Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mar Drugs ; 22(1)2023 Dec 28.
Article in English | MEDLINE | ID: mdl-38248645

ABSTRACT

Despite recent advances in the treatment of metastatic castration-resistant prostate cancer (CRPC), treatment is inevitably hampered by the development of drug resistance. Thus, new drugs are urgently needed. We investigated the efficacy, toxicity, and mechanism of action of the marine triterpene glycoside cucumarioside A2-2 (CA2-2) using an in vitro CRPC model. CA2-2 induced a G2/M-phase cell cycle arrest in human prostate cancer PC-3 cells and caspase-dependent apoptosis executed via an intrinsic pathway. Additionally, the drug inhibited the formation and growth of CRPC cell colonies at low micromolar concentrations. A global proteome analysis performed using the 2D-PAGE technique, followed by MALDI-MS and bioinformatical evaluation, revealed alterations in the proteins involved in cellular processes such as metastatic potential, invasion, and apoptosis. Among others, the regulation of keratin 81, CrkII, IL-1ß, and cathepsin B could be identified by our proteomics approach. The effects were validated on the protein level by a 2D Western blotting analysis. Our results demonstrate the promising anticancer activity of CA2-2 in a prostate cancer model and provide insights on the underlying mode of action.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Triterpenes , Male , Humans , Glycosides/pharmacology , Triterpenes/pharmacology , Prostate
2.
Mar Drugs ; 20(12)2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36547906

ABSTRACT

The field of marine bioactive compounds (marine drugs) has evolved significantly in recent years [...].


Subject(s)
Biological Products , Neoplasms , Humans , Aquatic Organisms , Neoplasms/drug therapy , Biological Products/pharmacology
3.
Mar Drugs ; 18(12)2020 Dec 15.
Article in English | MEDLINE | ID: mdl-33333876

ABSTRACT

By the end of the year 2020, there are nine marine-derived anticancer drugs available on the market, and the field is currently growing exponentially [...].


Subject(s)
Antineoplastic Agents/therapeutic use , Marine Biology , Marine Toxins/therapeutic use , Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Biological Products , Humans , Marine Toxins/pharmacology
4.
Mar Drugs ; 18(1)2020 Jan 14.
Article in English | MEDLINE | ID: mdl-31947523

ABSTRACT

Autophagy (Ancient Greek αὐτόφαγος [autóphagos]-"self-devouring") is defined as a regulated mechanism of the degradation of unnecessary or dysfunctional cellular components [...].


Subject(s)
Autophagy/drug effects , Biological Products/therapeutic use , Pharmaceutical Preparations/administration & dosage , Humans
5.
Mar Drugs ; 18(1)2019 Dec 26.
Article in English | MEDLINE | ID: mdl-31887976

ABSTRACT

In 2019, the scientific and medical community celebrated the 50th anniversary of the introduction of the very first marine-derived drug, Cytarabine, into clinics [...].


Subject(s)
Antineoplastic Agents/isolation & purification , Aquatic Organisms , Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , History, 21st Century , Humans
6.
Mar Drugs ; 16(2)2018 Jan 24.
Article in English | MEDLINE | ID: mdl-29364147

ABSTRACT

By the end of 2017, there were seven marine-derived pharmaceutical substances that have been approved by the FDA for clinical use as drugs[...].


Subject(s)
Antineoplastic Agents/pharmacology , Marine Toxins/pharmacology , Animals , Humans , Neoplasms/drug therapy , Periodicals as Topic
7.
Int J Mol Sci ; 20(1)2018 Dec 21.
Article in English | MEDLINE | ID: mdl-30577584

ABSTRACT

Despite high cure rates, about 20% of patients with advanced germ cell tumors (GCTs) fail cisplatin-based chemotherapy. High levels of DNA methylation have been identified in GCTs and linked to cisplatin resistance. Here, we examined the effects of DNA hypomethylating 5-azacitidine (5-aza) on two embryonal carcinoma cell lines (NCCIT, 2102Ep) and their cisplatin-resistant isogenic derivatives. Effects on cell viability and cisplatin sensitivity were assessed by the trypan blue exclusion method. Western blotting was used to examine induction of apoptosis 5-aza and results were validated by flow cytometry. Single agent treatment with 5-aza strongly impacted viability and induced apoptosis at low nanomolar concentrations, both in cisplatin-sensitive and -resistant cell lines. 5-aza exerted an immediate apoptotic response, followed by a prolonged inhibitory effect on cell viability and cell-cycle progression. Sequential treatment with 5-aza and cisplatin reduced cellular survival of the cisplatin-resistant sublines already at nanomolar concentrations, suggesting a partial restoration of cisplatin sensitivity by the compound. 5-aza demonstrated anti-tumor activity as a single agent at low nanomolar concentrations in GCT cells, irrespective of cisplatin-sensitivity. 5-aza may also have the potential at least to partially restore cisplatin-sensitivity in non-seminoma cells, supporting the hypothesis that combining DNA demethylating agents with cisplatin-based chemotherapy may be a valid therapeutic approach in patients with refractory GCTs.


Subject(s)
Apoptosis/drug effects , Azacitidine/pharmacology , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/metabolism , Testicular Neoplasms/genetics , Testicular Neoplasms/metabolism , Apoptosis/genetics , Biomarkers , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , DNA Methylation , Humans , Inhibitory Concentration 50 , Male
8.
Proteomics ; 17(11)2017 Jun.
Article in English | MEDLINE | ID: mdl-28445005

ABSTRACT

Rhizochalinin (Rhiz) is a novel marine natural sphingolipid-like compound, which shows promising in vitro and in vivo activity in human castration-resistant prostate cancer. In the present study, a global proteome screening approach was applied to investigate molecular targets and biological processes affected by Rhiz in castration-resistant prostate cancer. Bioinformatical analysis of the data predicted an antimigratory effect of Rhiz on cancer cells. Validation of proteins involved in the cancer-associated processes, including cell migration and invasion, revealed downregulation of specific isoforms of stathmin and LASP1, as well as upregulation of Grp75, keratin 81, and precursor IL-1ß by Rhiz. Functional analyses confirmed an antimigratory effect of Rhiz in PC-3 cells. Additionally, predicted ERK1/2 activation was confirmed by Western blotting analysis, and revealed prosurvival effects in Rhiz-treated prostate cancer cells indicating a potential mechanism of resistance. A combination of Rhiz with MEK/ERK inhibitors PD98059 (non-ATP competitive MEK1 inhibitor) and FR180204 (ATP-competitive ERK1/2 inhibitor) resulted in synergistic effects. This work provides further insights into the molecular mechanisms underlying Rhiz bioactivity. Furthermore, our research is exemplary for the ability of proteomics to predict drug targets and mode of action of natural anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Fatty Alcohols/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Prostatic Neoplasms/metabolism , Proteome/analysis , Adaptor Proteins, Signal Transducing/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cytoskeletal Proteins/metabolism , Drug Screening Assays, Antitumor , Fibroblasts/drug effects , Fibroblasts/metabolism , HSP70 Heat-Shock Proteins/metabolism , Humans , Interleukin-1beta/metabolism , Keratins, Hair-Specific/metabolism , Keratins, Type II/metabolism , LIM Domain Proteins/metabolism , MAP Kinase Signaling System/drug effects , Male , Membrane Proteins/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Proteomics/methods , Rhizoctonia/chemistry , Stathmin/metabolism
9.
J Cell Mol Med ; 21(7): 1300-1314, 2017 07.
Article in English | MEDLINE | ID: mdl-28026145

ABSTRACT

Type II testicular germ cell cancers (TGCT) are the most frequently diagnosed tumours in young men (20-40 years) and are classified as seminoma or non-seminoma. TGCTs are commonly treated by orchiectomy and chemo- or radiotherapy. However, a subset of metastatic non-seminomas (embryonal carcinomas) displays only incomplete remission or relapse and requires novel treatment options. Recent studies have shown effective application of the small-molecule inhibitor JQ1 in tumour therapy, which interferes with the function of 'bromodomain and extraterminal (BET)' proteins. JQ1-treated TGCT cell lines display up-regulation of genes indicative for DNA damage and cellular stress response and induce cell cycle arrest. Embryonal carcinoma (EC) cell lines, which presented as JQ1 sensitive, display down-regulation of pluripotency factors and induction of mesodermal differentiation. In contrast, seminoma-like TCam-2 cells tolerated higher JQ1 concentrations and were resistant to differentiation. ECs xenografted in vivo showed a reduction in tumour size, proliferation rate and angiogenesis in response to JQ1. Finally, the combination of JQ1 and the histone deacetylase inhibitor romidepsin allowed for lower doses and less frequent application, compared with monotherapy. Thus, we propose that JQ1 in combination with romidepsin may serve as a novel therapeutic option for (mixed) TGCTs.


Subject(s)
Apoptosis/drug effects , Azepines/administration & dosage , Cell Cycle Checkpoints/drug effects , Neoplasms, Germ Cell and Embryonal/drug therapy , Testicular Neoplasms/drug therapy , Triazoles/administration & dosage , Adult , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/pathology , Testicular Neoplasms/genetics , Testicular Neoplasms/pathology , Xenograft Model Antitumor Assays
10.
BMC Cancer ; 17(1): 93, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28143426

ABSTRACT

BACKGROUND: Advanced urothelial carcinomas represent a considerable clinical challenge as they are difficult to treat. Platinum-based combination regimens obtain response rates ranging from 40 to 70% in first-line therapy of advanced urothelial carcinoma. In the majority of cases, however, the duration of these responses is limited, and when progression occurs, the outcome is generally poor. Therefore, novel therapeutic strategies are urgently needed. The purpose of the current research is to investigate the anticancer effects and the mode of action of the marine triterpene glycoside frondoside A in p53-wild type and p53-deficient human urothelial carcinoma cells. METHODS: Activity of frondoside A was examined in the human urothelial carcinoma cell lines RT112, RT4, HT-1197, TCC-SUP, T-24, and 486p. Effects of frondoside A on cell viability, either alone or in combination with standard cytotoxic agents were investigated, and synergistic effects were analyzed. Pro-apoptotic activity was assessed by Western blotting and FACS, alone and in combination with a caspases-inhibitor. The impact of functional p53 was investigated by siRNA gene silencing and the p53 inhibitor pifithrin-α. Effects on autophagy were studied using LC3B-I/II and SQSTM/p62 as markers. The unpaired Student's t-test was used for comparison of the data sets. RESULTS: Frondoside A shows high cytotoxicity in urothelial carcinoma cells with IC50s ranging from 0.55 to 2.33 µM while higher concentrations of cisplatin are required for comparable effects (IC50 = 2.03 ~ 5.88 µM). Induction of apoptosis by frondoside A was associated with the regulation of several pro-apoptotic factors, like caspase-3, -8, and -9, PARP, Bax, p21, DNA fragmentation, and externalization of phosphatidylserine. Remarkably, inhibition of p53 by gene silencing or pifithrin-α pretreatment, as well as caspase inhibition, did not suppress apoptotic activity of frondoside A, while cisplatin activity, in contrast, was significantly decreased. Frondoside A inhibited pro-survival autophagy, a known mechanism of drug resistance in urothelial carcinoma and showed synergistic activity with cisplatin and gemcitabine. CONCLUSIONS: A unique combination of properties makes marine compound frondoside A a promising candidate for the treatment of human urothelial carcinomas.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Glycosides/pharmacology , Triterpenes/pharmacology , Animals , Carcinoma, Transitional Cell/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Screening Assays, Antitumor , Drug Synergism , Humans , Sea Cucumbers/chemistry , Tumor Suppressor Protein p53/metabolism , Urinary Bladder Neoplasms/drug therapy , Gemcitabine
11.
Proteomics ; 16(10): 1590-603, 2016 05.
Article in English | MEDLINE | ID: mdl-27001414

ABSTRACT

Monanchocidin A (MonA) is a novel marine alkaloid with promising anti-cancer properties. We recently demonstrated its high efficacy in human urogenital cancers including germ cell tumors. Here, we applied a global proteome screening approach to investigate molecular targets and biological processes affected by MonA in the human cisplatin-resistant germ cell cancer cell line NCCIT-R. Bioinformatical analysis of the proteomics data predicted an effect of MonA on cancer cell migration. Thus, proteins known to be involved in cancer cell migration and invasion were chosen for further validation. The protein alterations identified by proteomics resulted from both, regulation of the total protein expression and post-transcriptional modifications. Among others, regulation of an isoform of vimentin, up-regulation of multiple apolipoprotein E isoforms, and inhibition of hypusination of eukaryotic translation initiation factor 5A-1 were found upon treatment with MonA. Further functional analyses were performed and revealed decreased cell migration and colony formation of cancer cells treated with MonA at non-cytotoxic and non-antiproliferative concentrations. This work provides further insights into the molecular mechanisms behind MonA bioactivity. Furthermore, our research is exemplary for the ability of proteomics to predict drug targets and mode of action of natural anti-cancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Guanidine/analogs & derivatives , Proteome/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Gene Expression/drug effects , Guanidine/pharmacology , Humans , Proteome/genetics , Proteomics
12.
Int J Cancer ; 138(10): 2450-65, 2016 May 15.
Article in English | MEDLINE | ID: mdl-26695519

ABSTRACT

Despite recent advances in the treatment of metastatic castration-resistant prostate cancer (CRPC), outcome of patients remains poor due to the development of drug resistance. Thus, new drugs are urgently needed. We investigated efficacy, toxicity and mechanism of action of marine triterpene glycoside frondoside A (FrA) using CRPC cell lines in vitro and in vivo. FrA revealed high efficacy in human prostate cancer cells, while non-malignant cells were less sensitive. Remarkably, proliferation and colony formation of cells resistant to enzalutamide and abiraterone (due to the androgen receptor splice variant AR-V7) were also significantly inhibited by FrA. The marine compound caused cell type specific cell cycle arrest and induction of caspase-dependent or -independent apoptosis. Up-regulation or induction of several pro-apoptotic proteins (Bax, Bad, PTEN), cleavage of PARP and caspase-3 and down-regulation of anti-apoptotic proteins (survivin and Bcl-2) were detected in treated cells. Global proteome analysis revealed regulation of proteins involved in formation of metastases, tumor cell invasion, and apoptosis, like keratin 81, CrkII, IL-1ß and cathepsin B. Inhibition of pro-survival autophagy was observed following FrA exposure. In vivo, FrA inhibited tumor growth of PC-3 and DU145 cells with a notable reduction of lung metastasis, as well as circulating tumor cells in the peripheral blood. Increased lymphocyte counts of treated animals might indicate an immune modulating effect of FrA. In conclusion, our results suggest that FrA is a promising new drug for the treatment of mCRPC. Induction of apoptosis, inhibition of pro-survival autophagy, and immune modulatory effects are suspected modes of actions.


Subject(s)
Antineoplastic Agents/pharmacology , Glycosides/pharmacology , Triterpenes/pharmacology , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Reproducibility of Results , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
13.
Anticancer Drugs ; 27(2): 89-98, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26479145

ABSTRACT

Germ cell tumors (GCTs) are the most frequent malignancy in male patients between 15 and 45 years of age. Cisplatin-based chemotherapy shows excellent cure rates, but patients with cisplatin-resistant GCTs have a poor prognosis. Nintedanib (BIBF 1120, Vargatef) inhibits the receptor classes vascular endothelial growth factor receptor, platelet derived growth factor receptor, and fibroblast growth factor receptor, and has shown activity against many tumors, as well as in idiopathic lung fibrosis and bleomycin-induced lung injury. Here, we investigated the antineoplastic and antiangiogenic properties of nintedanib in cisplatin-resistant and cisplatin-sensitive GCT cells, both alone and in combination with classical cytotoxic agents such as cisplatin, etoposide, and bleomycin. The half-maximal inhibitory concentration (IC50) of nintedanib was 4.5 ± 0.43 µmol/l, 3.1 ± 0.45 µmol/l, and 3.6 ± 0.33 µmol/l in cisplatin-sensitive NTERA2, 2102Ep, and NCCIT cells, whereas the IC50 doses of the cisplatin-resistant counterparts were 6.6 ± 0.37 µmol/l (NTERA2-R), 4.5 ± 0.83 µmol/l (2102Ep-R), and 6.1 ± 0.41 µmol/l (NCCIT-R), respectively. Single treatment with nintedanib induced apoptosis and resulted in a sustained reduction in the capacity of colony formation in both cisplatin-sensitive and cisplatin-resistant GCT cells. Cell cycle analysis showed that nintedanib induced a strong G0/G1-phase arrest in all investigated cell lines. Combination treatment with cisplatin did not result in additive, synergistic, or antagonistic effects. The in-vivo activity was studied using the chorioallantoic membrane assay and indicated the antiangiogenic potency of nintedanib with markedly reduced microvessel density. Topical treatment of inoculated tumor plaques resulted in a significant reduction of the tumor size. This indicates that nintedanib might be a promising substance in the treatment of GCT.


Subject(s)
Antineoplastic Agents/pharmacology , Indoles/pharmacology , Neoplasms, Germ Cell and Embryonal/pathology , Angiogenesis Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Bleomycin/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor/drug effects , Cell Survival/drug effects , Chick Embryo , Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/drug effects , Cisplatin/pharmacology , Drug Interactions , Drug Resistance, Neoplasm , Etoposide/pharmacology , Humans , Male , Neoplasms, Germ Cell and Embryonal/drug therapy , Neoplasms, Germ Cell and Embryonal/metabolism
14.
Mar Drugs ; 14(7)2016 Jul 15.
Article in English | MEDLINE | ID: mdl-27428983

ABSTRACT

Guanidine alkaloids from sponges Monanchora spp. represent diverse bioactive compounds, however, the mechanisms underlying bioactivity are very poorly understood. Here, we report results of studies on cytotoxic action, the ability to inhibit EGF-induced neoplastic transformation, and the effects on MAPK/AP-1 signaling of eight rare guanidine alkaloids, recently isolated from the marine sponge Monanchora pulchra, namely: monanchocidin A (1), monanchocidin B (2), monanchomycalin C (3), ptilomycalin A (4), monanchomycalin B (5), normonanchocidin D (6), urupocidin A (7), and pulchranin A (8). All of the compounds induced cell cycle arrest (apart from 8) and programmed death of cancer cells. Ptilomycalin A-like compounds 1-6 activated JNK1/2 and ERK1/2, following AP-1 activation and caused p53-independent programmed cell death. Compound 7 induced p53-independent cell death without activation of AP-1 or caspase-3/7, and the observed JNK1/2 activation did not contribute to the cytotoxic effect of the compound. Alkaloid 8 induced JNK1/2 (but not ERK1/2) activation leading to p53-independent cell death and strong suppression of AP-1 activity. Alkaloids 1-4, 7, and 8 were able to inhibit the EGF-induced neoplastic transformation of JB6 P⁺ Cl41 cells. Our results suggest that investigated guanidine marine alkaloids hold potential to eliminate human cancer cells and prevent cancer cell formation and spreading.


Subject(s)
Alkaloids/pharmacology , Cell Transformation, Neoplastic/drug effects , Epidermal Growth Factor/metabolism , Guanidines/pharmacology , Porifera/chemistry , Alkaloids/chemistry , Animals , Cell Cycle Checkpoints/drug effects , Cell Death/drug effects , Cell Line , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Guanidine/analogs & derivatives , Guanidine/chemistry , Guanidine/pharmacology , Guanidines/chemistry , HeLa Cells , Humans , MAP Kinase Signaling System/drug effects , Mice , Transcription Factor AP-1/metabolism , Tumor Suppressor Protein p53/metabolism
15.
Oncology ; 89(2): 70-8, 2015.
Article in English | MEDLINE | ID: mdl-25765871

ABSTRACT

OBJECTIVE: The efficacy of second-line treatment after failure of platinum-based chemotherapy in patients with advanced urothelial cancer is limited. Based on encouraging preclinical and clinical phase I data, we evaluated the safety and efficacy of the combination of paclitaxel and everolimus in these patients. METHODS: In this trial, patients having failed to respond to prior platinum-based combination treatment of urothelial cancer were treated with paclitaxel (175 mg/m(2) i.v., 3-weekly) and the mTOR-inhibitor everolimus (10 mg p.o., once daily). The patients were treated until tumor progression or until a maximum of 6 cycles was completed. A one-stage design was used to evaluate the objective response rate (ORR) as the primary endpoint. RESULTS: A total of 27 patients (67% male; median age 63 years) were enrolled. The most frequent grade III/IV toxicities were anemia (28%), peripheral neuropathy (28%), and fatigue (24%). No treatment-related deaths were reported. Complete and partial remissions were observed in 0/24 and 3/24 patients eligible for efficacy analysis, respectively (ORR 13%). Progression-free survival was 2.9 months [95% confidence interval (95% CI) 1.9-4.2], and the median overall survival was 5.6 months (95% CI 4.8-10.2). CONCLUSION: The combination of paclitaxel and everolimus has not achieved the expected efficacy in second-line treatment of urothelial cancer and should not be further explored.


Subject(s)
Antineoplastic Agents/administration & dosage , Everolimus/administration & dosage , Paclitaxel/administration & dosage , Urologic Neoplasms/drug therapy , Urothelium/drug effects , Adult , Aged , Antineoplastic Agents/adverse effects , Everolimus/adverse effects , Female , Germany , Humans , Male , Middle Aged , Paclitaxel/adverse effects , Quality of Life , Treatment Outcome , Urologic Neoplasms/pathology , Urothelium/pathology
16.
Curr Oncol Rep ; 17(12): 54, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26449842

ABSTRACT

Male germ cell tumors (GCTs) are curable cancers, yet 10-15 % of patients with metastatic disease fail cisplatin-based first-line treatments. While therapeutic options have increased for various other cancers, little progress has been made in the management of GCT in the last decades. A better understanding of the molecular alterations underlying the disease and identification of new therapeutic targets are needed. Several phase I/II studies with promising new agents are ongoing or have been completed, but most of those trials have been small and have not included translational research. Therefore, molecular profiles predictive for response or new agents have not been identified in male GCT so far. The purpose of this review is to highlight emerging targets and therapies with the potential to improve systemic treatment of metastatic male GCT and to develop strategies for future clinical trials.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms, Germ Cell and Embryonal/drug therapy , Testicular Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cisplatin/therapeutic use , Clinical Trials as Topic , Humans , Immunotherapy/methods , Male , Molecular Targeted Therapy/methods , Neoplasms, Second Primary/drug therapy , Protein Kinase Inhibitors/therapeutic use
17.
Breast ; 77: 103765, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-39002281

ABSTRACT

PURPOSE: Although younger age has been negatively associated with persistence to adjuvant endocrine therapy (ET), factors contributing to non-persistence remain poorly understood. We assessed factors associated with non-persistence to ET and described the 5-year trajectories of quality of life (QoL) and symptoms in young women (≤40 years) with hormone receptor-positive breast cancer (BC). METHODS: We retrieved data on clinical characteristics and non-persistence from the medical annual records in the European cohort of the "Helping Ourselves, Helping Others: The Young Women's BC Study" (IBCSG 43-09 HOHO). Women completed surveys at baseline, biannually for three years, and annually for another seven years. Data collection included sociodemographic information, QoL aspects assessed by the Cancer Rehabilitation Evaluation System-Short Form and symptoms assessed by the Breast Cancer Prevention Trial symptom scales. Cox regression models were applied to identify factors associated with non-persistence. RESULTS: The cumulative risk of interrupting ET within 5 years was 27.7 % (95 % CI, 21.5-35.2). The QoL subscale scores remained stable over 5 years, with slight improvements in the physical subscale. Hot flashes decreased (p < 0.001), while vaginal problems intensified (p < 0.001) over time. Being married without children and having difficulties interacting and communicating with the medical team were significantly associated with non-persistence. CONCLUSIONS: Discussing the desire to conceive with partnered childless women and establishing a good relationship with the medical team may be important in addressing the non-persistence in young BC survivors. As recent data suggests the safety of pausing ET to conceive, this approach may be a reasonable future option to limit non-persistence.

18.
Onkologie ; 36(6): 371-3, 2013.
Article in English | MEDLINE | ID: mdl-23774153

ABSTRACT

BACKGROUND: Intracranial germinomas (IG) are rare and highly curable tumors. The incidence and optimal treatment of recurrences are not well defined. CASE REPORT: A 34-year-old male was diagnosed with a late recurrence of an IG 14 years after the initial diagnosis and treatment. The diagnosis was complicated by the absence of tumor markers and delayed histological sampling of the lesion. Upon histological confirmation, the patient received 2 cycles of conventional chemotherapy, followed by 2 cycles of highdose chemotherapy and peripheral blood stem cell transplantation. The patient achieved a complete remission on magnetic resonance imaging scan. Consolidating radiation of the involved field was performed after termination of the chemotherapy. CONCLUSION: Limited information on the optimal management of late relapses of IG call for individualized therapeutic approaches. Platinum-based chemotherapy, followed by high-dose chemotherapy and consolidative radiation, appears to be feasible and effective in this situation.


Subject(s)
Brain Neoplasms/therapy , Chemoradiotherapy , Germinoma/therapy , Neoplasm Recurrence, Local/therapy , Peripheral Blood Stem Cell Transplantation , Pineal Gland , Pinealoma/therapy , Adult , Antineoplastic Agents/administration & dosage , Brain Neoplasms/diagnosis , Combined Modality Therapy/methods , Germinoma/diagnosis , Humans , Male , Neoplasm Recurrence, Local/diagnosis , Pinealoma/diagnosis , Radiotherapy, Conformal , Treatment Outcome
19.
Onkologie ; 36(11): 663-8, 2013.
Article in English | MEDLINE | ID: mdl-24192771

ABSTRACT

BACKGROUND: Germ cell tumor (GCT) patients are at risk of venous thromboembolic events (VTEEs). A higher incidence of VTEEs has been reported in GCT patients undergoing cisplatin-based chemotherapy. PATIENTS AND METHODS: A retrospective analysis of the incidence of and risk factors for VTEEs in 193 GCT patients receiving platinum-based chemotherapy in Hamburg, Germany, between 2000 and 2009 was performed. RESULTS: VTEEs occurred in 22 patients (11%). In only 4 patients, the VTEEs occurred during cisplatin-based chemotherapy, while 18 patients (81%) experienced VTEEs prior to initiation of chemotherapy. Pure seminoma, 'intermediate risk' (International Germ Cell Cancer Collaborative Group (IGCCCG)), retroperitoneal or supraclavicular lymph node metastases, elevated lactate dehydrogenase (LDH) levels, a central venous catheter (CVC), arterial hypertension, application of granulocyte colony-stimulating factor (G-CSF) and of ≥ 3 cycles of cisplatin-based chemotherapy could be identified as risk factors. 2 risk groups could be described: (i) VTEEs manifesting before chemotherapy in patients with seminoma, retroperitoneal tumor masses, and elevated LDH levels, and (ii) VTEEs occurring during chemotherapy applied via CVC in patients with supraclavicular lymph node metastases. CONCLUSIONS: The incidence of VTEEs in GCT patients was 11%, but in the majority of patients, the VTEEs occurred before the initiation of platinum-based chemotherapy. Supraclavicular lymph node metastases and use of a CVC are risk factors for VTEEs during chemotherapy.


Subject(s)
Cisplatin/therapeutic use , Drug-Related Side Effects and Adverse Reactions/mortality , Neoplasms, Germ Cell and Embryonal/drug therapy , Neoplasms, Germ Cell and Embryonal/mortality , Venous Thromboembolism/mortality , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Female , Germany/epidemiology , Humans , Incidence , Male , Middle Aged , Retrospective Studies , Risk Factors , Survival Rate , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL