Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cureus ; 15(9): e45365, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37849608

ABSTRACT

Kaposi sarcoma (KS) is a low-grade mesenchymal angioproliferative disorder that requires infection with human herpes virus 8 (HHV-8) for it to develop. It is commonly seen in HIV-positive patients and rarely in immunosuppressed HIV-negative patients. Rituximab is a monoclonal anti-CD20 chimeric murine/human immunoglobulin G antibody used to treat B cell lymphoproliferative diseases as well as a variety of autoimmune disorders. Several cases of iatrogenic Kaposi sarcoma (iKS) have been described after rituximab treatment. The purpose of this narrative review is to identify the presence of common clinical characteristics among rituximab-induced KS patients that could facilitate better management of this rare condition.

2.
Rom J Morphol Embryol ; 64(2): 173-180, 2023.
Article in English | MEDLINE | ID: mdl-37518874

ABSTRACT

The formation, proliferation, and evolution of glioblastoma (GB) are significantly influenced by pathological angiogenesis. This is supported by several growth factor receptors, such as the vascular endothelial growth factor receptor (VEGFR). In this experiment, we examined how the Food and Drug Administration (FDA) approved VEGFR blockers Sorafenib and Axitinib affect the viability of GB cells in vitro. Cells were cultivated in 96-well culture plates for the experiments, afterwards Sorafenib and Axitinib were administered at doses ranging from 0.3 µM to 80 µM. 2,5-Diphenyl-2H-tetrazolium bromide (MTT) assay was used to assess the impact of VEGFR inhibition on high-grade glioma (HGG) cell lines. To observe the morphological changes in cell shape, we used a 10× magnification microscopy. Our results showed that both Axitinib and Sorafenib retarded GB1B culture proliferation in a dose- and time-dependent manner in comparison to control cohorts that had not received any treatment. The half maximal inhibitory concentration (IC50) value for Axitinib was 3.5839 µM after three days of drug administration and 2.2133 µM after seven days of drug administration. The IC50 value for Sorafenib was 3.5152 µM after three days of drug administration and 1.6846 µM after seven days of drug administration. After the treatment with Axitinib or Sorafenib, very few cells became rounded and detached from the support, others remained adherent to the culture substrate, but acquired a larger, flatter shape. Our results indicate that VEGFR might serve as a key target in the treatment of GB. Although it is known that in vitro some drugs block the VEGFR more potently, clinical evidence is required to show whether this actually translates to better clinical outcomes.


Subject(s)
Antineoplastic Agents , Glioblastoma , Humans , Axitinib/pharmacology , Sorafenib/pharmacology , Glioblastoma/drug therapy , Cell Survival , Vascular Endothelial Growth Factor A/metabolism , Indazoles/pharmacology , Indazoles/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
3.
Cureus ; 14(7): e26843, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35974841

ABSTRACT

Background The neutrophil-to-lymphocyte ratio (NLR) at baseline treatment is an important marker of systemic inflammation, which is correlated with survival benefits in lung, breast, ovarian, bladder, and colorectal cancer. Programmed death-ligand 1 (PD-L1) expression is a biomarker with discording results regarding survival benefits in lung cancer. In our research, we studied the relationship between these two markers in patients with lung cancer. Methods Patients with stage I, II, III, and IV lung cancer (n = 80) were included in this retrospective study. The NLR baseline was recorded before the initiation of treatment. The NLR cut-off value was 4. PD-L1 expression was determined by immunohistochemical staining. Univariate and multivariate survival analyses were conducted to test their prognostic value. Results NLR proved to be a significant prognostic factor for progression-free survival (PFS) (p=0.002, Log Rank) with a mean PFS of 27.7 months for low NLR patients and 12.8 months for high NLR patients. It was also significant for overall survival (OS) (p=0.007, Log Rank) with a mean OS of 52 months for low NLR patients and 41.6 months for high NLR patients. The prognostic impact of PD-L1 expression on PFS and OS was not statistically significant with a mean PFS of 23.1 months for PD-L1-negative patients and 15.8 months for PD-L1-positive patients (p=0.422, Log Rank). Mean OS was 49 months for PD-L1-negative patients while for PD-L1-positive patients, it was 43.3 months (p=0.550 Log Rank). Regarding the correlation between PD-L1 expression and NLR value, PFS mean survival times were 13.1 months for PD-L1(+)/NLR>4, 15.1 months for PD-L1(-)/NLR>4, 16.4 months for PD-L1(+)/NLR<4 and 27.8 months for PD-L1(-)/NLR<4. This correlation between PFS and the combined PD-L1 and NLR prognostic factor was statistically relevant (p=0.04). For OS, the PD-L1/NLR combined prognostic factor was not statistically relevant (p=0.055). A mean PFS time of 27.8 months was reported for PD-L1(-)/NLR<4 group patients while for the other groups, the mean PFS was 14.9 months (p=0.045). In univariate analysis, the elevated NLR was significantly associated with a decreased PFS time (HR=2.31, 95% CI =1.323- 4.051, p=0.03) as well as OS (HR=3.555, 95% CI=1.310- 9.652, p=0.013). In multivariate analysis, NLR remained statistically significant for PFS (HR=2.160, 95% CI=1.148- 4.062, p=0.013) and OS (HR=4.364, 95% CI=1.474- 12.921, p=0.008) after adjusting for the factors of age, gender, tumor stage, lymph node stage, clinical stage, histology, and PD-L1 expression. PD-L1 expression was not a valid prognostic factor for progression or death in either univariate or multivariate analysis. We also stratified the disease control rate (DCR) depending on PD-L1/NLR combined factor expression. In the PD-L1(-)/NLR<4 group, we had the highest number of partial responses (PRs) and only one complete response (CR) compared to the other groups (p=0.006). Conclusions As the number of patients is limited in the present analysis, it is hypothesized that these two markers can be useful in dividing patients into two prognostic groups: the good prognostic group reunites PD-L1(+)/NLR<4 and PD-L1(-)/NLR<4 and the poor prognostic group reunites PD-L1(+)/NLR>4 and PD-L1(-)/NLR>4.

4.
Curr Health Sci J ; 48(4): 407-412, 2022.
Article in English | MEDLINE | ID: mdl-37304796

ABSTRACT

The ß-arrestins (ß-arr) family are proteins that regulate the signaling and trafficking of various G protein-coupled receptors. Out of the four members, ß-arr 1 and 2 have been proven as essential actors behind different processes that lead to the progression of cancer as cell proliferation, migration, invasion and metastasis. In addition to this, these proteins are also capable of transmitting anti-apoptotic signals, influence tumor growth rate and drug resistance. Several studies have demonstrated that ß-arr 2 overexpression corelates with an impaired overall survival and also showed that it may mediate multidrug resistance in certain types of cancer. In the current study we analyzed the effect of ß-arr 2 overexpression on proliferation and how it affects Temozolomide (TMZ) response on the CL2:6 High Grade Glioma (HGG) cell line. We observed contradictory results after transfection, with ß-arr 2 overexpressing cells having a superior proliferation rate after 24 and 48h, when compared to untransfected cells, while the opposite was noted after 72h. In terms of response to TMZ, we observed a similar contradictory pattern with modest differences between doses being observed at 24h, while the smallest and largest doses in our experiment produced opposite effects after 48h and 72h. This further underscores the scarcity of information regarding the exact roles and the importance of ß-arrs in the intrinsic mechanisms which govern cancer cells.

5.
Curr Health Sci J ; 48(3): 263-269, 2022.
Article in English | MEDLINE | ID: mdl-36815076

ABSTRACT

The oncological field benefits of extensive medical research and various types of cancer notice improvements, however glioblastoma multiforme remains one of the deadliest cancers in humans with virtually no advance in survival and clinical outcome. Temozolomide, the FDA approved drug for glioblastoma, faces numerous challenges such as resistance and side effects. To overcome these challenges, many combination therapies are currently studied. The present study analyses the effects of temozolomide in combination with doxorubicin on a glioblastoma cell line. Our results showed that both drugs displayed a cytotoxic effect on the studied cells in single administration (55% for 100µM temozolomide at 14 days, 53% for 100µM doxorubicin at 14 days), but without a synergistic effect in dual therapy. Although the results failed to produce the expected effect, they propose new research perspectives in the future.

6.
Int J Mol Sci ; 22(10)2021 May 13.
Article in English | MEDLINE | ID: mdl-34068040

ABSTRACT

The epidermal growth factor, latrophilin, and seven transmembrane domain-containing protein 1 (ELTD1), is a member of the G-protein coupled receptors (GPCRs) superfamily. Although discovered in 2001, ELTD1 has been investigated only by a few research groups, and important data about its role in normal and tumor cells is still missing. Even though its functions and structure are not yet fully understood, recent studies show that ELTD1 has a role in both physiological and pathological angiogenesis, and it appears to be a very important biomarker and a molecular target in cancer diseases. Upregulation of ELTD1 in malignant cells has been reported, and correlated with poor cancer prognosis. This review article aims to compile the existing data and to discuss the current knowledge on ELTD1 structure and signaling, and its role in physiological and neoplastic conditions.


Subject(s)
Biomarkers, Tumor/metabolism , Neoplasms/pathology , Receptors, G-Protein-Coupled/metabolism , Humans , Neoplasms/metabolism , Signal Transduction
7.
Int J Mol Sci ; 22(2)2021 Jan 08.
Article in English | MEDLINE | ID: mdl-33435537

ABSTRACT

Nowadays, due to recent advances in molecular biology, the pathogenesis of glioblastoma is better understood. For the newly diagnosed, the current standard of care is represented by resection followed by radiotherapy and temozolomide administration, but because median overall survival remains poor, new diagnosis and treatment strategies are needed. Due to the quick progression, even with aggressive multimodal treatment, glioblastoma remains almost incurable. It is known that epidermal growth factor receptor (EGFR) amplification is a characteristic of the classical subtype of glioma. However, targeted therapies against this type of receptor have not yet shown a clear clinical benefit. Many factors contribute to resistance, such as ineffective blood-brain barrier penetration, heterogeneity, mutations, as well as compensatory signaling pathways. A better understanding of the EGFR signaling network, and its interrelations with other pathways, are essential to clarify the mechanisms of resistance and create better therapeutic agents.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioma/genetics , Signal Transduction/genetics , Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Combined Modality Therapy , ErbB Receptors/genetics , ErbB Receptors/metabolism , Glioblastoma/metabolism , Glioblastoma/therapy , Glioma/metabolism , Glioma/pathology , Glioma/therapy , Humans , Signal Transduction/drug effects , Temozolomide/therapeutic use
8.
Contemp Oncol (Pozn) ; 24(1): 55-66, 2020.
Article in English | MEDLINE | ID: mdl-32514239

ABSTRACT

From all central nervous system tumors, gliomas are the most common. Nowadays, researchers are looking for more efficient treatments for these tumors, as well as ways for early diagnosis. Receptor tyrosine kinases (RTKs) are major targets for oncology and the development of small-molecule RTK inhibitors has been proven successful in cancer treatment. Mutations or aberrant activation of the RTKs and their intracellular signaling pathways are linked to several malignant diseases, including glioblastoma. The progress in the understanding of malignant glioma evolution has led to RTK targeted therapies with high capacity to improve the therapeutic response while reducing toxicity. In this review, we present the most important RTKs (i.e. EGFR, IGFR, PDGFR and VEGFR) currently used for developing cancer therapeutics together with the potential of RTK-related drugs in glioblastoma treatment. Also, we focus on some therapeutic agents that are currently at different stages of research or even in clinical phases and proved to be suitable as re-purposing candidates for glioblastoma treatment.

SELECTION OF CITATIONS
SEARCH DETAIL
...