-rating scales. This study aims to demonstrate feasibility and safety of max. CPET in GBM patients, concurrently evaluating their physical fitness status. METHODS: Newly diagnosed GBM patients undergoing adjuvant chemotherapy were offered participation in an exercise program. At baseline, max. CPET assessed cardiorespiratory fitness including peak oxygen consumption (VO2peak), peak workload, and physical work capacity (PWC) at 75% of age-adjusted maximal heart rate (HR). Criteria for peak workload were predefined based on threshold values in HR, respiratory quotient, respiratory equivalent, lactate, and rate of perceived effort. Data were compared to normative values. Adverse events were categorized according to standardized international criteria. Further, self-reported exercise data pre- and post-diagnosis were gathered. RESULTS: All 36 patients (median-aged 60; 21 men) met the predefined criteria for peak workload. Mean absolute VO2peak was 1750 ± 529 ml/min, peak workload averaged 130 ± 43 W, and mean PWC was 0.99 ± 0.38 W/kg BW, all clinically meaningful lower than age- and sex-predicted normative values (87%, 79%, 90%, resp.). Only once (3%) a minor, transient side effect occurred (post-test dizziness, no intervention needed). Self-reported exercise decreased from 15.8 MET-h/week pre-diagnosis to 7.2 MET-h/week post-diagnosis. CONCLUSION: Max. CPET in this well-defined population proved feasible and safe. GBM patients exhibit reduced cardiorespiratory fitness, indicating the need for tailored exercise to enhance health and quality of life. CPET could be essential in establishing precise exercise guidelines.


Subject(s)
Brain Neoplasms , Exercise Test , Feasibility Studies , Glioblastoma , Physical Fitness , Humans , Male , Female , Middle Aged , Glioblastoma/drug therapy , Exercise Test/methods , Brain Neoplasms/drug therapy , Physical Fitness/physiology , Aged , Oxygen Consumption/drug effects , Adult , Cardiorespiratory Fitness/physiology
3.
J Neurooncol ; 163(2): 367-376, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37306887

ABSTRACT

PURPOSE: Exercise proved to reduce cancer-related symptoms and prolong survival in some cancer types. However, brain tumor patients are often advised against strenuous exercise. Here, we summarize our experience with a submaximal exercise program for glioma patients: ActiNO (Active in Neuro-Oncology). METHODS: Glioma patients were invited to participate in the program. Since 2011, a sports scientist individualized two one-hour sessions per week adapted to the patients' symptoms. One session consisted of bicycle ergometry (average workload: 75% of maximum heart rate), the other of whole-body resistance training. Both sessions were further complimented by coordinative elements. Cardiorespiratory fitness was assessed using the "Physical Work Capacity" procedure. Patients were followed up regularly to assess adherence to the program and disease activity. RESULTS: Until December 2019, 45 glioma patients, median-aged 49 years (IQR 42-59), were included in the analysis. Most patients suffered from glioblastoma (58%), followed by diffuse lower-grade astrocytoma (29%). In overall 1828 training sessions, two minor epileptic events occurred (1 speech arrest; 1 focal seizure). During fitness assessment, all patients achieved at least 75% of their age-adjusted maximum heart rate. Peak workload averaged 172 W (95% CI 156-187). Median survival of participating glioblastoma patients was 24.1 months (95% CI 8.6-39.5). CONCLUSION: This supervised training program with submaximal exertion was feasible and safe in glioma regardless of WHO grading. Based on these experiences, we initiated a prospective multicenter study to objectify improvements in physical performance and quality of life in patients with glioblastoma.


Subject(s)
Glioblastoma , Glioma , Humans , Quality of Life , Prospective Studies , Glioma/therapy , Exercise Therapy/methods
4.
Cancers (Basel) ; 13(6)2021 Mar 13.
Article in English | MEDLINE | ID: mdl-33805663

ABSTRACT

The COVID-19 pandemic is associated with significant morbidity, mortality, and restrictions on everyday life worldwide. This may be especially challenging for brain tumor patients given increased vulnerability due to their pre-existing condition. Here, we aimed to investigate the quality of life (QoL) in brain tumor patients and relatives in this setting. Over twelve weeks during the first wave of the pandemic (04-07/2020), brain tumor patients and their families from two large German tertiary care centers were asked to complete weekly questionnaires for anxiety, depression, distress, and well-being. Information regarding social support and living conditions was also collected. One hundred participants (63 patients, 37 relatives) completed 729 questionnaires over the course of the study. Compared to relatives, patients showed more depressive symptoms (p < 0.001) and reduced well-being (p = 0.013). While acceptance of lockdown measures decreased over time, QoL remained stable. QoL measures between patients and their families were weakly or moderately correlated. The number of social contacts was strongly associated with QoL. Age, living conditions, ongoing therapy, employment, and physical activity were other predictors. QoL is correlated between patients and their families and heavily depends on social support factors, indicating the need to focus on the entire family and their social situation for QoL interventions during the pandemic.

5.
J Clin Med ; 9(4)2020 Apr 02.
Article in English | MEDLINE | ID: mdl-32252441

ABSTRACT

A brain tumor diagnosis poses a significant psychological burden and it severely impacts quality of life (QOL), both in patients and relatives. However, comprehensive strategies addressing QOL in this setting remain rare. Here, we aim to share our findings of a one-week ski exercise intervention, with emphasis on feasibility, safety, QOL, and physical exercise. The intervention consisted of week-long daily ski sessions with professional ski guides as well as dedicated physicians present. The participants were handed questionnaires, including distress and QOL items before, during, and after the intervention. Using fitness watches, exercise intensity was also tracked at these timepoints. During the intervention, patients were checked for adverse events daily. Fifteen participants, nine patients after multidisciplinary treatment, and six relatives were included in the study. Additionally, 13 children participated in the exercise, but not in the study. All of the participants completed the entire program. No severe adverse events were documented during daily checks. There was a strong increase in quantified activity and QOL with a corresponding decrease in distress during the intervention, and, partly, afterwards. This prospective brain tumor rehabilitation study demonstrates the feasibility and safety of challenging ski exercise in brain tumor patients. The findings also underline the exercise-mediated QOL benefits, emphasizing the need for more comprehensive brain tumor rehabilitation programs.

6.
Med Sci Sports Exerc ; 51(12): 2429-2433, 2019 12.
Article in English | MEDLINE | ID: mdl-31730561

ABSTRACT

INTRODUCTION: Glioblastoma multiforme (GBM) carries a strongly unfavorable prognosis despite intensive multidisciplinary therapy. Physical exercise is rarely offered to patients for fear of adverse events such as falls, epileptic seizures, or bleeding, despite little supporting evidence. Here, we report a study of high-level and long-term exercise in a GBM patient. METHODS: A 33-yr-old male, diagnosed with a large cystic GBM, was included in our institution's Personal Training Program after initial tumor resection and adjuvant radiochemotherapy. The program was designed to facilitate individual long-term high-intensity exercise. Supervised by a certified personal trainer, it consisted of at least four weekly training sessions and intermittent performance diagnostics. An activity tracker quantified training intensity. RESULTS: In this setting, the patient exercised at high intensity without adverse events for 87 continuous weeks (21 months). He averaged 43.7 metabolic equivalent of task hours per week (MET·h·wk), well above the 75th percentile of healthy males the same age, while undergoing multiple surgeries, chemotherapy, and radiation therapy regimens. The patient completed two marathons averaging less than 5 min·km both times, despite tumor progression. Performance diagnostics indicated a gain of fitness even during continuous GBM treatment. Due to multiple intraventricular lesions and increasing intracranial pressure, training was stopped 6 wk before the patient passed away 2 yr after initial diagnosis. CONCLUSIONS: This study demonstrates that high-intensity, long-term physical training regimens are feasible in GBM patients during full multidisciplinary therapy. In this patient, the exercise was pursued without adverse events and led to a gain of fitness despite tumor progression and intensive multiple therapies. We conclude that, in GBM patients, exercise regimens require further study instead of general discouragement.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , High-Intensity Interval Training , Physical Fitness , Adult , Brain Neoplasms/surgery , Chemoradiotherapy, Adjuvant , Glioblastoma/surgery , Humans , Male , Prognosis , Quality of Life
7.
Sci Rep ; 7(1): 13998, 2017 10 25.
Article in English | MEDLINE | ID: mdl-29070896

ABSTRACT

Proteolytic enzymes have shown efficacy in cancer therapy. We present a combination of the two pro-enzymes Trypsinogen and Chymotrypsinogen A with potent in vitro and in vivo anti-tumour efficacy. A synergetic anti-tumour effect for Trypsinogen and Chymotrypsinogen A was determined at a ratio 1:6 (named PRP) using 24 human cancer cell lines. The antiangiogenic effect of PRP was analysed by matrigel-based tube formation and by fibrous capsule formation assays. Furthermore, cell invasion and wound healing assays together with qRT-PCR determination of epithelial-to-mesenchymal transition (EMT) markers were performed on human cancer cells treated with PRP. Additionally, in vivo pharmacokinetic studies were implemented and the PRP's anti-tumour efficacy was explored against orthotopic pancreatic and ovarian cancer tumours. PRP formulation was proven to inhibit in vitro angiogenesis, tumour growth, cancer cell migration and invasiveness; and to be an effective and well tolerated in vivo anti-tumour treatment. Finally, the clinical efficacy of a suppository formulation containing both pancreatic pro-enzymes in the context of a UK Pharmaceuticals Special Scheme was evaluated in advanced cancer patients. Consequently, PRP could have relevant oncological clinical applications for the treatment of advanced or metastatic pancreatic adenocarcinoma and advanced epithelial ovarian cancer.


Subject(s)
Chymotrypsinogen/administration & dosage , Enzyme Precursors/administration & dosage , Ovarian Neoplasms/prevention & control , Pancreas/enzymology , Pancreatic Neoplasms/prevention & control , Trypsinogen/administration & dosage , Animals , Apoptosis , Cell Proliferation , Female , Humans , Mice , Mice, Nude , Neoplasm Metastasis , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pilot Projects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
PLoS One ; 7(12): e52175, 2012.
Article in English | MEDLINE | ID: mdl-23300607

ABSTRACT

PG545 is a clinically relevant heparan sulfate (HS) mimetic which, in addition to possessing anti-angiogenic properties, also acts as a heparanase inhibitor which may differentiate its mechanism(s) of action from approved angiogenesis inhibitors. The degradation of HS by heparanase has been strongly implicated in cell dissemination and the metastatic process. Thus, the anti-metastatic activity of PG545 has been linked to the enzymatic function of heparanase - the only endoglycosidase known to cleave HS, an important component of the extracellular matrix (ECM) which represents a potential avenue for therapeutic intervention for certain metastatic cancer indications. Recent concerns raised about the paucity of overall survival as an endpoint in mouse models of clinically relevant metastasis led us to examine the effect of PG545 on the progression of both primary tumor growth and the spontaneously metastasizing disease in the 4T1 syngeneic breast carcinoma model in a non-surgical and surgical (mastectomy) setting. PG545 significantly inhibited primary tumor growth but importantly also inhibited lung metastasis in treated mice, an effect not observed with the tyrosine kinase inhibitor sorafenib. Importantly, PG545 significantly enhanced overall survival compared to vehicle control and the sorafenib group, suggesting PG545's inhibitory effect on heparanase is indeed a critical attribute to induce anti-metastatic activity. In addition to blocking a common angiogenic signalling pathway in tumor cells, the expression of heparanase in the primary tumor and lung was also significantly reduced by PG545 treatment. These results support the ongoing development of PG545 and highlight the potential utility in metastatic disease settings.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Glucuronidase/antagonists & inhibitors , Lung Neoplasms/mortality , Mammary Neoplasms, Experimental/mortality , Neovascularization, Pathologic/prevention & control , Saponins/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols , Cells, Cultured , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , Glucuronidase/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Immunoenzyme Techniques , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mice , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Phenylurea Compounds/pharmacology , Sorafenib
9.
Anticancer Drugs ; 22(1): 58-78, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20890178

ABSTRACT

The aim was to determine the potential of the allosteric mammalian target of rapamycin inhibitor, everolimus, to act in combination with cytotoxic anticancer compounds in vitro and in vivo. A concomitant combination in vitro showed no evidence of antagonism, but enhanced the antiproliferative effects (additive to synergistic) with cisplatin, doxorubicin, 5-fluorouracil, gemcitabine, paclitaxel, and patupilone. Everolimus (1-5 mg/kg/d orally) was evaluated for antitumor activity in vivo alone or in combination with suboptimal cytotoxic doses using athymic nude mice bearing subcutaneous human H-596 lung, KB-31 cervical, or HCT-116 colon tumor xenografts. Everolimus monotherapy was very well tolerated and caused inhibition of tumor growth, rather than regression, and this was associated with a dose-dependent decline in tumor pS6 levels, a key downstream protein of mammalian target of rapamycin. At the doses used, the cytotoxics inhibited tumor growth and caused tolerable body-weight loss. Concomitant combinations of cisplatin, doxorubicin, paclitaxel, or patupilone with everolimus produced cooperative antitumor effects, in some cases producing regressions without clinically significant increases in toxicity. In contrast, combinations with gemcitabine and 5-fluorouracil were less well tolerated. Alternative administration schedules were tested for cisplatin, gemcitabine, or paclitaxel combined with everolimus: these did not dramatically affect cisplatin or gemcitabine activity or tolerability but were antagonistic for paclitaxel. Everolimus showed promising maintenance activity after treatment with doxorubicin or paclitaxel ceased. Overall, the results confirm that everolimus is an effective, well-tolerated suppressor of experimental human tumor growth, and although it did not show strong potentiation of efficacy, antitumor activity in vivo was increased without marked increases in toxicity, supporting clinical use of everolimus as a partner for conventional cytotoxics.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/administration & dosage , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Drug Screening Assays, Antitumor , Drug Synergism , Epothilones/administration & dosage , Everolimus , Female , Fluorouracil/administration & dosage , HCT116 Cells , Humans , Mice , Mice, Nude , Paclitaxel/administration & dosage , Paclitaxel/antagonists & inhibitors , Sirolimus/administration & dosage , Sirolimus/analogs & derivatives , Xenograft Model Antitumor Assays , Gemcitabine
10.
Cancer Res ; 70(4): 1544-54, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20145120

ABSTRACT

Accumulating evidence suggests important roles for the receptor tyrosine kinase Axl in cancer progression, invasion, metastasis, drug resistance, and patient mortality, highlighting Axl as an attractive target for therapeutic development. We have generated and characterized a potent and selective small-molecule inhibitor, R428, that blocks the catalytic and procancerous activities of Axl. R428 inhibits Axl with low nanomolar activity and blocked Axl-dependent events, including Akt phosphorylation, breast cancer cell invasion, and proinflammatory cytokine production. Pharmacologic investigations revealed favorable exposure after oral administration such that R428-treated tumors displayed a dose-dependent reduction in expression of the cytokine granulocyte macrophage colony-stimulating factor and the epithelial-mesenchymal transition transcriptional regulator Snail. In support of an earlier study, R428 inhibited angiogenesis in corneal micropocket and tumor models. R428 administration reduced metastatic burden and extended survival in MDA-MB-231 intracardiac and 4T1 orthotopic (median survival, >80 days compared with 52 days; P < 0.05) mouse models of breast cancer metastasis. Additionally, R428 synergized with cisplatin to enhance suppression of liver micrometastasis. Our results show that Axl signaling regulates breast cancer metastasis at multiple levels in tumor cells and tumor stromal cells and that selective Axl blockade confers therapeutic value in prolonging survival of animals bearing metastatic tumors.


Subject(s)
Benzocycloheptenes/pharmacology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carcinoma/mortality , Carcinoma/pathology , Oncogene Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Triazoles/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Benzocycloheptenes/therapeutic use , Breast Neoplasms/drug therapy , Carcinoma/drug therapy , Female , HeLa Cells , Humans , K562 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins , Survival Analysis , Triazoles/therapeutic use , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Axl Receptor Tyrosine Kinase
11.
Cancer Res ; 64(14): 4931-41, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15256466

ABSTRACT

Aberrant epidermal growth factor receptor (EGFR) and ErbB2 expression are associated with advanced disease and poor patient prognosis in many tumor types (breast, lung, ovarian, prostate, glioma, gastric, and squamous carcinoma of head and neck). In addition, a constitutively active EGFR type III deletion mutant has been identified in non-small cell lung cancer, glioblastomas, and breast tumors. Hence, members of the EGFR family are viewed as promising therapeutic targets in the fight against cancer. In a similar vein, vascular endothelial growth factor (VEGF) receptor kinases are also promising targets in terms of an antiangiogenic treatment strategy. AEE788, obtained by optimization of the 7H-pyrrolo[2,3-d]pyrimidine lead scaffold, is a potent combined inhibitor of both epidermal growth factor (EGF) and VEGF receptor tyrosine kinase family members on the isolated enzyme level and in cellular systems. At the enzyme level, AEE788 inhibited EGFR and VEGF receptor tyrosine kinases in the nm range (IC(50)s: EGFR 2 nm, ErbB2 6 nm, KDR 77 nm, and Flt-1 59 nm). In cells, growth factor-induced EGFR and ErbB2 phosphorylation was also efficiently inhibited (IC(50)s: 11 and 220 nm, respectively). AEE788 demonstrated antiproliferative activity against a range of EGFR and ErbB2-overexpressing cell lines (including EGFRvIII-dependent lines) and inhibited the proliferation of epidermal growth factor- and VEGF-stimulated human umbilical vein endothelial cells. These properties, combined with a favorable pharmacokinetic profile, were associated with a potent antitumor activity in a number of animal models of cancer, including tumors that overexpress EGFR and or ErbB2. Oral administration of AEE788 to tumor-bearing mice resulted in high and persistent compound levels in tumor tissue. Moreover, AEE788 efficiently inhibited growth factor-induced EGFR and ErbB2 phosphorylation in tumors for >72 h, a phenomenon correlating with the antitumor efficacy of intermittent treatment schedules. Strikingly, AEE788 also inhibited VEGF-induced angiogenesis in a murine implant model. Antiangiogenic activity was also apparent by measurement of tumor vascular permeability and interstitial leakage space using dynamic contrast enhanced magnetic resonance imaging methodology. Taken together, these data indicate that AEE788 has potential as an anticancer agent targeting deregulated tumor cell proliferation as well as angiogenic parameters. Consequently, AEE788 is currently in Phase I clinical trials in oncology.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Purines/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Animals , BALB 3T3 Cells , Cell Division/drug effects , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , ErbB Receptors/metabolism , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Melanoma, Experimental/drug therapy , Melanoma, Experimental/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Phosphorylation , Purines/pharmacokinetics , Receptor, ErbB-2/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Xenograft Model Antitumor Assays
12.
Exp Toxicol Pathol ; 55(4): 237-45, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14703768

ABSTRACT

UNLABELLED: The aim of the study was to determine the effects of a specific epithelial growth factor Receptor kinase inhibitor (EGFR-KI) and Taxol on tumor growth in a novel tumor model. MATERIAL & METHODS: A genetically engineered tumor model which uses "transgenic" organs in immune competent mice was used. NeuT-transfected immortalized HC11 epithelial cells and primary mouse mammary epithelial cells have been transplanted into the gland-free mammary fat pad of female BALB/c mice. Mammary tumors developed after a latency period of three to four weeks. The mice were thereafter daily orally treated over a 19 or 22-day period with 0, 38, 75, 2 x 75 mg/kg body weight (b.w.) EGFR-KI (n: 7-9 per group) or intravenously with 10 mg/kg b.w. Taxol. After necropsy the histopathological evaluation of the tumors was performed in a coded manner. The proliferation activity of tumor cells was analyzed by laser scanning cytometry (LSC) using anti-Ki67-antibodies. RESULTS: Oral Treatment with EGFR-KI in this transgenic organ model showed clear antitumor efficacy in a dose-dependent manner in the range between 38 and 75 mg/kg b.w. This antiproliferative effect appears to be minimally increased at 75 mg/kg/day twice per day. For all treatments a strong correlation between the biological behavior of the tumor, histopathology and cell proliferation could be established. In contrast, treatment with Taxol showed no significant reduction of tumor growth or cell proliferation in this model. This new transgenic organ model comprising histopathological evaluation and cell proliferation analysis appears to be a suitable test system for drug candidates that affect specific biochemical pathways. It may have greater predictive nature for clinical effects in humans as compared to conventional tumor models because of its c-erb B2 gene overexpression.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , ErbB Receptors/antagonists & inhibitors , Mammary Glands, Animal/transplantation , Mammary Neoplasms, Experimental/drug therapy , Protein-Tyrosine Kinases/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Transformed , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor/methods , Enzyme Inhibitors/administration & dosage , Female , Image Cytometry , Injections, Intravenous , Ki-67 Antigen/metabolism , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Paclitaxel/administration & dosage , Paclitaxel/therapeutic use , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...