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










Publication year range
1.
J Clin Med ; 11(1)2022 Jan 05.
Article in English | MEDLINE | ID: mdl-35012014

ABSTRACT

Immune cell reconstitution after stem cell transplantation is allocated over several stages. Whereas cells mediating innate immunity recover rapidly, adaptive immune cells, including T and B cells, recover slowly over several months. In this study we investigated kinetics and reconstitution of de novo B cell formation in patients receiving CD3 and CD19 depleted haploidentical stem cell transplantation with additional in vivo T cell depletion with monoclonal anti-CD3 antibody. This model enables a detailed in vivo evaluation of hierarchy and attribution of defined lymphocyte populations without skewing by mTOR- or NFAT-inhibitors. As expected CD3+ T cells and their subsets had delayed reconstitution (<100 cells/µL at day +90). Well defined CD19+ B lymphocytes of naïve and memory phenotype were detected at day +60. Remarkably, we observed a very early reconstitution of antibody-secreting cells (ASC) at day +14. These ASC carried the HLA-haplotype of the donor and secreted the isotypes IgM and IgA more prevalent than IgG. They correlated with a population of CD19- CD27- CD38low/+ CD138- cells. Of note, reconstitution of this ASC occurred without detectable circulating T cells and before increase of BAFF or other B cell stimulating factors. In summary, we describe a rapid reconstitution of peripheral blood ASC after CD3 and CD19 depleted haploidentical stem cell transplantation, far preceding detection of naïve and memory type B cells. Incidence before T cell reconstitution and spontaneous secretion of immunoglobulins allocate these early ASC to innate immunity, eventually maintaining natural antibody levels.

2.
Nat Commun ; 12(1): 7005, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34853305

ABSTRACT

Immune-checkpoint inhibitors (ICI) have transformed oncological therapy. Up to 20% of all non-small cell lung cancers (NSCLCs) show durable responses upon treatment with ICI, however, robust markers to predict therapy response are missing. Here we show that blood platelets interact with lung cancer cells and that PD-L1 protein is transferred from tumor cells to platelets in a fibronectin 1, integrin α5ß1 and GPIbα-dependent manner. Platelets from NSCLC patients are found to express PD-L1 and platelet PD-L1 possess the ability to inhibit CD4 and CD8 T-cells. An algorithm is developed to calculate the activation independent adjusted PD-L1 payload of platelets (pPD-L1Adj.), which is found to be superior in predicting the response towards ICI as compared to standard histological PD-L1 quantification on tumor biopsies. Our data suggest that platelet PD-L1 reflects the collective tumor PD-L1 expression, plays important roles in tumor immune evasion and overcomes limitations of histological quantification of often heterogeneous intratumoral PD-L1 expression.


Subject(s)
B7-H1 Antigen/metabolism , Blood Platelets/metabolism , Carcinoma, Non-Small-Cell Lung/therapy , Immunotherapy , Lung Neoplasms/therapy , Adult , Aged , Aged, 80 and over , B7-H1 Antigen/genetics , Biomarkers, Tumor , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Immunologic Factors , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Male , Middle Aged , T-Lymphocytes , Young Adult
3.
Front Immunol ; 12: 653081, 2021.
Article in English | MEDLINE | ID: mdl-33936075

ABSTRACT

Soft tissue sarcoma (STS) constitutes a rare group of heterogeneous malignancies. Effective treatment options for most subtypes of STS are still limited. As a result, especially in metastatic disease, prognosis is still dismal. The ligands for the activating immunoreceptor NKG2D (NKG2DL) are commonly expressed in STS, but generally absent in healthy tissues. This provides the rationale for utilization of NKG2DL as targets for immunotherapeutic approaches. We here report on the preclinical characterization of bispecific fusion proteins (BFP) consisting of the extracellular domain of the NKG2D receptor fused to Fab-fragments directed against CD3 (NKG2D-CD3) or CD16 (NKG2D-CD16) for treatment of STS. After characterization of NKG2DL expression patterns on various STS cell lines, we demonstrated that both NKG2D-CD16 and NKG2D-CD3 induce profound T and NK cell reactivity as revealed by analysis of activation, degranulation and secretion of IFNγ as well as granule associated proteins, resulting in potent target cell lysis. In addition, the stimulatory capacity of the constructs to induce T and NK cell activation was analyzed in heavily pretreated STS patients and found to be comparable to healthy donors. Our results emphasize the potential of NKG2D-CD3 and NKG2D-CD16 BFP to target STS even in an advanced disease.


Subject(s)
CD3 Complex/antagonists & inhibitors , NK Cell Lectin-Like Receptor Subfamily K/genetics , Receptors, IgG/antagonists & inhibitors , Recombinant Fusion Proteins/therapeutic use , Sarcoma/drug therapy , Adult , Aged , Antibodies, Bispecific/genetics , Antibodies, Bispecific/pharmacology , CD3 Complex/metabolism , Cell Degranulation/drug effects , Cell Degranulation/immunology , Cell Line, Tumor , Female , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation/drug effects , Male , Middle Aged , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Primary Cell Culture , Protein Domains/genetics , Receptors, IgG/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/pharmacology , Sarcoma/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Young Adult
4.
Genes (Basel) ; 13(1)2021 12 23.
Article in English | MEDLINE | ID: mdl-35052377

ABSTRACT

Mutations of the IL2RG gene, which encodes for the interleukin-2 receptor common gamma chain (γC, CD132), can lead to X-linked severe combined immunodeficiency (X-SCID) associated with a T-B+NK- phenotype as a result of dysfunctional γC-JAK3-STAT5 signaling. Lately, hypomorphic mutations of the IL2RG gene have been described causing atypical SCID with a milder phenotype. Here, we report three brothers with low-normal lymphocyte counts and susceptibility to recurrent respiratory infections and cutaneous warts. The clinical presentation combined with dysgammaglobulinemia suspected an inherited immunity disorder, which has been proven by Next Generation Sequencing as a novel c.458T > C; p.Ile153Thr IL2RG missense-mutation. Subsequent functional characterization revealed impaired T-cell proliferation, low TREC levels and a skewed TCR Vß repertoire in all three patients. Interestingly, investigation of various subpopulations showed normal expression of CD132 but with partially impaired STAT5 phosphorylation compared to healthy controls. Additionally, we performed precise genetic analysis of subpopulations revealing spontaneous somatic reversion, predominately in lymphoid derived CD3+, CD4+ and CD8+ T cells. Our data demonstrate that the atypical SCID phenotype noticed in these three brothers is due to the combination of hypomorphic IL-2RG function and somatic reversion.


Subject(s)
Interleukin Receptor Common gamma Subunit/genetics , Mutation/genetics , X-Linked Combined Immunodeficiency Diseases/genetics , Adult , CD8-Positive T-Lymphocytes , Cell Proliferation/genetics , Humans , Male , Phenotype , Young Adult
5.
Cells ; 9(12)2020 12 11.
Article in English | MEDLINE | ID: mdl-33322555

ABSTRACT

Sclerosing spindle cell rhabdomyosarcoma (SSRMS) is a rare rhabdomyosarcomas (RMS) subtype. Especially cases bearing a myogenic differentiation 1 (MYOD1) mutation are characterized by a high recurrence and metastasis rate, often leading to a fatal outcome. SSRMS cell lines are valuable in vitro models for studying disease mechanisms and for the preclinical evaluation of new therapeutic approaches. In this study, a cell line established from a primary SSRMS tumor of a 24-year-old female after multimodal chemotherapeutic pretreatment has been characterized in detail, including immunohistochemistry, growth characteristics, cytogenetic analysis, mutation analysis, evaluation of stem cell marker expression, differentiation potential, and tumorigenicity in mice. The cell line which was designated SRH exhibited a complex genomic profile, including several translocations and deletions. Array-comparative genomic hybridization (CGH) revealed an overall predominating loss of gene loci. The mesenchymal tumor origin was underlined by the expression of mesenchymal markers and potential to undergo adipogenic and osteogenic differentiation. Despite myogenic marker expression, terminal myogenic differentiation was inhibited, which might be elicited by the MYOD1 hotspot mutation. In vivo tumorigenicity could be confirmed after subcutaneous injection into NOD/SCID/γcnull mice. Summarized, the SRH cell line is the first adult SSRMS cell line available for preclinical research on this rare RMS subtype.


Subject(s)
Genomics , Rhabdomyosarcoma/pathology , Adipogenesis , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Line Authentication/methods , Comparative Genomic Hybridization , Female , Humans , Karyotyping , Mice , Mice, Inbred NOD , Mice, SCID , MyoD Protein/genetics , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/genetics , Signal Transduction , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Young Adult
6.
Cell Death Dis ; 11(8): 701, 2020 08 24.
Article in English | MEDLINE | ID: mdl-32839432

ABSTRACT

Soft tissue sarcomas (STS) are a heterogeneous group of malignancies predominantly affecting children and young adults. Despite improvements in multimodal therapies, 5-year survival rates are only 50% and new treatment options in STS are urgently needed. To develop a rational combination therapy for the treatment of STS we focused on ABT-199 (Venetoclax), a BCL-2 specific BH3-mimetic, in combination with the proteasome inhibitor bortezomib (BZB). Simultaneous inhibition of BCL-2 and the proteasome resulted in strongly synergistic apoptosis induction. Mechanistically, ABT-199 mainly affected the multidomain effector BAX by liberating it from BCL-2 inhibition. The combination with BZB additionally resulted in the accumulation of BOK, a BAX/BAK homologue, and of the BH3-only protein NOXA, which inhibits the anti-apoptotic protein MCL-1. Thus, the combination of ABT-199 and BZB sensitizes STS cells to apoptosis by simultaneously releasing several defined apoptotic restraints. This synergistic mechanism of action was verified by CRISPR/Cas9 knock-out, showing that both BAX and NOXA are crucial for ABT-199/BZB-induced apoptosis. Noteworthy, efficient induction of apoptosis by ABT-199/BZB was not affected by the p53 status and invariably detected in cell lines and patient-derived tumor cells of several sarcoma types, including rhabdomyo-, leiomyo-, lipo-, chondro-, osteo-, or synovial sarcomas. Hence, we propose the combination of ABT-199 and BZB as a promising strategy for the treatment of STS, which should warrant further clinical investigation.


Subject(s)
Bortezomib/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Sarcoma/drug therapy , Sulfonamides/pharmacology , Adult , Aged , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Biphenyl Compounds/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Cell Line, Tumor , Drug Synergism , Female , Humans , Male , Middle Aged , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Nitrophenols/pharmacology , Proteasome Endopeptidase Complex/drug effects , Proteasome Inhibitors/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Sarcoma/metabolism , Sulfonamides/metabolism , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
7.
BMC Res Notes ; 7: 313, 2014 May 23.
Article in English | MEDLINE | ID: mdl-24885681

ABSTRACT

BACKGROUND: While paraneoplastic leukocytosis is a common phenomenon in solid tumors, extreme elevations of white blood counts (WBC) in the range of more than 100,000/µl are uncommon in patients with non-hematologic malignancies. Leukocytosis with mature neutrophils due to a granulocyte colony-stimulating factor (G-CSF) producing tumor is only seen on rare occasions. CASE PRESENTATION: Massive neutrophil leukocytosis of approximately 100,000/µl was diagnosed in a 57-year-old Caucasian woman with metastatic undifferentiated endometrial sarcoma. A bone marrow trephine biopsy revealed massively increased granulopoiesis, but no evidence of monoclonal myeloproliferative disease. After the primary tumor had been resected, white blood count (WBC) plummeted and went back to nearly normal levels within one week. With progressive metastatic disease, granulocyte colony-stimulating factor (G-CSF) plasma levels were found to be increased by 10-fold. White blood count (WBC) strictly correlated with tumor burden and response to chemotherapy. In the final stage of treatment resistent disease, white blood count (WBC) approximated 300,000/µl. CONCLUSION: We report on a granulocyte colony-stimulating factor (G-CSF) secreting undifferentiated endometrial sarcoma, which was associated with extreme neutrophil counts. White blood count (WBC) were closely correlated with tumor burden and associated with an aggressive clinical course. We suggest that paraneoplastic neutrophilia represents a poor prognostic sign in soft tissue sarcoma. In patients with similar constellations, antitumor therapy must not be delayed.


Subject(s)
Bone Marrow Neoplasms/diagnosis , Granulocyte Colony-Stimulating Factor/metabolism , Paraneoplastic Syndromes/complications , Paraneoplastic Syndromes/diagnosis , Sarcoma/complications , Sarcoma/diagnosis , Sarcoma/metabolism , Blood Cell Count , Bone Marrow/pathology , Bone Marrow/surgery , Bone Marrow Neoplasms/blood , Bone Marrow Neoplasms/diagnostic imaging , Bone Marrow Neoplasms/surgery , Diagnosis, Differential , Fatal Outcome , Female , Humans , Middle Aged , Paraneoplastic Syndromes/blood , Paraneoplastic Syndromes/diagnostic imaging , Paraneoplastic Syndromes/surgery , Radiography, Thoracic , Sarcoma/surgery , Tomography, X-Ray Computed
8.
Stem Cells Transl Med ; 2(1): 53-60, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23283492

ABSTRACT

Advanced adult soft-tissue sarcomas (STSs) are rare tumors with a dismal prognosis and limited systemic treatment options. STSs may originate from mesenchymal stem cells (MSCs); the latter have mainly been isolated from adult bone marrow as plastic-adherent cells with differentiation capacity into mesenchymal tissues. Recently, a panel of antibodies has been established that allows for the prospective isolation of primary MSCs with high selectivity. Similar to cancer stem cells in other malignancies, sarcoma stem cells may bear immunophenotypic similarity with the corresponding precursor, that is, MSCs. We therefore set out to establish the expression pattern of MSC markers in sarcoma cell lines and primary tumor samples by flow cytometry. In addition, fibroblasts from different sources were examined. The results document a significant amount of MSC markers shared by sarcoma cells. The expression pattern includes uniformly expressed markers, as well as MSC markers that only stained subpopulations of sarcoma cells. Expression of W5C5, W8B2 (tissue nonspecific alkaline phosphatase [TNAP]), CD344 (frizzled-4), and CD271 marked subpopulations displaying increased proliferation potential. Moreover, CD271+ cells displayed in vitro doxorubicin resistance and an increased capacity to form spheres under serum-free conditions. Interestingly, another set of antigens, including the bona fide progenitor cell markers CD117 and CD133, were not expressed. Comparative expression patterns of novel MSC markers in sarcoma cells, as well as fibroblasts and MSCs, are presented. Our data suggest a hierarchical cytoarchitecture of the most common adult type sarcomas and introduce W5C5, TNAP, CD344, and CD271 as potential sarcoma progenitor cell markers.


Subject(s)
Antigens, Surface/metabolism , Biomarkers, Tumor/metabolism , Mesenchymal Stem Cells/metabolism , Neoplastic Stem Cells/metabolism , Sarcoma/metabolism , Antibiotics, Antineoplastic/pharmacology , Cell Differentiation , Cell Proliferation , Cell Shape , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Humans , Spheroids, Cellular/drug effects , Tumor Cells, Cultured
9.
Blood ; 121(5): 723-33, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23223360

ABSTRACT

UNLABELLED: Polymorphonuclear neutrophil granulocytes (neutrophils) are tightly controlled by an incompletely understood homeostatic feedback loop adjusting the marrow's supply to peripheral needs. Although it has long been known that marrow cellularity is inversely correlated with G-CSF levels, the mechanism linking peripheral clearance to production remains unknown. Herein, the feedback response to antibody induced neutropenia is characterized to consist of G-CSF­dependent shifts of marrow hematopoietic progenitor populations including expansion of the lin-/Sca-1/c-kit (LSK) and granulocyte macrophage progenitor (GMP) compartments at the expense of thrombopoietic and red cell precursors. Evidence is provided that positive feedback regulation is independent from commensal germs as well as T, B, and NK cells. However, in vivo feedback is impaired in TLR4-/- and TRIF-/-, but not MyD88-/- animals. In conclusion, steady-state neutrophil homeostasis is G-CSF­dependent and regulated through pattern-recognition receptors,thereby directly linking TLR-triggering to granulopoiesis. KEY POINTS: Steady-state and emergency granulopoiesis are both dependent on TLR signaling.


Subject(s)
Adaptor Proteins, Vesicular Transport/immunology , Granulocyte Precursor Cells/immunology , Homeostasis/immunology , Neutrophils/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Adaptor Proteins, Vesicular Transport/genetics , Animals , Granulocyte Colony-Stimulating Factor/genetics , Granulocyte Colony-Stimulating Factor/immunology , Granulocyte Precursor Cells/cytology , Homeostasis/genetics , Lymphocytes/immunology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Neutrophils/cytology , Signal Transduction/genetics , Toll-Like Receptor 4/genetics
10.
Mol Cancer Ther ; 7(7): 1940-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18645004

ABSTRACT

The biosynthesis of immunoglobulin leads to constitutive endoplasmic reticulum (ER) stress in myeloma cells, which activates the unfolded protein response (UPR). The UPR promotes protein folding by chaperones and increases proteasomal degradation of misfolded protein. Excessive ER stress induces apoptosis and represents a molecular basis for the bortezomib sensitivity of myeloma. Most solid malignancies such as sarcoma, by contrast, are poorly bortezomib sensitive and display low levels of ER stress. We hypothesized that pharmacologic induction of ER stress might sensitize malignancies to bortezomib treatment. We show that the HIV protease inhibitor ritonavir induces ER stress in bortezomib-resistant sarcoma cells. Ritonavir triggered the UPR, decreased the degradation of newly synthesized protein, but did not directly inhibit proteasomal active sites in the therapeutic dose range in contrast to bortezomib. Whereas neither bortezomib nor ritonavir monotherapy translated into significant apoptosis at therapeutic drug levels, the combination strongly increased the level of ER stress and activated PERK, IRE1, and ATF6, synergistically induced CHOP, JNK, caspase-4, and caspase-9, and resulted in >90% apoptosis. In summary, ritonavir increases the level of ER stress induced by bortezomib, which sensitizes bortezomib-resistant cells to bortezomib-induced apoptosis. Ritonavir may therefore be tested clinically to improve the sensitivity of solid malignancies toward bortezomib treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Boronic Acids/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/pathology , Pyrazines/pharmacology , Ritonavir/pharmacology , Sarcoma/pathology , Binding Sites , Bortezomib , Cell Death/drug effects , Cell Line, Tumor , Drug Screening Assays, Antitumor , G2 Phase/drug effects , Humans , Neoplasm Proteins/biosynthesis , Proteasome Inhibitors , Protein Biosynthesis/drug effects , Protein Folding , Protein Subunits/metabolism
11.
Int J Oncol ; 29(2): 471-9, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16820891

ABSTRACT

Testicular germ cell tumors (GCTs) are highly sensitive to cisplatin-based chemotherapy. It has been suggested that the chemosensitivity of GCTs can be partially attributed to the preference of apoptosis induction over a p21-mediated G1/S phase cell-cycle arrest following induction of p53. Since cell-cycle progression can be manipulated by a growing number of targeted agents, a thorough understanding of the impact of cell-cycle progression on drug-induced cell death might help to enhance the efficacy of chemotherapy. The aim of this study was to assess the cell-cycle dependence of cisplatin-induced cell death in an in vitro model of GCTs. Cell-cycle progression and induction of apoptosis were assessed by flow cytometry and Western blot analysis of PARP cleavage in the GCT derived cell lines, NT2 and 2102 EP, and compared with the breast carcinoma cell line MCF-7. Response to treatment was assessed in different phases of the cell cycle after synchronization by serum depletion and contact inhibition. Following cisplatin exposure, unsynchronized cells accumulated in G2/M after 28 h. This arrest was reversible at sublethal cisplatin doses (0.5-4.5 microM for 2 h). At higher concentrations, cells accumulated in G2 and died in G2/M-arrest. A 2-h exposure of cells in G2/M with 10 microM cisplatin resulted in a higher apoptotic index 70 h after treatment (74 and 70% for NT2 and 2102 EP, respectively) compared to treatment in G1/S (34 and 38%). Synchronized cells treated in G1 showed PARP cleavage after 48 h following cisplatin exposure, whereas treatment in G2 resulted in PARP cleavage already after 24 h. Cisplatin-induced cell death in GCTs is highly dependent on cell-cycle phase. All crucial events are restricted to the G2/M phase: cisplatin-induced DNA-damage is sensed, the apoptotic process is initiated and eventually executed in this phase of the cell cycle. The cells are most sensitive to cisplatin in this phase of the cell cycle. As far as the development of targeted agents is concerned, inhibition of the cell cycle in G1/S phase is likely to result in a protective effect against cisplatin, whereas agents arresting cells in G2/M may exert a synergistic effect.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Neoplasms, Germ Cell and Embryonal/drug therapy , Neoplasms, Germ Cell and Embryonal/pathology , Apoptosis , Cell Cycle , Cell Line, Tumor , DNA Damage , Drug Resistance, Neoplasm , Humans , In Vitro Techniques , Poly(ADP-ribose) Polymerases/metabolism
12.
Invest New Drugs ; 23(3): 205-11, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15868376

ABSTRACT

BACKGROUND: Germ cell tumours (GCTs) are highly sensitive to cisplatin-based chemotherapy. The inability to arrest the cell cycle at the G1/S-check-point due to a lack of retinoblastoma gene product RB has been suggested as one potential explanation for this feature. Flavopiridol (FP), an inhibitor of cyclin dependent kinases, causes cell cycle arrest or apoptosis depending on the relation of the transcription factor E2F1 and RB. METHODS: The effect of FP was evaluated in GCT-derived cell lines NT2, 2102 EP and NCCIT in comparison to cell lines derived from ovarian cancer (SKOV), breast cancer (MCF7), and cervical cancer (HeLa) using the MTT-assay. Cell cycle progression and induction of apoptosis were assessed by flow cytometry and immunoblot analysis of PARP-cleavage. RESULTS: FP did not affect cell cycle progression and proliferation of GCT cell lines at sublethal doses. At higher concentrations, cell death occurred independent of cell cycle progression. The IC50 was approximately fivefold lower for the three GCT cell lines (60/60/70 nM) than for the other tumour cell lines tested (350/280/300 nM). Lethal doses in vitro were markedly lower than plasma concentrations of FP achieved in clinical studies. In vitro sensitivity to FP did not correlate with that to cisplatin. The cell lines NTera2 and NCCIT showed comparable responses to FP despite differing in their IC50 to cisplatin by factor 4. Flow cytometry and immunoblot for PARP indicated apoptotic cell death induced by FP. Synergism between either cisplatin or paclitaxel and FP was not observed. However, at low concentrations, cytotoxicity of FP and cisplatin appeared to be additive. CONCLUSION: These prelinical investigations suggest a significant antitumour activity of FP in GCT. GCT derived cell lines were far more responsive to FP than cell lines derived from other solid tumours. In contrast to other models, FP did not induce cell cycle arrest in the GCT-derived cell lines tested, possibly due to the known lack of RB-expression in GCTs. However, apoptosis was induced unrelated to cell cycle progression already at low concentrations. No cross resistance between FP and cisplatin was observed. A clinical trial evaluating the activity of FP in patients with cisplatin-refractory GCTs appears to be warranted.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Embryonal/pathology , Cell Cycle/drug effects , Flavonoids/pharmacology , Piperidines/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Cisplatin/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Female , Humans , Inhibitory Concentration 50 , Ovarian Neoplasms/pathology , Paclitaxel/pharmacology , Uterine Cervical Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...