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











Publication year range
1.
Clin Oncol (R Coll Radiol) ; 34(7): 459-468, 2022 07.
Article in English | MEDLINE | ID: mdl-35012901

ABSTRACT

AIMS: To evaluate the efficacy and safety of nintedanib plus docetaxel in patients with advanced adenocarcinoma non-small cell lung cancer (NSCLC) who progressed after chemotherapy and immune checkpoint inhibitor (ICI) therapy. MATERIALS AND METHODS: VARGADO (NCT02392455) is an ongoing, prospective, non-interventional, real-world study of nintedanib plus docetaxel after first-line chemotherapy in the routine clinical treatment of patients with locally advanced, metastatic or locally recurrent adenocarcinoma NSCLC. Data were collected during routine visits. We report the results from cohort B (n = 80), who received third-line nintedanib plus docetaxel after first-line chemotherapy and second-line ICI therapy. RESULTS: The median duration of follow-up was 12.4 months. Median progression-free survival from initiation of third-line nintedanib plus docetaxel was 6.4 months (95% confidence interval 4.8, 7.3); median overall survival was 12.1 months (95% confidence interval 9.4, 13.5). The 1-year overall survival rate after initiation of third-line nintedanib plus docetaxel treatment (primary end point) was 52% (95% confidence interval 38.0%, 64.4%). Among 64 patients with a documented response, the objective response rate was 50% (n = 32; one complete response and 31 partial responses) and the disease control rate was 86% (n = 55). There were no new safety signals or unexpected toxicities. Among all treated patients, 74% (n = 59) experienced drug-related adverse events, most commonly (nintedanib-related/docetaxel-related) diarrhoea (34%/24%), a decreased white blood cell count (11%/19%) and nausea (13%/16%). CONCLUSIONS: Nintedanib plus docetaxel demonstrated a high response rate and disease stabilisation in the third-line setting after failure of prior chemotherapy and ICI treatment, with a manageable safety profile. These results suggest that nintedanib plus docetaxel represents an efficient treatment option after failure of prior ICIs. The ongoing VARGADO study provides valuable real-world data to inform clinical decision-making regarding treatment sequencing after chemotherapy and ICI failure in patients with adenocarcinoma NSCLC.


Subject(s)
Adenocarcinoma , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Adenocarcinoma/pathology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma, Non-Small-Cell Lung/pathology , Docetaxel , Humans , Immune Checkpoint Inhibitors , Indoles , Lung Neoplasms/pathology , Neoplasm Recurrence, Local/drug therapy , Prospective Studies , Treatment Outcome
2.
Clin. transl. oncol. (Print) ; 14(5): 376-381, mayo 2012. ilus, tab
Article in English | IBECS | ID: ibc-126912

ABSTRACT

INTRODUCTION: In patients, a transient decrease in peripheral blood lymphocyte counts was observed following intraperitoneal administration of the trifunctional monoclonal antibody catumaxomab (anti-human EpCAM x anti-human CD3). The aim of this study was to clarify the observed effect in a preclinical mouse model and to analyse the related mechanism of action in vitro. MATERIALS AND METHODS: A related antibody, BiLu (antihuman EpCAM x anti-mouse CD3), was administered to mice and blood leukocytes were analysed. In vitro studies measured activation and cytokine secretion from human peripheral blood mononuclear cells (PBMC). For the analysis of T cell adhesion, PBMC were preincubated with catumaxomab and then co-cultured with human endothelial cells (HUVEC); T cell adhesion was assessed in the presence or absence of endothelial cell preactivation by TNFα. Adherent T cells were determined by flow cytometry. RESULTS: Treatment of mice with BiLu resulted in a dosedependent transient decrease in CD3+ T cells (both CD4+ and CD8+) that returned to the normal range within 48 h. Catumaxomab physiologically activated T cells in vitro (increased CD69 expression) and induced cytokine release (TNFα, IFNγ). TNFα increased expression of adhesion molecules CD54 and CD62E on endothelial cells. Furthermore, catumaxomab dose-dependently enhanced adhesion of T cells to endothelial cells. Adhesion was further increased when endothelial cells were preactivated with TNFα. CONCLUSIONS: Catumaxomab increases adhesion of T cells to endothelial cells due to antibody-mediated activation of T cells and production of T cell cytokines that up-regulate endothelial cell adhesion molecules. These results provide a mechanistic rationale for the transient, reversible decrease in lymphocyte counts observed following catumaxomab administration in patients, which is likely to be due to redistribution of lymphocytes (AU)


Subject(s)
Animals , Tumor Necrosis Factor Receptor Superfamily, Member 7/administration & dosage , Tumor Necrosis Factor Receptor Superfamily, Member 7/analysis , Tumor Necrosis Factor Receptor Superfamily, Member 7/blood , Tumor Necrosis Factor Receptor Superfamily, Member 7/chemical synthesis , Tumor Necrosis Factor Receptor Superfamily, Member 7/classification , Tumor Necrosis Factor Receptor Superfamily, Member 7/deficiency , Tumor Necrosis Factor Receptor Superfamily, Member 7 , Tumor Necrosis Factor Receptor Superfamily, Member 7/toxicity
3.
Clin Transl Oncol ; 13(12): 889-98, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22126733

ABSTRACT

BACKGROUND Trifunctional antibodies, such as catumaxomab (anti-EpCAM×anti-CD3) and ertumaxomab (anti- HER-2/neu×anti-CD3), transiently link immune effector cells to tumour cells, which results in cellular cytotoxicity towards the tumour cells. A functional immune system is therefore essential for effective anti-tumour activity. However, the commonly observed haematotoxicity of chemotherapeutics and radiation therapy may be associated with some degree of immunosuppression. Combining chemotherapy and trifunctional antibodies in cancer treatment requires understanding of the impact of chemotherapeutics on immune cell function and, thus, on the activity of trifunctional antibodies. METHODS The effect of chemotherapeutic treatment on trifunctional antibody-mediated anti-tumour activity was assessed in vitro. Blood samples were collected from 12 head and neck squamous cell carcinoma patients after chemotherapy (5-fluorouracil, cisplatin) and radiotherapy, and from one healthy control donor. The immune cell status was analysed and mononuclear cells (MNC) were isolated. The potency of catumaxomab and ertumaxomab was assessed in a cytotoxicity assay using MNC isolated from each patient sample in co-culture with a tumour target cell line. The release of infl ammatory cytokines was also monitored in the cell culture supernatant. RESULTS Most patients included in this study had decreased immune cell counts during the course of chemotherapy. Nonetheless, an effective and concentration-dependent anti- tumour activity mediated by trifunctional antibodies was demonstrated using these patient immune effector cells. The immune response activity of the patient immune cells was not impaired one week after cisplatin administration or even three days after the last 5-fluorouracil treatment. CONCLUSION This study shows for the first time that immune effector cells from cancer patients undergoing standard chemotherapy and radiotherapy can be activated by trifunctional antibodies for efficient killing of tumour cells.


Subject(s)
Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/therapy , Chemoradiotherapy , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/therapy , Aged , Case-Control Studies , Cisplatin/administration & dosage , Cytokines/metabolism , Female , Fluorouracil/administration & dosage , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Staging , Survival Rate , Treatment Outcome , Tumor Cells, Cultured
4.
Clin. transl. oncol. (Print) ; 13(12): 889-898, dic. 2011. ilus
Article in English | IBECS | ID: ibc-125998

ABSTRACT

BACKGROUND Trifunctional antibodies, such as catumaxomab (anti-EpCAM×anti-CD3) and ertumaxomab (anti- HER-2/neu×anti-CD3), transiently link immune effector cells to tumour cells, which results in cellular cytotoxicity towards the tumour cells. A functional immune system is therefore essential for effective anti-tumour activity. However, the commonly observed haematotoxicity of chemotherapeutics and radiation therapy may be associated with some degree of immunosuppression. Combining chemotherapy and trifunctional antibodies in cancer treatment requires understanding of the impact of chemotherapeutics on immune cell function and, thus, on the activity of trifunctional antibodies. METHODS The effect of chemotherapeutic treatment on trifunctional antibody-mediated anti-tumour activity was assessed in vitro. Blood samples were collected from 12 head and neck squamous cell carcinoma patients after chemotherapy (5-fluorouracil, cisplatin) and radiotherapy, and from one healthy control donor. The immune cell status was analysed and mononuclear cells (MNC) were isolated. The potency of catumaxomab and ertumaxomab was assessed in a cytotoxicity assay using MNC isolated from each patient sample in co-culture with a tumour target cell line. The release of infl ammatory cytokines was also monitored in the cell culture supernatant. RESULTS Most patients included in this study had decreased immune cell counts during the course of chemotherapy. Nonetheless, an effective and concentration-dependent anti- tumour activity mediated by trifunctional antibodies was demonstrated using these patient immune effector cells. The immune response activity of the patient immune cells was not impaired one week after cisplatin administration or even three days after the last 5-fluorouracil treatment. CONCLUSION This study shows for the first time that immune effector cells from cancer patients undergoing standard chemotherapy and radiotherapy can be activated by trifunctional antibodies for efficient killing of tumour cells (AU)


Subject(s)
Male , Female , Middle Aged , Aged , Humans , Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/therapy , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/therapy , Chemoradiotherapy/methods , Chemoradiotherapy , Case-Control Studies , Cisplatin/administration & dosage , Fluorouracil/administration & dosage , Follow-Up Studies , Neoplasm Staging , Survival Rate , Treatment Outcome , Tumor Cells, Cultured , Cytokines/metabolism
5.
Br J Cancer ; 97(3): 315-21, 2007 Aug 06.
Article in English | MEDLINE | ID: mdl-17622246

ABSTRACT

Epithelial cell adhesion molecule EpCAM is a transmembrane glycoprotein that is frequently overexpressed in a variety of carcinomas. This pan-carcinoma antigen has served as the target for a plethora of immunotherapies. Innovative therapeutic approaches include the use of trifunctional antibodies (trAbs) that recruit and activate different types of immune effector cells at the tumour site. The trAb catumaxomab has dual specificity for EpCAM and CD3. In patients with malignant ascites, catumaxomab significantly increased the paracentesis-free interval, corroborating the high efficacy of this therapeutic antibody. Here, we characterised the monoclonal antibody (mAb) HO-3, that is, the EpCAM-binding arm of catumaxomab. Peptide mapping indicated that HO-3 recognises a discontinuous epitope, having three binding sites in the extracellular region of EpCAM. Studies with glycosylation-deficient mutants showed that mAb HO-3 recognised EpCAM independently of its glycosylation status. High-affinity binding was not only detected for mAb HO-3, but also for the monovalent EpCAM-binding arm of catumaxomab with an excellent K(D) of 5.6 x 10(-10) M. Furthermore, trAb catumaxomab was at least a 1000-fold more effective in eliciting the eradication of tumour cells by effector peripheral blood mononuclear cells compared with mAb HO-3. These findings suggest the great therapeutic potential of trAbs and clearly speak in favour of EpCAM-directed cancer immunotherapies.


Subject(s)
Antigens, Neoplasm/therapeutic use , Cell Adhesion Molecules/therapeutic use , Immunotherapy , Neoplasms/therapy , Amino Acid Sequence , Antibodies, Bispecific/metabolism , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Base Sequence , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , DNA Primers , Epidermal Growth Factor/chemistry , Epidermal Growth Factor/metabolism , Epithelial Cell Adhesion Molecule , Epitopes/chemistry , Glycosylation , Humans , Molecular Sequence Data
6.
Mar Biotechnol (NY) ; 3(Supplement 1): S3-5, 2001 Jun.
Article in English | MEDLINE | ID: mdl-14961294
7.
J Cell Biochem ; 76(4): 572-84, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10653977

ABSTRACT

Mutations that affect the oligomerization domain (OD) of the p53 tumor suppressor may be of particular interest because of the remarkable contradiction between the conservation of the OD and its relative functional resistance to amino acid substitutions, and because of recent hints that cellular protein factors may interact with the OD. Both point to the possibility that this domain fulfills tasks beyond oligomerization. We report that the tumor-associated mutants 330H, 334V, and 337C are defective for homo-oligomerization by three criteria. Accordingly, 330H and 337C failed to bind to a p53 recognition motif in gel-shift assays and to stimulate reporter genes efficiently in transient transfections. 334V retained some activity in both assays despite being oligomerization-defective. The ability of the mutants to induce apoptosis correlated with their performance in the DNA binding and transactivation assays. However, mutants 330H and 337C were able to provoke cell death when overexpressed, which in combination with their failure to transactivate genes suggests competence for the induction of transactivation-independent apoptosis at high protein levels. Although 334V and 337C failed to homo-oligomerize, they were able to hetero-oligomerize with a p53 with wild-type OD, and 334V was able to interfere with transactivation by wt p53. All mutants showed a reduced reactivity with antibody PAb421 and a distinct calpain cleavage pattern indicative of conformational alterations. In conclusion, tumor-associated OD mutants of p53 can be functionally competent to different degrees despite of being oligomerization defective.


Subject(s)
Protein Conformation , Tumor Suppressor Protein p53/chemistry , Antibodies/metabolism , Apoptosis/genetics , Calpain/metabolism , Centrifugation, Density Gradient , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Epitopes/genetics , Epitopes/immunology , Genes, Reporter/genetics , Humans , Mutation , Repressor Proteins/genetics , Transcriptional Activation/genetics , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
8.
J Biol Chem ; 274(27): 19136-44, 1999 Jul 02.
Article in English | MEDLINE | ID: mdl-10383418

ABSTRACT

The Epstein-Barr virus-encoded nuclear antigens EBNA2 and EBNA3C both interact with the cellular transcription factor RBP-Jkappa and modulate the expression of several shared target genes, suggesting a tight cooperation in latently infected cells. In a survey for additional cellular factors that bind to EBNA2 as well as EBNA3C, we have isolated and characterized DP103, a novel human member of the DEAD box family of putative ATP-dependent RNA helicases. The interaction with DP103 is mediated by amino acids (aa) 121-213 of EBNA2 and aa 534-778 of EBNA3C, regions that are not involved in binding of the viral proteins to RBP-Jkappa. The DP103-cDNA encodes a protein of 824 aa that harbors all of the common DEAD box motifs. Monoclonal antibodies raised against DP103 detect a protein of 103 kDa in mammalian cells that resides in high molecular weight complexes in vivo. We have detected an ATPase activity intrinsic to or closely associated with DP103. By subcellular fractionation, we find DP103 in both a soluble nuclear fraction as well as in the insoluble skeletal fraction. Whereas the protein and its mRNA are uniformly expressed in all tested cell lines, we observed differential expression of the mRNA in normal human tissues.


Subject(s)
Epstein-Barr Virus Nuclear Antigens/metabolism , RNA Helicases/chemistry , RNA Helicases/metabolism , Adenosine Triphosphatases/metabolism , Amino Acid Sequence , Animals , B-Lymphocytes/metabolism , Base Sequence , Cell Line , Cloning, Molecular , DEAD Box Protein 20 , DEAD-box RNA Helicases , DNA, Complementary/isolation & purification , Humans , Molecular Sequence Data , RNA, Messenger/metabolism , Random Amplified Polymorphic DNA Technique , Rats , Spodoptera , T-Lymphocytes/metabolism
9.
Int J Cancer ; 71(5): 858-66, 1997 May 29.
Article in English | MEDLINE | ID: mdl-9180157

ABSTRACT

p53-mediated programmed cell death (PMCD) often requires an intact transactivation domain of the p53 tumor suppressor and is therefore usually interpreted to rely upon the transactivation of genes. As a notable exception, murine GHFT1 cells have been documented to perish in a p53-dependent manner even in the presence of transcription inhibitor actinomycin D (Act D) and have since served as one model system for transactivation-independent apoptosis. We report here that p53 transactivation domain mutant Q22,S23 nonetheless fails to mediate apoptosis in these cells as efficiently as wild-type p53. This suggests that some function of the NH2-terminal domain other than the transactivation of genes supports PMCD of GHFT1 cells. To substantiate this suggestion, we employed a p53 whose transactivation domain had been replaced with the one of VP16, which acts through the same elements of the basal transcription machinery. Although the hybrid was fully competent for transactivation, it was impaired for the mediation of apoptosis to the same extent as mutant Q22,S23. Thus, a function of the transactivation domain other than the binding to the transcription co-activators hTAF(II)31 and 70 is required for the efficient induction of apoptosis in GHFT1 cells.


Subject(s)
Apoptosis , Peptide Fragments/physiology , Transcriptional Activation , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/physiology , Animals , Blotting, Western , DNA/metabolism , DNA Fragmentation , Mice , Mice, Transgenic , Mutation , Promoter Regions, Genetic , Transcription Factors , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
10.
Am J Vet Res ; 51(11): 1806-14, 1990 Nov.
Article in English | MEDLINE | ID: mdl-2173450

ABSTRACT

Our objectives were to describe the ultrastructural morphogenesis of pulmonary lesions induced by 3-methylindole in 30- to 45-day-old Holstein calves and to determine whether toxic exposure to 3-methylindole exacerbates pulmonary lesions induced by bovine respiratory syncytial virus. Administration of 3-methylindole (0.25 g/kg) to calves resulted in interstitial edema and ultrastructural swelling of type-I alveolar epithelial cells and nonciliated bronchiolar epithelial cells as early as 4 to 6 hours after intraruminal administration. More severe alveolar edema containing protein was associated with swelling of capillary endothelial cells at 2 days after administration. Proliferation of type-II alveolar epithelial cells was first observed at 2 days after 3-methylindole administration, and marked hyperplasia of type-II epithelial cells and nonciliated bronchiolar epithelial cells was evident by 4 days after administration. Pulmonary cytochrome P-450 monooxygenase concentrations decreased significantly (P less than 0.001) by 12 hours after administration and did not increase significantly again by 8 days after administration. Calves were inoculated with bovine respiratory syncytial virus 3 days after administration of 3-methylindole, and pulmonary lesions were assessed 5 days after viral inoculation. Viral replication was demonstrated by fluorescence microscopy for viral antigen or by transmission electron microscopy in ciliated and nonciliated airway epithelial cells. Viral antigen was identified infrequently in alveolar macrophages and in type-II alveolar epithelial cells. 3-Methylindole exposure in calves did not result in more widespread distribution of viral antigen in alveolar tissue of respiratory syncytial virus-inoculated calves or in significant enhancement of viral pneumonia.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Cattle Diseases/chemically induced , Lung Diseases/veterinary , Pneumonia, Viral/veterinary , Skatole/toxicity , Animals , Cattle , Cattle Diseases/pathology , Disease Susceptibility , Lung Diseases/chemically induced , Lung Diseases/pathology , Male , Microscopy, Electron/veterinary , Pneumonia, Viral/etiology , Pneumonia, Viral/pathology , Skatole/administration & dosage , Spumavirus/pathogenicity , Spumavirus/ultrastructure
11.
Cornell Vet ; 75(4): 473-83, 1985 Oct.
Article in English | MEDLINE | ID: mdl-4053607

ABSTRACT

Severe respiratory disease associated with bovine respiratory syncytial virus (BRSV) infection has been identified in dairy cattle in New York State. The cases identified occurred in dairy calves and heifers. The disease was characterized in 4 animals by pathologic changes including interstitial pneumonia, necrotizing bronchiolitis with multinucleated syncytial epithelial cells and interstitial emphysema. BRSV antigen was demonstrated in lung samples or was isolated in tissue culture in all 4 cases. A retrospective survey of 6279 bovine diagnostic accessions between 1977 and 1982 revealed 66 cases of interstitial pneumonia, often with concurrent bronchiolitis. In this 5 year period, only 1 case in 1981 had interstitial pneumonia and bronchiolitis with pathologic features consistent with BRSV infection. It is concluded that pathogenic BRSV has entered New York State and that it is contributing to clinical respiratory disease in dairy cattle.


Subject(s)
Bronchiolitis, Viral/veterinary , Cattle Diseases/complications , Pulmonary Fibrosis/veterinary , Respiratory Tract Diseases/veterinary , Respirovirus Infections/veterinary , Animals , Antigens, Viral/analysis , Bronchiolitis, Viral/complications , Bronchiolitis, Viral/pathology , Cattle , Cattle Diseases/microbiology , Cattle Diseases/pathology , Female , Lung/microbiology , Microscopy, Fluorescence , New York , Pulmonary Fibrosis/complications , Pulmonary Fibrosis/pathology , Respiratory Syncytial Viruses , Respiratory Tract Diseases/complications , Respiratory Tract Diseases/microbiology , Respiratory Tract Diseases/pathology , Respirovirus Infections/complications , Respirovirus Infections/microbiology , Respirovirus Infections/pathology , Retrospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL