Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Klin Onkol ; 33(4): 248-259, 2020.
Article in English | MEDLINE | ID: mdl-32894953

ABSTRACT

Recently, the World Health Organization (WHO) classification of tumours of the central nervous system (CNS) has brought essential changes. The currently valid revised WHO 2016 classification of CNS tumours introduced the concept of integrated dia-gnostics, which incorporated not only histopathological morphological finding and immunophenotype but also molecular-genetic characteristics of the tumour. Thus, the final integrated dia-gnosis comprises the traditional morphological and growth pattern characteristics of a tumour including histopathological grade and also specific molecular bio-markers. The classification of tumour based on a combination of both tumour phenotype and genotype enables more precise prognostic stratification, increases the objectivity of dia-gnostics and prediction of response to treatment. In 2017, an international platform, The Consortium to Inform Molecular and Practical Approaches to CNS Tumor Taxonomy - not official WHO (cIMPACT-NOW), was established to create and formulate practical recommendations for integrated dia-gnostics of CNS tumours and upcoming WHO classification. The incorporation of molecular bio-markers into the integrated dia-gnostics radically changed the classification of diffuse gliomas, which include entities with different morphological characteristics, genetic alterations and bio-logical behaviour. This review article summarizes essential morphological, immunophenotypical and molecular genetic characteristics of diffuse gliomas within the scope of integrated dia-gnostics according to the valid WHO classification of tumours of the CNS and subsequent recommendations of dia-gnostic approaches. This work was supported by grant of the Ministry of Health of the Czech Republic - Conceptual Development of a Research Organization (MMCI 00209805) and Grant Agency of Masaryk University (MUNI/A/1562/2018). The authors declare they have no potential conflicts of interest concerning drugs, products, or services used in the study. The Editorial Board declares that the manuscript met the ICMJE recommendation for biomedical papers.


Subject(s)
Biomarkers, Tumor/analysis , Brain Neoplasms/classification , Brain Neoplasms/diagnosis , Glioma/classification , Glioma/diagnosis , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Glioma/genetics , Glioma/metabolism , Humans
2.
Klin Onkol ; 33(1): 15-19, 2020.
Article in English | MEDLINE | ID: mdl-32075383

ABSTRACT

Modern immunotherapy with checkpoint inhibitors has become the backbone treatment for many cancers. However, it is often accompanied by immune-related side effects, which may differ depending on the nature of the treatment. The frequency of adverse reactions increases with the number of patients receiving immunotherapy. The situation has become even more difficult with the advent of combination immunotherapy. Although the kinetics of the onset and duration of toxicity have been well described, caution should be exercised. In clinical practice, cases with atypical courses often occur. Ignorance of the problem can lead to underestimation of symptoms and damage to the patient. Immune-related side effects are variable and any organ can be affected. In addition to skin, intestinal and liver toxicity, immune-related endocrinopathy is another relatively frequent toxicity. Thyroid, pituitary and adrenal glands are most commonly affected. Symptoms of endocrinopathy are often nonspecific, which may complicate a differential diagnosis. Fortunately, most toxicities are grade 1 and 2; however, in routine clinical practice, care must be exercised to detect the onset of life-threatening toxicity such as an adrenal crisis or type 1 diabetes mellitus with ketoacidosis. It is unclear whether high doses of corticosteroids are effective in preserving endocrine gland function. Long-term hormone replacement therapy is essential because immune-related endocrinopathy is often irreversible, unlike other immune-related toxicities. Close cooperation with an endocrinologist is therefore very important. This work was supported by MH CZ - DRO (MMCI, 00209805). The authors declare they have no potential conflicts of interest concerning drugs, products, or services used in the study. The Editorial Board declares that the manuscript met the ICMJE recommendation for biomedical papers.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , Endocrine System Diseases/chemically induced , Endocrine System Diseases/diagnosis , Endocrine System Diseases/epidemiology , Humans , Immunotherapy/adverse effects , Neoplasms/drug therapy
3.
Klin Onkol ; 32(2): 97-100, 2019.
Article in English | MEDLINE | ID: mdl-30995848

ABSTRACT

BACKGROUND: With advancements in diagnostic techniques, oligometastatic prostate cancer is diagnosed in patients who were, in the past, considered to have localized disease. Moreover, evidence of the effectiveness of treatment intensification for this disease is increasing, focusing on primary tumors as well as metastatic lesions. Thus, we can delay the start of systemic palliative treatment and improve overall survival. Many questions remain unclear, such as the definition of oligometastasis disease, or which patients should be offered aggressive treatment. Data are limited and come from small retrospective studies but show conclusively the benefits of survival in targeted primary prostate and metastatic prostate cancer therapy with surgery or radiotherapy. Often, stereotactic radiotherapy is used in this indication, with minimal side effects. In retrospective studies, 3-5 metastatic lesions were generally accepted for definition of oligometastatic disease, but patient subgroups were heterogeneous. A recent study attempts to better define oligometastatic disease and find out the right degree of intensification of treatment. When and in which patient to use metastasis-targeted therapy and when the standard systemic treatment is already meaningful. It is already clear that selected patients benefit from targeted personalized treatment. PURPOSE: The purpose of this review is to offer an update of the problem of oligometastatic prostate cancer. The article presents an overview of data from contemporary literature, modern possibilities of diagnostic imaging methods and treatment options of oligometastatic prostate cancer including surgery and radiotherapy.  authors declare they have no potential conflicts of interest concerning drugs, products, or services used in the study. The Editorial Board declares that the manuscript met the ICMJE recommendation for biomedical papers. Submitted: 8. 2. 2019 Accepted: 5. 3. 2019.


Subject(s)
Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Humans , Male , Neoplasm Metastasis , Prognosis
4.
Neoplasma ; 65(2): 287-291, 2018.
Article in English | MEDLINE | ID: mdl-29534590

ABSTRACT

Patients with clinically node-positive bladder cancer have a poor prognosis, with many receiving only palliative chemo- therapy. We evaluated oncological results in bladder cancer patients with clinically regional and supraregional lymph- adenopathy treated with induction chemotherapy (IC) and consolidative cystectomy. Twenty-five patients with clinically node-positive bladder cancer (including pelvic and retroperitoneal nodes) were treated with 2-4 cycles of IC followed by consolidative cystectomy between 2010 and 2016. Pathologic complete response (pCR) was defined as no residual tumor in the final specimen (ypT0N0).The 3-year cancer-specific (CSS) and recurrence-free survival (RFS) for the whole cohort were 52% and 39%, respectively. The 3-year RFS differed according to volume of nodal metastases, the rates were 56% for minimal nodal disease (cN1) versus 33% for cN2-3 and 0% for cM1 disease (p<0.001). pCR was seen in 7 (28%) patients; 50% in cN1 versus 13% in cN3-M1. pCR associated with 3-year CSS of 80% versus 45% in patients with persistent disease after IC. In conclusion, a multimodal approach to patients with clinically node-positive bladder cancer, consisting of IC followed by consolidative surgery, may achieve long-term survival in selected patients. Better results may be expected in patients with initially minimal nodal burden and complete pathologic response to chemotherapy. Further studies are warranted to improve patient selection for consolidative surgery, especially with supra-regional metastases.


Subject(s)
Lymph Nodes/pathology , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/surgery , Cystectomy , Disease-Free Survival , Humans , Induction Chemotherapy , Prognosis , Survival Rate , Treatment Outcome
5.
Klin Onkol ; 30(Supplementum3): 55-61, 2017.
Article in Czech | MEDLINE | ID: mdl-29239194

ABSTRACT

Treatment of renal cell carcinoma is still palliative. Targeted therapy increases response rates and prolongs overall survival and progression-free survival compared with cytokines and chemotherapy. Checkpoint inhibitors constitute the up-date of therapeutic approaches, and anti-PD-1 antibody, one checkpoint inhibitor, is now well established as a second and/or third palliative treatment for patients with renal cell carcinoma. In this study, we present the latest data from current studies on cytokines, cancer vaccines, ipilimumab, and nivolumab. The therapeutic efficacies of combinations such as targeted therapy with immune checkpoint inhibitors and anti-CTLA-4 with anti PD-1 (-L1) have been reported in many studies. Preliminary results are encouraging but the high toxicities and elevated cost are limiting. Treatments with combinations of bevacizumab and atezolizumab, axitinib and pembrolizumab or avelumab, lenvatinib and pembrolizumab, and nivolumab and ipilimumab (results from study phase I, II, and sometimes III) are reported to be highly effective and to result in long-lasting responses with response-rates of 70-100%. So far, valid predictors for these therapies have not been forthcoming, but considerable work is being exerted in this area. Heng and Memorial Sloan Kettering Cancer Center (MSKCC) models are still being used to select patients for immunotherapy. Immunotherapy will definitely continue to play an important role in the treatment of patients with renal cell carcinoma; however, many questions remain.Key words: renal cell carcinoma - immunotherapy - checkpoint inhibitors - target therapy Supported by MH CZ - DRO (MMCI, 00209805) This work was supported by program of the Czech Ministry of Health No. P03-15-34 678A. The authors declare they have no potential conflicts of interest concerning drugs, products, or services used in the study. The Editorial Board declares that the manuscript met the ICMJE recommendation for biomedical papers.Submitted: 16. 8. 2017Accepted: 7. 9. 2017.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Cancer Vaccines/therapeutic use , Carcinoma, Renal Cell/therapy , Immunotherapy , Kidney Neoplasms/therapy , B7-H1 Antigen/antagonists & inhibitors , CTLA-4 Antigen/antagonists & inhibitors , Cytokines/immunology , Humans , Programmed Cell Death 1 Receptor/antagonists & inhibitors
6.
Klin Onkol ; 30(5): 337-342, 2017.
Article in Czech | MEDLINE | ID: mdl-29031035

ABSTRACT

BACKGROUND: The optimal treatment for low-grade gliomas remains controversial. Neurosurgery, radiotherapy, and chemotherapy are the main treatment options. Despite advances in oncology, there are still a lot of uncertainties, and the optimal sequences, combinations, and timings of these procedures have not yet been optimized. It is still unclear whether temozolomide can replace effective, but toxic PCV chemotherapy (procarbazine, lomustine, vincristine) and whether temozolomide can be used upfront alone instead of radiotherapy alone. Mature results from phase III trials (CODEL, EORTC 22033-26033) will provide answers to these questions. Correlative analyses of survival data and molecular marker findings (1p/19q codeletion, IDH1/2 mutation, and MGMT promoter methylation status) are essential. Due to slow progressive nature of the disease, all clinical trials with low-grade gliomas are complicated by the need for long-term follow-up to obtain valid mature data, which makes any new treatment procedures or developments in basic research developed during the course of closed clinical trials difficult to apply in daily clinical practice. An example is the recently published RTOG 9802 study evaluating the role of adjuvant PCV in combination with radiotherapy for the treatment of high-risk low-grade glioma patients where the recruitment of patients was initiated almost two decades ago. Health-related quality of life after treatment of patients with expected long-term survival is also very important and its maintenance is currently the focus of considerable interest. AIM: The main objective of the present review is to summarize the results of key clinical trials and highlight controversial issues that could have an impact on future daily practice. Another aim is to discuss these issues in the light of newly established molecular markers from the new 2016 WHO Classification of Tumors of the Central Nervous System.Key words: glioma - astrocytoma - radiotherapy - temozolomide - PCV - cognition This work was supported by MH CZ - RVO (MMCI, 00209805) and by project of the Ministry of Education, Youths and Sports of the Czech Republic CEITEC 2020 (LQ1601). The authors declare they have no potential conflicts of interest concerning drugs, products, or services used in the study. The Editorial Board declares that the manuscript met the ICMJE recommendation for biomedical papers.Submitted: 21. 2. 2017Accepted: 20. 3. 2017.


Subject(s)
Brain Neoplasms/therapy , Chemotherapy, Adjuvant/methods , Glioma/therapy , Radiotherapy, Adjuvant/methods , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Clinical Trials as Topic , Humans
7.
Klin Onkol ; 30(5): 343-348, 2017.
Article in Czech | MEDLINE | ID: mdl-29031036

ABSTRACT

BACKGROUND: The standard postsurgical options for low-grade gliomas include watchful waiting or radiotherapy depending on the risk factors for recurrence. The use of chemotherapy for the treatment of this disease is generally controversial, although the recently published results of the first of two large randomized phase III clinical trials (RTOG 9802 a EORTC 22033-26033), focusing on the evaluation of chemotherapy for the upfront treatment of newly diagnosed low-grade gliomas, are reassuring in this respect. The long-term results of a RTOG 9802 comparing radiotherapy alone with radiotherapy and six cycles of adjuvant PCV chemotherapy (procarbazine, lomustine, vincristine) in patients with high-risk low-grade gliomas will probably have an impact on daily clinical practice. The increase in median overall survival from 7.8 years to 13.3 years, mainly for patients with oligodendrogliomas, is unprecedented, but the toxicity of PCV is too high and molecular marker analysis remains inadequate. It is still unclear whether less toxic temozolomide can replace PCV and whether temozolomide can be used upfront alone instead of with radiotherapy. This question is addressed by the ongoing EORTC 22033-26033 study. The preliminary results show no significant difference in progression-free survival between patients receiving radiotherapy and those receiving temozolomide alone. Treatment with temozolomide was not associated with an improvement in cognitive function compared with treatment with radiotherapy. Despite limited follow-up, the study clearly confirmed the importance of molecular characterization of low-grade gliomas, as currently defined in the new 2016 WHO Classification of Tumors of the Central Nervous System. AIM: The aim of the review is to summarize available information from listed key clinical trials of chemotherapy for low-grade gliomas and draw attention to unresolved issues concerning the use of chemotherapy for the treatment of this disease.Key words: glioma - astrocytoma - chemotherapy - PCV - temozolomide - RTOG 9802 This work was supported by MH CZ - RVO (MMCI, 00209805) and by project of the Ministry of Education, Youths and Sports of the Czech Republic CEITEC 2020 (LQ1601). The authors declare they have no potential conflicts of interest concerning drugs, products, or services used in the study. The Editorial Board declares that the manuscript met the ICMJE recommendation for biomedical papers.Submitted: 21. 2. 2017Accepted: 20. 3. 2017.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Chemotherapy, Adjuvant/methods , Glioma/drug therapy , Brain Neoplasms/radiotherapy , Brain Neoplasms/surgery , Clinical Trials as Topic , Glioma/radiotherapy , Glioma/surgery , Humans , Radiotherapy, Adjuvant
8.
Neoplasma ; 64(3): 329-337, 2017.
Article in English | MEDLINE | ID: mdl-28253711

ABSTRACT

In many ongoing clinical trials, new strategies for radiotherapy of brain metastases are currently being investigated. A post surgical focal cavity stereotactic radiosurgery and the developing role of a hippocampal-sparing whole brain radiotherapy are of the highest importance. The evaluation of spatial patterns of metastases failure after radiotherapy is a powerful tool for assessing the potential benefit of new different radiotherapy approaches, which enables to identify possible directions leading to better radiotherapy techniques and to modify general management for newly diagnosed brain metastases. The purpose of this article is to present a mix between trial data and philosophical point of view for discussion about the importance of systematic evaluation of spatial patterns of failure in all ongoing trials investigating new approaches in local brain metastases treatment.


Subject(s)
Brain Neoplasms/radiotherapy , Clinical Trials as Topic , Radiosurgery , Combined Modality Therapy , Cranial Irradiation , Humans , Neoplasm Metastasis/diagnosis , Treatment Failure
9.
Klin Onkol ; 29 Suppl 4(Suppl 4): 78-87, 2016.
Article in Czech | MEDLINE | ID: mdl-27846725

ABSTRACT

BACKGROUND: The treatment of early or locally advanced stages of non-small cell lung cancer (NSCLC) is based on surgical resection or radiotherapy. Metastatic disease is always incurable, treatment is palliative, systemic based on chemotherapy or target therapy. NSCLC is the most common cause of cancer-related death worldwide, and new therapeutic approaches are needed. Based on the emerging data on the role of immune system in shaping of tumor outbreak and outcome, immunotherapy is currently in the center of interest of cancer research and therapy of solid cancers including NSCLC. Various anti-cancer vaccination approaches and antigen-independent immunomodulatory drugs are being developed and trialed. The most advanced in terms of approaching clinical practice are the so-called checkpoint inhibitors blocking cytotoxic T-lymphocyte antigen-4 (CTLA-4) or programmed cell death of the protein and its ligand (PD-1, PD-L1). Beside innovative drug development, the field of cancer immunotherapy focuses on the identification and clinical application of effective biomarkers of clinical efficacy and on the evaluation of combinations of immunotherapeutic drugs or with classical anti-cancer approaches, such as chemotherapy, radiotherapy or with targeted therapy. AIM: In this review, we summarize basic principles of immnobiology of NSCLC in the context of innovative immunotherapeutics, strategy and phase III results of anti-cancer vaccines in NSCLC, results of NSCLC treatment with checkpoint inhibitors, and current challenges in immunotherapy of lung cancers.Key words: non-small cell lung cancer - immunotherapy - cancer vaccines - drug response biomarkersThis work was supported by MEYS - NPS I - LO1413.The authors declare they have no potential conflicts of interest concerning drugs, products, or services used in the study.The Editorial Board declares that the manuscript met the ICMJE recommendation for biomedical papers.Submitted: 10. 6. 2016Accepted: 16. 6. 2016.


Subject(s)
Antineoplastic Agents/therapeutic use , Cancer Vaccines/therapeutic use , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/therapy , Immunotherapy , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , CTLA-4 Antigen/antagonists & inhibitors , Humans , Ipilimumab , Nivolumab , Programmed Cell Death 1 Receptor/antagonists & inhibitors
10.
Klin Onkol ; 29(2): 107-10, 2016.
Article in Czech | MEDLINE | ID: mdl-27081799

ABSTRACT

Thanks to a multidisciplinary approach, testicular germinative tumors are now assigned to a group of highly curable oncologic diseases with favorable prognoses. Despite the gradual increase in the incidence of this type of cancer in recent years, mortality remains low. Yet, guidelines for postoperative treatment of stage I non-seminomatous germ cell testicular tumors remain inconsistent due to the low number of randomized studies. The probability of relapse is strongly associated with the occurrence of lymphangiogenesis. The period after primarily orchiectomy can be utilized for close monitoring, cisplatin-based adjuvant chemotherapy, or retroperitoneal lymphadenectomy. All variants of therapy offer a cure rate of about 99 %. The use of adjuvant chemotherapy, as well as retroperitoneal lymph node dissection, is associated with acute and late adverse effects. The effort to minimize side effects with preserving the lowest number of relapses resulted in the need for comparing the number of chemotherapy cycles and chemotherapy vs. retroperitoneal lymphadenectomy. The aim of this review is to evaluate the different treatment modalities for stage I testicular germ cell tumors with respect to their efficacy and toxicity.


Subject(s)
Neoplasms, Germ Cell and Embryonal/therapy , Testicular Neoplasms/therapy , Humans , Male , Neoplasm Staging , Neoplasms, Germ Cell and Embryonal/pathology , Testicular Neoplasms/pathology
11.
Neoplasma ; 63(2): 263-8, 2016.
Article in English | MEDLINE | ID: mdl-26774148

ABSTRACT

Ototoxicity is an important adverse effect of using Cisplatin (cis-diamminedichloroplatinum) (CDDP) as a form of chemotherapy. The clinical picture of CDDP induced ototoxicity includes perceptive hearing impairment (reversible or permanent) and tinnitus. Ototoxicity manifests with considerable variability between patients. The objective of this prospective study was to investigate a possible genetic background to this variability. We assessed ototoxicity induced by therapeutic doses of CDDP in adult patients with germinative testicular tumors, or other tumors treated with an identical CDDP dosage scheme. Audiological examination before, during and after the treatment has shown deterioration in hearing; first in the high-frequencies and with increased CDDP cumulative doses, impairment in other frequencies as well. Occurrence of tinnitus was not dependent on the administered dose of CDDP, or the other risk factors examined in this study. The association of CDDP induced ototoxicity with genetic polymorphisms in candidate genes was examined. Our study has demonstrated an association of early onset of CDDP induced ototoxicity with the presence of two copies of GSTT1 gene (p=0,009) and with T allele of rs9332377 polymorphism in COMT gene (p=0,001).


Subject(s)
Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Genetic Predisposition to Disease/genetics , Hearing Loss/chemically induced , Hearing Loss/genetics , Tinnitus/chemically induced , Tinnitus/genetics , Adult , Antineoplastic Agents/therapeutic use , Cisplatin/therapeutic use , DNA Copy Number Variations/genetics , Female , Gene Dosage/genetics , Humans , Male , Middle Aged , Neoplasms/drug therapy , Polymorphism, Genetic/genetics , Prospective Studies , Risk Factors , Young Adult
12.
Klin Onkol ; 28 Suppl 4: 4S103-14, 2015.
Article in Czech | MEDLINE | ID: mdl-26647898

ABSTRACT

BACKGROUND: The standard treatment of advanced melanoma has been changing in recent years. Palliative chemotherapy is being replaced by more efficient targeted therapies and modern immunotherapies based on antibodies against checkpoints of the immune response (so  called checkpoint inhibitors). Todays standard ipilimumab (ant-iCTLA 4 antibody) could significantly prolong overall survival and achieved longterm disease control in about 20% of patients. There are other perspective immune modulating agents, such as anti-PD 1 antibodies (nivolumab, pembrolizumab, pidilizumab) and anti-PD L1 antibodies. Unique mechanism of action is accompanied by new types of immunerelated adverse events. AIM: The aim of the article is to summarize current knowledge about the toxicity of these antibodies and propose solutions in routine clinical practice.


Subject(s)
Antibodies, Monoclonal/adverse effects , CTLA-4 Antigen/antagonists & inhibitors , Melanoma/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Humans , Ipilimumab , Nivolumab
13.
Klin Onkol ; 26(2): 114-23, 2013.
Article in Czech | MEDLINE | ID: mdl-23718670

ABSTRACT

BACKGROUND: The incidence of renal cell carcinoma in the Czech Republic is one of the highest in the world. Curative treatment is still possible only surgically, while in the palliative treatment, partial success was reached using targeted therapies. While prognostic factors and models are commonly used in clinical practice, unfortunately, predictive biomarkers have not been found. The aim of our study was to verify the validity of selected prognostic factors on a consecutive patient cohort from the Czech population. PATIENTS AND METHODS: The patient cohort consisted of 544 patients with RCC diagnosed and/or treated at our institute from 2003 to 2010. Individual clinical and histological prognostic factors and Heng prognostic model were validated. RESULTS: Median time of follow-up for our cohort was 42 months (range 0.3-326 months), median age at diagnosis was 62 years, and almost 64% of patients were men. Distribution of clinical stages was as follows: 46.5% of I, II. 10.7%, III. 13.1%, IV. 20%. 26.4% of patients in stage I-III relapsed. We diagnosed mainly clear cell (84.6%) and papillary carcinoma (9.2%). Initially, 95.8% of patients underwent surgical treatment, systemic adjuvant and palliative treatment was applied in 3.7 and 37.7% of patients, respectively. Palliative targeted therapy was received by a total of 163 patients (30%). In first-line targeted therapy, the following median TTP was reached (in months): 10.8 for sunitinib, 6.3 for sorafenib and 5.2 months for immunotherapy. The most significant prognostic factors (p < 0.00001) were: stage of disease (HR = 9.61), size of the primary tumor (HR = 5.83), lymph nodes (HR = 8.26), presence of sarcomatoid tumor sections in the tumor (HR = 7.29), and tumor grade (HR = 4.0). Besides these, we also confirmed the prognostic importance of presence of eosinophilic granulations in the tumor (HR = 1.91, p = 0.02). When applying the Heng prognostic model, we achieved similar results for patients treated with targeted therapies. CONCLUSION: The obtained epidemiological and clinico-pathological data are consistent with previously published data. These prognostic factors can be used for a differentiated approach to patients with RCC, both for establishing follow-up plan for patients after surgery as well as indication for targeted therapies.


Subject(s)
Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/therapy , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Female , Humans , Male , Middle Aged
15.
Klin Onkol ; 26(1): 42-6, 2013.
Article in Czech | MEDLINE | ID: mdl-23528172

ABSTRACT

BACKGROUND: Malignant melanoma is considered to be highly resistant to chemotherapy, radiotherapy, hormonotherapy and standard immunotherapy (interleukin 2, interferon alpha). Radical surgery in the early stages of the disease is still the most efficient method. Since the development of immunotherapy and targeted therapy, the role of palliative chemotherapy for advanced disease may be changing. CASE: A case report regarding 44-year-old woman with extensive tumor of the pectoral wall with contralateral axillary lymphadenopathy is presented. On the basis of imaging methods, histology and immunohistochemistry, the tumor was defined as a sarcoma. Due to PAX7-FKHR fusion gene positivity, rhabdomyosarcoma was the most probable classification. The patient was treated with radical chemotherapy including iphosphamide, vincristine, actinomycin D and doxorubicin with the effect of partial regression of the tumor. This enabled radical surgery of the chest wall tumor. Pathology proved 70% necrosis of the tumor. A contralateral axillary dissection was performed with a finding of two lymph nodes infiltrated with melanoma. The immunohistochemistry markers S100, HMB-45 and Melan A were positive. This resulted in a reclassification of the chest wall tumor to malignant melanoma. The following PET/CT scan was negative. A massive progression of the disease occurred after 5 months. B-RAF mutation leads to a plan of targeted therapy with vemurafenib. CONCLUSION: The case demonstrates the limits of the sensitivity and specificity of immunohistochemical markers of melanoma and the ability of this tumor to imitate various tumors including soft tissue sarcomas. A rare -PAX7-FKHR fusion gene positivity considered specific for rhabdomyosarcoma was found. An extraordinary response to radical chemotherapy with surgical resection led to an improvement of the quality of life and to a prolonged survival comparable with the effect of new targeted treatment for malignant melanoma.


Subject(s)
Melanoma/diagnosis , Rhabdomyosarcoma/diagnosis , Sarcoma/diagnosis , Skin Neoplasms/diagnosis , Thoracic Neoplasms/diagnosis , Thoracic Wall , Adult , Combined Modality Therapy , Diagnosis, Differential , Female , Humans , Melanoma/drug therapy , Rhabdomyosarcoma/surgery , Sarcoma/surgery , Skin Neoplasms/drug therapy , Thoracic Neoplasms/surgery
16.
Klin Onkol ; 25(3): 184-7, 2012.
Article in Czech | MEDLINE | ID: mdl-22724567

ABSTRACT

BACKGROUND: Cisplatin induced ototoxicity is a serious adverse effect of cisplatin therapy. Cisplatin induced ototoxicity shows significant interindividual variability. This variability is probably based on genetic background. Recent papers describe association of cisplatin ototoxicity with allelic variants of glutathion-S-transferase coding genes. PATIENTS AND METHODS: We have analyzed 55 patients treated with cisplatin therapy without any previous hearing impairment. Audiometric examination was performed before the start of cisplatin therapy and then before and after each cycle of cisplatin. DNA isolated from peripheral blood samples was used to analyze genetic polymorphisms of selected genes coding for glutathion-S-transferases. RESULTS: We have demonstrated association of early onset of cisplatin induced hearing impairment with absence of null allele of GSTT1 (p = 0.009). Both GSTM1 gene deletion and single nucleotide polymorphism in GSTP1 gene (rs1695) did not show any association with cisplatin induced ototoxicity. CONCLUSION: Early onset of cisplatin induced hearing impairment is more probable in persons with two functional alleles of GSTT1 gene.


Subject(s)
Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Glutathione Transferase/genetics , Hearing Loss/chemically induced , Polymorphism, Genetic , Audiometry, Pure-Tone , Hearing Loss/diagnosis , Hearing Loss/genetics , Humans
17.
Diabetologia ; 54(10): 2702-12, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21773684

ABSTRACT

AIMS/HYPOTHESIS: Reactive oxygen species (ROS) generated during hyperglycaemia are implicated in the development of diabetic vascular complications. High glucose increases oxidative stress in endothelial cells and induces apoptosis. A major source of ROS in endothelial cells exposed to glucose is the NAD(P)H oxidase enzyme. Several studies demonstrated that C-peptide, the product of proinsulin cleavage within the pancreatic beta cells, displays anti-inflammatory effects in certain models of vascular dysfunction. However, the molecular mechanism underlying this effect is unclear. We hypothesised that C-peptide reduces glucose-induced ROS generation by decreasing NAD(P)H oxidase activation and prevents apoptosis METHODS: Human aortic endothelial cells (HAEC) were exposed to 25 mmol/l glucose in the presence or absence of C-peptide and tested for protein quantity and activity of caspase-3 and other apoptosis markers by ELISA, TUNEL and immunoblotting. Intracellular ROS were measured by flow cytometry using the ROS sensitive dye chloromethyl-2',7'-dichlorodihydrofluorescein diacetate (CM-H(2)-DCDFA). NAD(P)H oxidase activation was assayed by lucigenin. Membrane and cytoplasmic levels of the NAD(P)H subunit ras-related C3 botulinum toxin substrate 1 (rho family, small GTP binding protein Rac1) (RAC-1) and its GTPase activity were studied by immunoblotting and ELISA. RAC-1 (also known as RAC1) gene expression was investigated by quantitative real-time PCR. RESULTS: C-peptide significantly decreased caspase-3 levels and activity and upregulated production of the anti-apoptotic factor B cell CLL/lymphoma 2 (BCL-2). Glucose-induced ROS production was quenched by C-peptide and this was associated with a decreased NAD(P)H oxidase activity and reduced RAC-1 membrane production and GTPase activity. CONCLUSIONS/INTERPRETATION: In glucose-exposed endothelial cells, C-peptide acts as an endogenous antioxidant molecule by reducing RAC-1 translocation to membrane and NAD(P)H oxidase activation. By preventing oxidative stress, C-peptide protects endothelial cells from glucose-induced apoptosis.


Subject(s)
C-Peptide/pharmacology , Endothelial Cells/metabolism , Glucose/pharmacology , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Aorta/cytology , Apoptosis/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunoblotting , In Situ Nick-End Labeling , Real-Time Polymerase Chain Reaction , rac1 GTP-Binding Protein/genetics
18.
Klin Onkol ; 24(2): 112-20, 2011.
Article in Czech | MEDLINE | ID: mdl-21644366

ABSTRACT

BACKGROUNDS: Glioblastoma multiforme is the most common malignant primary tumor of the brain in adults. Standard therapy consists in maximal surgical resection and adjuvant concurrent chemoradiotherapy and adjuvant therapy with temozolomid. This approach improves survival in comparison with postsurgical radiotherapy alone. PATIENTS AND METHODS: Consecutive patients with histologically confirmed glioblastoma multiforme in the period from January 2003 to December 2009 underwent postoperative radiotherapy (1.8-2.0 Gy/d, total of 60 Gy) plus concurrent daily chemotherapy (temozolomide 75 mg/m2/d), followed by 6 cycles of temozolomide (150 to 200 mg/m2 for 5 days, every 28 days) and were analyzed retrospectively. The primary end point was to describe the correlation between known clinical factors, treatment and progression free survival (PFS) and overall survival (OS). We assessed the toxicity and safety of the chemoradiotherapy. RESULTS: Eighty-six patients (median age, 56 years; 60% male) were included. Most of them (> 80%) were of performance status (PS) 0-1 at the beginning of chemoradiotherapy. Total macroscopic resection was performed in 20% of the patients, subtotal in 65%, partial in 9%, and just biopsy in 6%. Median PFS was 7.0 months (2.0-35.5), median OS was 13.0 months (2.5-70). Postoperative performance status (PS), the extent of resection, and administration of planned treatment without reduction had statistically significant influences on PFS and OS. Median PFS and OS were 22.0, 7.0 and 6.0 months for PFS (p = 0.0018) in patients with PS O, 1 and 2 respectively and 32.0, 13.0 and 9.0 months for OS (p = 0.0023). Patients with total removal of tumor had longer PFS (14.0 vs 6.0 months, HR = 0.5688; p = 0.0301) and OS (23.0 vs 12.0 months, HR 0.4977; p = 0.0093), as did patients without dose reduction of radiotherapy and/or chemotherapy. Patients with radiotherapy dose of over 54 Gy had PFS 8.0 vs 3.0 months (HR = 0.3313; p = 0.0001) and OS 15.0 vs 5.0 months (HR = 0.1730; p < 0.0001). Similarly, treatment with concurrent chemotherapy for more than 40 days was also important: PFS 8.0 vs 5.0 months (HR = 0.5300; p = 0.0023) and OS 17.0 vs 9.5 months (HR = 0.5943; p = 0.0175). Age, gender and position of tumor had no significant influence. Treatment-related hematology toxicity grades 3 and 4 occurred relatively often: thrombocytopenia (9%), leukopenia (6%), neutropenia (6%) and lymphopenia (25%). Thrombo-embolic events were dominant in non-hematology toxicity. Serious toxicity occurred mainly in the subgroup of patients with PS 2. Treatment of progression was useful in selected patients. Second surgery was of the most benefit (OS 24.0 vs 12.5 months, HR = 0.5325; p = 0.0111). CONCLUSION: Postoperative performance status, extent of resection, successful administration of the majority of planned concurrent chemoradiotherapy and possibility of surgical treatment at the time of recurrence correlate with better prognosis for our patients with glioblastoma. Our experience indicates that performance status should be the main factor in decisions about treatment intensity. Treatment of malignant glioma requires a multidisciplinary team.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Adult , Aged , Brain Neoplasms/mortality , Combined Modality Therapy , Female , Glioblastoma/mortality , Humans , Karnofsky Performance Status , Male , Middle Aged , Survival Rate , Young Adult
20.
Clin Exp Immunol ; 165(1): 60-76, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21413939

ABSTRACT

We have identified a novel interleukin (IL)-7-responsive T cell population [forkhead box P3 (FoxP3(+) ) CD4(+) CD25(+) CD127(+) ] that is comparably functionally suppressive to conventional FoxP3(+) CD4(+) CD25(+) regulatory T cells (T(regs) ). Although IL-2 is the most critical cytokine for thymic development of FoxP3(+) T(regs) , in the periphery other cytokines can be compensatory. CD25(+) CD127(+) T cells treated with IL-7 phenotypically 'matured' into the known 'classical' FoxP3(+) CD4(+) CD25(high) CD127(-) FoxP3(+) T(regs) . In freshly isolated splenocytes, the highest level of FoxP3 expression was found in CD127(+) CD25(+) T cells when compared with CD127(-) CD25(+) or CD127(+) CD25(-) cells. IL-7 treatment of CD4(+) CD25(+) T cells induced an increase in the accumulation of FoxP3 in the nucleus in vitro. IL-7-mediated CD25 cell surface up-regulation was accompanied by a concurrent down-regulation of CD127 in vitro. IL-7 treatment of the CD127(+) CD25(+) FoxP3(+) cells also resulted in up-regulation of cytotoxic T lymphocyte antigen 4 without any changes in CD45RA at the cell surface. Collectively, these data support emerging evidence that FoxP3(+) T cells expressing CD127 are comparably functionally suppressive to CD25(+) CD127(-) FoxP3(+) T cells. This IL-7-sensitive regulation of FoxP3(+) T(reg) phenotype could underlie one peripheral non-IL-2-dependent compensatory mechanism of T(reg) survival and functional activity, particularly for adaptive T(regs) in the control of autoimmunity or suppression of activated effector T cells.


Subject(s)
Cell Nucleus/metabolism , Forkhead Transcription Factors/metabolism , Interleukin-7/pharmacology , T-Lymphocyte Subsets/drug effects , T-Lymphocytes, Regulatory/drug effects , Active Transport, Cell Nucleus , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD/metabolism , CTLA-4 Antigen , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cells, Cultured , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-7 Receptor alpha Subunit/biosynthesis , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/immunology , Leukocyte Common Antigens/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...