Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Lung Cancer ; 192: 107824, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38761665

ABSTRACT

Surgical resection of pulmonary adenocarcinoma is considered to be curative but progression-free survival (PFS) has remained highly variable. Antitumor immune response may be important, however, the prognostic significance of tumor-infiltrating natural killer (NK) and regulatory T (Treg) lymphocytes is uncertain. Resected pulmonary adenocarcinoma tissues (n = 115) were studied by immunohistochemical detection of NKp46 and FoxP3 positivity to identify NK and Treg cells, respectively. Association of cell densities with clinicopathological features and progression-free survival (PFS) as well as overall survival (OS) were analyzed with a follow-up time of 60 months. Both types of immune cells were accumulated predominantly in tumor stroma. NK cell density showed association with female gender, non-smoking and KRAS wild-type status. According to Kaplan-Meier analysis, PFS and OS proved to be longer in patients with high NK or Treg cell densities (p = 0.0293 and p = 0.0375 for PFS, p = 0.0310 and p = 0.0448 for OS, respectively). Evaluating the prognostic effect of the combination of NK and Treg cell density values revealed that PFS and OS were significantly longer in NKhigh/Treghigh cases compared to the other groups combined (p = 0.0223 and p = 0.0325, respectively). Multivariate Cox regression analysis indicated that high NK cell density was independent predictor of longer PFS while high NK and high Treg cell densities both proved significant predictors of longer OS. The NKhigh/Treghigh combination also proved to be an independent prognostic factor for both PFS and OS. In conclusion, NK and Treg cells can be components of the innate and adaptive immune response at action against progression of pulmonary adenocarcinoma.

3.
Biomedicines ; 11(12)2023 Dec 13.
Article in English | MEDLINE | ID: mdl-38137525

ABSTRACT

High mortality in head and neck squamous cell carcinoma (HNSCC) is due to recurrence, metastasis, and radiochemotherapy (RCT) resistance. These phenomena are related to the tumor cell subpopulation undergoing partial epithelial to mesenchymal transition (pEMT). Repeated transforming growth factor-beta (TGF-beta-1) treatment via the p38 mitogen-activated protein kinase (p38 MAPK) signaling pathway induces pEMT in SCC-25 HNSCC cells, and activates and stabilizes the pro-EMT transcription factor Slug. We investigated the growth inhibitory, cisplatin-sensitizing, and pro-apoptotic effects of p38 MAPK inhibition in cisplatin-resistant (SCC-25) and -sensitive (UPCI-SCC090) HNSCC cell lines, using two specific p38 MAPK inhibitors, SB202190 and ralimetinib. Cell viability was measured by MTT assay; cell cycle distribution and cell death were evaluated by flow cytometry; p38 MAPK phosphorylation, Slug protein stabilization, and p38 MAPK downstream targets were investigated by Western blot. p-p38 inhibitors achieved sustained phosphorylation of p38 MAPK (Thr180/Tyr182) and inhibition of its function, which resulted in decreased phosphorylation (Thr69/71) of the downstream target pATF2 in pEMT cells. Subsequently, the p-p38 inhibition resulted in reduced Slug protein levels. In accordance, p-p38 inhibition led to sensitization of pEMT cells to cisplatin-induced cell death; moreover, p-p38 inhibitor treatment cycles significantly decreased the viability of cisplatin-surviving cells. In conclusion, clinically relevant p38 inhibitors might be effective for RCT-resistant pEMT cells in HNSCC patients.

4.
Int J Mol Sci ; 24(24)2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38139365

ABSTRACT

Similarly to our healthy organs, the tumor tissue also constitutes an ecosystem. This implies that stromal cells acquire an altered phenotype in tandem with tumor cells, thereby promoting tumor survival. Cancer cells are fueled by abnormal blood vessels, allowing them to develop and proliferate. Tumor-associated fibroblasts adapt their cytokine and chemokine production to the needs of tumor cells and alter the peritumoral stroma by generating more collagen, thereby stiffening the matrix; these processes promote epithelial-mesenchymal transition and tumor cell invasion. Chronic inflammation and the mobilization of pro-tumorigenic inflammatory cells further facilitate tumor expansion. All of these events can impede the effective administration of tumor treatment; so, the successful inhibition of tumorous matrix remodeling could further enhance the success of antitumor therapy. Over the last decade, significant progress has been made with the introduction of novel immunotherapy that targets the inhibitory mechanisms of T cell activation. However, extensive research is also being conducted on the stromal components and other cell types of the tumor microenvironment (TME) that may serve as potential therapeutic targets.


Subject(s)
Neoplasms , Tumor Microenvironment , Humans , Ecosystem , Neoplasms/metabolism , Carcinogenesis , Immunotherapy
5.
Cells ; 12(14)2023 07 24.
Article in English | MEDLINE | ID: mdl-37508582

ABSTRACT

Malignant melanoma is challenging to treat, and metastatic cases need chemotherapy strategies. Targeted inhibition of commonly mutant BRAF V600E by inhibitors is efficient but eventually leads to resistance and progression in the vast majority of cases. Numerous studies investigated the mechanisms of resistance in melanoma cell lines, and an increasing number of in vivo or clinical data are accumulating. In most cases, bypassing BRAF and resulting reactivation of the MAPK signaling, as well as alternative PI3K-AKT signaling activation are reported. However, several unique changes were also shown. We developed and used a patient-derived tumor xenograft (PDTX) model to screen resistance evolution in mice in vivo, maintaining tumor heterogeneity. Our results showed no substantial activation of the canonical pathways; however, RNAseq and qPCR data revealed several altered genes, such as GPR39, CD27, SLC15A3, IFI27, PDGFA, and ABCB1. Surprisingly, p53 activity, leading to apoptotic cell death, was unchanged. The found biomarkers can confer resistance in a subset of melanoma patients via immune modulation, microenvironment changes, or drug elimination. Our resistance model can be further used in testing specific inhibitors that could be used in future drug development, and combination therapy testing that can overcome inhibitor resistance in melanoma.


Subject(s)
Melanoma , Proto-Oncogene Proteins B-raf , Vemurafenib , Animals , Humans , Mice , Cell Line, Tumor , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Phosphatidylinositol 3-Kinases/genetics , Receptors, G-Protein-Coupled/genetics , Vemurafenib/pharmacology , Vemurafenib/therapeutic use
6.
Cancers (Basel) ; 15(4)2023 Feb 15.
Article in English | MEDLINE | ID: mdl-36831566

ABSTRACT

This Topical Collection, comprising 13 papers (10 original articles and 3 reviews), addresses various aspects of the field of melanoma progression: genomic and proteomic approaches, experimental studies, the questions of sentinel lymph node dissection, and metastasis formation of uveal and conjunctival melanomas is also discussed [...].

7.
Magy Onkol ; 66(2): 101-108, 2022 Jun 20.
Article in Hungarian | MEDLINE | ID: mdl-35724386

ABSTRACT

Skin melanoma became one of the most frequent malignancies of the skin mainly due to the increased exposure to environmental UV irradiation. However, this did not lead to the increase of mortality, mainly due to the efficient early diagnostics as well as to the development of new therapies which were based on the identification of the mutation patterns and understanding of the immunobiology. Molecular markers are important to differentiate malignant tumors from precancerous ones but also routinely available to define prognosis. Immunotherapy can be used in any molecular class of melanoma, while target therapy is only available in case of BRAF mutant form. Meanwhile these novel therapies induce new resistance mechanisms based on the development of acquired genetic alterations. Due to these facts the initial molecular profiling of the resected primary tumors must gradually be replaced by continuous monitoring.


Subject(s)
Melanoma , Skin Neoplasms , Humans , Melanoma/genetics , Melanoma/pathology , Melanoma/therapy , Pathology, Molecular , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/therapy , Melanoma, Cutaneous Malignant
8.
Pathol Oncol Res ; 28: 1610297, 2022.
Article in English | MEDLINE | ID: mdl-35531074

ABSTRACT

Characterization of the molecular mechanisms underlying antitumor immune responses and immune escape mechanisms has resulted in the development of more effective immunotherapeutic strategies, including immune checkpoint inhibitor (ICI) therapy. ICIs can induce durable responses in patients with advanced cancer in a wide range of cancer types, however, the majority of the patients fail to respond to this therapy or develop resistance in the course of the treatment. Information about the molecular mechanisms underlying primary and acquired resistance is limited. Although HLA class I molecules are crucial in the recognition of tumor antigens by cytotoxic T lymphocytes, only a few studies have investigated the role of their expression level on malignant cells in ICI resistance. To address this topic, utilizing immunohistochemical staining with monoclonal antibodies (mAbs) we analyzed HLA class I expression level in pre-treatment and post-treatment tumor samples from melanoma patients treated with ipilimumab. Twenty-nine metastases removed from six patients were available for the study, including 18 pre-treatment and 11 post-treatment lesions. Compared to metastases excised before ipilimumab therapy, post-treatment lesions displayed a significantly lower HLA class I expression level on melanoma cells; HLA class I downregulation was most marked in progressing metastases from nonresponding patients. We also evaluated the level of infiltration by CD8+ T cells and NK cells but did not find consistent changes between pre- and post-treatment samples. Our results indicate the potential role of HLA class I downregulation as a mechanism of ICI resistance.


Subject(s)
Melanoma , Skin Neoplasms , CD8-Positive T-Lymphocytes , Down-Regulation , Humans , Ipilimumab/therapeutic use , Melanoma/pathology , Skin Neoplasms/pathology
9.
Cancers (Basel) ; 14(10)2022 May 13.
Article in English | MEDLINE | ID: mdl-35626010

ABSTRACT

BACKGROUND: Head and neck squamous cell carcinomas (HNSCCs) are among the most abundant malignancies worldwide. Patients with recurrent/metastatic disease undergo combination chemotherapy containing cetuximab, the monoclonal antibody used against the epidermal growth factor receptor (EGFR). Cetuximab augments the effect of chemotherapy; however, a significant number of patients show therapy resistance. The mechanism of resistance is yet to be unveiled, although extracellular alterations of the receptor have been reported, and their role in cetuximab failure has been proposed. AIMS: Here, we investigate possible effects of the multi-exon deletion variant (EGFRvIII), and the single nucleotide polymorphism EGFR R521K on cetuximab efficacy. RESULTS: Our results show that in HNSCC patients, the EGFRvIII allele frequency is under 1%; therefore, it cannot lead to common resistance. EGFR R521K, present in 42% of the patients, is investigated in vitro in four HNSCC cell lines (two wild-type and two heterozygous for EGFR R521K). While no direct effect is found to be related to the EGFR status, cells harboring R521K show a reduced sensitivity in ADCC experiments and in vivo xenograft experiments. However, this preclinical difference is not reflected in the progression-free or overall survival of HNSCC patients. Furthermore, NK cell and macrophage presence in tumors is not related to EGFR R521K. DISCUSSION: Our results suggest that EGFR R521K, unlike reported previously, is unable to cause cetuximab resistance in HNSCC patients; therefore, its screening before therapy selection is not justifiable.

10.
Int J Mol Sci ; 23(10)2022 May 11.
Article in English | MEDLINE | ID: mdl-35628196

ABSTRACT

Similar to other malignancies, TCGA network efforts identified the detailed genomic picture of skin melanoma, laying down the basis of molecular classification. On the other hand, genome-wide association studies discovered the genetic background of the hereditary melanomas and the susceptibility genes. These genetic studies helped to fine-tune the differential diagnostics of malignant melanocytic lesions, using either FISH tests or the myPath gene expression signature. Although the original genomic studies on skin melanoma were mostly based on primary tumors, data started to accumulate on the genetic diversity of the progressing disease. The prognostication of skin melanoma is still based on staging but can be completed with gene expression analysis (DecisionDx). Meanwhile, this genetic knowledge base of skin melanoma did not turn to the expected wide array of target therapies, except the BRAF inhibitors. The major breakthrough of melanoma therapy was the introduction of immune checkpoint inhibitors, which showed outstanding efficacy in skin melanoma, probably due to their high immunogenicity. Unfortunately, beyond BRAF, KIT mutations and tumor mutation burden, no clinically validated predictive markers exist in melanoma, although several promising biomarkers have been described, such as the expression of immune-related genes or mutations in the IFN-signaling pathway. After the initial success of either target or immunotherapies, sooner or later, relapses occur in the majority of patients, due to various induced genetic alterations, the diagnosis of which could be developed to novel predictive genetic markers.


Subject(s)
Melanoma , Proto-Oncogene Proteins B-raf , Genome-Wide Association Study , Humans , Melanoma/diagnosis , Melanoma/epidemiology , Melanoma/genetics , Pathology, Molecular , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms , Melanoma, Cutaneous Malignant
11.
Int J Mol Sci ; 23(5)2022 Feb 28.
Article in English | MEDLINE | ID: mdl-35269844

ABSTRACT

We developed a human melanoma model using the HT168-M1 cell line to induce IFN-α2 resistance in vitro (HT168-M1res), which was proven to be maintained in vivo in SCID mice. Comparing the mRNA profile of in vitro cultured HT168-M1res cells to its sensitive counterpart, we found 79 differentially expressed genes (DEGs). We found that only a 13-gene core of the DEGs was stable in vitro and only a 4-gene core was stable in vivo. Using an in silico cohort of IFN-treated melanoma tissues, we validated a differentially expressed 9-gene core of the DEGs. Furthermore, using an in silico cohort of immune checkpoint inhibitor (ICI)-treated melanoma tissues, we tested the predictive power of the DEGs for the response rate. Analysis of the top four upregulated and top four downregulated genes of the DEGs identified WFDC1, EFNA3, DDX10, and PTBP1 as predictive genes, and analysis of the "stable" genes of DEGs for predictive potential of ICI response revealed another 13 genes, out of which CDCA4, SOX4, DEK, and HSPA1B were identified as IFN-regulated genes. Interestingly, the IFN treatment associated genes and the ICI-therapy predictive genes overlapped by three genes: WFDC1, BCAN, and MT2A, suggesting a connection between the two biological processes.


Subject(s)
Melanoma , Transcriptome , Animals , Cell Cycle Proteins , Chromosomal Proteins, Non-Histone/genetics , DEAD-box RNA Helicases , Gene Expression Profiling , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Humans , Immunotherapy , Melanoma/drug therapy , Melanoma/genetics , Mice , Mice, SCID , Oncogene Proteins/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Polypyrimidine Tract-Binding Protein , Proteins/genetics , SOXC Transcription Factors/genetics
12.
BMC Public Health ; 21(1): 1707, 2021 09 19.
Article in English | MEDLINE | ID: mdl-34538241

ABSTRACT

BACKGROUND: In Hungary, the mortality rate for testicular germ cell cancer (TGCC) is 0,9/100000 which is significantly higher than the EU average. We prospectively evaluated the effect of socioeconomic position on patient delay and therapy outcomes. METHODS: Questionnaires on subjective social status (MacArthur Subjective Status Scale), objective socioeconomic position (wealth, education, and housing data), and on patient's delay were completed by newly diagnosed TGCC patients. RESULTS: Patients belonged to a relatively high socioeconomic class, a university degree was double the Hungarian average, Cancer-specific mortality in the highest social quartile was 1.56% while in the lowest social quartile 13.09% (p = 0.02). In terms of patient delay, 57.2% of deceased patients waited more than a year before seeking help, while this number for the surviving patients was 8.0% (p = 0.0000). Longer patient delay was associated with a more advanced stage in non-seminoma but not in seminoma, the correlation coefficient for non-seminoma was 0.321 (p < 0.001). For patient delay, the most important variables were the mother's and patient's education levels (r = - 0.21, p = 0.0003, and r = - 0.20, p = 0.0005), respectively. Since the patient delay was correlated with the social quartile and resulted in a more advanced stage in non-seminoma, the lower social quartile resulted in higher mortality in non-seminoma patients (p = 0.005) but not in seminoma patients (p = 0.36) where the patient delay was not associated with a more advanced stage. CONCLUSIONS: Based on our result, we conclude that to improve survival, we should promote testicular cancer awareness, especially among the most deprived populations, and their health care providers.


Subject(s)
Neoplasms, Germ Cell and Embryonal , Testicular Neoplasms , Humans , Hungary/epidemiology , Male , Neoplasms, Germ Cell and Embryonal/therapy , Prospective Studies , Risk Factors , Socioeconomic Factors , Testicular Neoplasms/therapy
13.
Magy Onkol ; 65(2): 188-195, 2021 06 03.
Article in Hungarian | MEDLINE | ID: mdl-34081766

ABSTRACT

Head and neck squamous cell carcinomas (HNSCC) take many lifes worldwide. Patients with recurrent/metastatic disease receive combination chemotherapy containing anti-EGFR antibody cetuximab. However, resistance often hurdles therapy. The mechanism is yet to unveil, although EGFR extracellular alterations and activity of c-Met signaling were accused. We investigated the effects of EGFR-vIII and EGFR-R521K on cetuximab efficacy in HNSCC in cellular, xenograft, and clinical setup. Furthermore, we investigated the efficacy of c-Met inhibition in HNSCC in vitro and in vivo. We showed that EGFR-vIII is very rare in HNSCC, while the common R521K polymorphism abolishes antibody-dependent cellular cytotoxicity and in vivo antitumor effect of cetuximab. This selectivity was not reflected in immunophenotype or survival data of HNSCC patients, suggesting a more complex mechanism behind. Interestingly, c-Met inhibitor SU11274 was more effective in cetuximab-resistant, EGFR R521K heterozygous cells and xenografts, raising the possible importance of simultaneous targeting of the two receptors.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cetuximab/pharmacology , Drug Resistance, Neoplasm , ErbB Receptors/genetics , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Humans , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/genetics , Xenograft Model Antitumor Assays
14.
Pathol Oncol Res ; 26(4): 2475-2481, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32572819

ABSTRACT

Real-world evidence from clinical practices is fundamental for understanding the efficacy and tolerability of medicinal products. Patients with renal cell cancer were studied to gain data not represented by analyses conducted on highly selected patients participating in clinical trials. Our goal was to retrospectively collect data from patients with advanced renal tumours treated with pazopanib (PZ) to investigate the efficacy, frequency of side effects, and searching for predictive markers. Eighty-one patients who had received PZ therapy as first-line treatment were retrospectively evaluated. Overall survival (OS), progression-free survival (PFS) were assessed as endpoints. Median PFS and OS were 11.8 months (95% CI: 8.8-22.4); and 30.2 months (95% CI: 20.3-41.7) respectively. Severe side effects were only encountered in 11 (14%) patients. The presence of liver metastasis shortened the median PFS (5.5 vs. 14.8 months, p = 0.003). Median PFS for patients with or without side effects was 25.6 vs. 7.3 months, respectively (p = 0.0001). Patients younger than 65 years had a median OS of 41.7 months vs. 25.2 months for those over 65 years of age (p = 0.008). According to our results absence of liver metastases, younger age (<65 years) and presence of side effects proved to be independent predictive markers of better PFS and OS.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/drug therapy , Indazoles/therapeutic use , Kidney Neoplasms/drug therapy , Pyrimidines/therapeutic use , Sulfonamides/therapeutic use , Adult , Age Factors , Aged , Aged, 80 and over , Carcinoma, Renal Cell/pathology , Female , Humans , Kidney Neoplasms/pathology , Male , Middle Aged , Progression-Free Survival , Protein Kinase Inhibitors/therapeutic use , Retrospective Studies , Treatment Outcome
15.
J Immunother Cancer ; 8(1)2020 06.
Article in English | MEDLINE | ID: mdl-32554608

ABSTRACT

BACKGROUND: The clinical response to immune checkpoint inhibitors (ICIs) in only part of the treated patients, in conjunction with the potentially serious side effects associated with this type of therapy, has emphasized the need to identify biomarkers to select patients who may benefit from ICI treatment. The aim of our study was to test human leukocyte antigen (HLA) class I and II expression in melanoma metastases as potential biomarkers of response to ipilimumab and survival in patients with metastatic melanoma, since these molecules play a crucial role in the interactions of malignant cells with host's immune system. MATERIALS AND METHODS: HLA class I and II antigen expression level in pretreatment surgical tissue samples (50 lymph node and 35 cutaneous or subcutaneous metastases) from 30 patients was analyzed by immunohistochemical staining with monoclonal antibodies. Expression levels were correlated to intratumoral density of lymphocytes expressing cluster of differentiation (CD)8, CD45RO, CD4, forkhead box P3 (FOXP3) and/or programmed cell death protein 1 (PD-1), to clinical response to treatment, and to patients' survival. RESULTS: HLA class I antigen expression level in lymph node metastases, but not in cutaneous or subcutaneous metastases was significantly correlated to density of CD8+ and CD45RO+ T cells and of lymphocytes expressing PD-1, as well as to clinical response and to patients' survival. CONCLUSIONS: Our results corroborate the role of HLA class I expression level (alone or in combination with T-cell density values) as a predictive biomarker of response to ipilimumab in patients with melanoma. In addition, our results show that this association is influenced by the anatomic site of the metastasis used to measure the HLA class I antigen expression level.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Histocompatibility Antigens Class I/metabolism , Ipilimumab/therapeutic use , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Antineoplastic Agents, Immunological/pharmacology , Female , Humans , Ipilimumab/pharmacology , Male , Melanoma/mortality , Neoplasm Metastasis , Skin Neoplasms/mortality , Survival Analysis
16.
Cells ; 9(2)2020 02 12.
Article in English | MEDLINE | ID: mdl-32059478

ABSTRACT

Epithelial to mesenchymal transition (EMT) contributes to tumor progression, cancer cell invasion, and therapy resistance. EMT is regulated by transcription factors such as the protein products of the SNAI gene family, which inhibits the expression of epithelial genes. Several signaling pathways, such as TGF-beta1, IL-6, Akt, and Erk1/2, trigger EMT responses. Besides regulatory transcription factors, RNA molecules without protein translation, micro RNAs, and long non-coding RNAs also assist in the initialization of the EMT gene cluster. A challenging novel aspect of EMT research is the investigation of the interplay between tumor microenvironments and EMT. Several microenvironmental factors, including fibroblasts and myofibroblasts, as well as inflammatory, immune, and endothelial cells, induce EMT in tumor cells. EMT tumor cells change their adverse microenvironment into a tumor friendly neighborhood, loaded with stromal regulatory T cells, exhausted CD8+ T cells, and M2 (protumor) macrophages. Several EMT inhibitory mechanisms are instrumental in reversing EMT or targeting EMT cells. Currently, these mechanisms are also significant for clinical use.


Subject(s)
Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/genetics , Neoplasms/pathology , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Epithelial-Mesenchymal Transition/drug effects , Humans , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism
17.
Pathol Oncol Res ; 26(1): 317-325, 2020 Jan.
Article in English | MEDLINE | ID: mdl-30225783

ABSTRACT

Ipilimumab was the first immunotherapy approved for metastatic melanoma in decades and is currently registered as a second-line treatment. However, new immunotherapies, in combination with ipilimumab, offer even better clinical outcomes for patients compared with single-agent treatments, at the expense of improved toxicity. The aim of this study was to evaluate the feasibility of ipilimumab outside the clinical trials and to identify survival predictors for treatment benefit. Data were collected on 47 advanced melanoma patients treated with ipilimumab between 2010 and 2015 at a single center. Association of clinical characteristics (including primary tumor characteristics), serum lactate dehydrogenase (LDH), erythrocyte sedimentation rate, absolute eosinophil, lymphocyte, and neutrophil count, neutrophil/lymphocyte and eosinophil/lymphocyte ratio with toxicity and clinical outcome were assessed using univariate and multivariate analysis. Median progression-free survival at a median follow-up of 10 months was 2.7 months and median overall survival was 9.8 months. Objective response was observed in 17% of patients and the disease control rate at week 24 was 40%. The 1- and 2-year survival rates documented were 40 and 28%, respectively. Significant association between high LDH level (>1.5× upper limit of normal) and decreased overall survival was demonstrated in uni- and multivariate analysis (hazard ratio [HR]: 3.554, 95% CI: 1.225-10.306, p = 0.019). Neither biomarkers nor clinical outcome were associated with toxicity. Using baseline serum LDH to identify patients most likely to benefit from ipilimumab therapy could serve as a simple and inexpensive biomarker of clinical outcome.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor/analysis , Ipilimumab/therapeutic use , L-Lactate Dehydrogenase/blood , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Blood Sedimentation , Eosinophils , Female , Humans , Lymphocyte Count , Male , Melanoma/blood , Melanoma/mortality , Middle Aged , Neutrophils , Progression-Free Survival , Retrospective Studies , Skin Neoplasms/blood , Skin Neoplasms/mortality , Treatment Outcome , Melanoma, Cutaneous Malignant
18.
Semin Cancer Biol ; 60: 249-261, 2020 02.
Article in English | MEDLINE | ID: mdl-31419526

ABSTRACT

Tumor progression to metastatic disease is characterized by continuous genetic alterations due to instability of the genome. Immune sensitivity was found to be linked to tumor mutational burden (TMB) and the resulting amount of neoantigens. However, APOBEC activity resulting in increase in TMB causes immune evasion. On the other hand, clonal or acquired genetic loss of HLA class I also hampers immune sensitivity of tumors. Rare amplification of the PD-L1 gene in cancers may render them sensitive to immune checkpoint inhibitors but involvement of broader regions of chromosome 9p may ultimately lead again to immune evasion due to inactivation of the IFN-γ signaling pathway. Such genetic changes may occur not only in the primary tumor but at any phase of progression: in lymphatic as well as in visceral metastases. Accordingly, it is rational to monitor these changes continuously during disease progression similar to target therapies. Moreover, beside temporal variability, genomic features of tumors such as mutation profiles, as well as the tumor immune microenvironment also show considerable inter- and intratumoral spatial heterogeneity, suggesting the necessity of multiple sampling in biomarker studies.


Subject(s)
Disease Susceptibility , Neoplasms/etiology , Neoplasms/metabolism , Animals , Antigen Presentation/immunology , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Biomarkers, Tumor , Disease Progression , Disease Susceptibility/immunology , Gene Amplification , Genetic Background , Genetic Heterogeneity , Genetic Predisposition to Disease , Humans , Immunogenetics/methods , Mutation , Neoplasm Metastasis , Neoplasm Staging , Neoplasms/pathology , Organ Specificity/genetics , Organ Specificity/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
19.
Magy Onkol ; 63(3): 165-171, 2019 Sep 18.
Article in Hungarian | MEDLINE | ID: mdl-31533135

ABSTRACT

In the immune defense against cancer, cell-mediated adaptive immune response is considered of primary importance, in which T lymphocytes play a key role. Activation, expansion and function of antigen-specific T cells is a multistep process and its outcome depends on the balance of positive and negative regulatory mechanisms controlling each step. Many factors can hamper the development of an efficient antitumor immune response, such as insufficient expression of tumor antigens or of molecules necessary for their processing, inhibitory molecular interactions (e.g. immune checkpoints), immune suppressive factors or suppressor cells. Various therapeutic strategies have been developed in order to increase the efficiency of antitumor immune defense, of which the application of immune checkpoint inhibitors, antibodies blocking the brakes of immune response, is the most widespread. These immunomodulatory antibodies had more favorable effect than traditional treatment modalities on a widening spectrum of tumor types, still the majority of patients do not or only transiently respond. Therefore, great efforts are made to develop rational combinations of immune and other therapies, and to identify resistance mechanisms and biomarkers predicting therapy outcome.


Subject(s)
Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Antigens, Neoplasm/immunology , Humans , Immunologic Factors/immunology
20.
Magy Onkol ; 63(3): 173-182, 2019 Sep 18.
Article in Hungarian | MEDLINE | ID: mdl-31533136

ABSTRACT

Genomic instability is a hallmark of cancer therefore of the metastatic disease as well. High tumor mutation burden is due to deficiencies of the DNA repair systems and leads to immunosensitivity due to generation of neoantigens. However, APOBEC activation of that system, though increases mutation rate, but causes immunoresistance. Deficient antigen presentation due to HLA class I defects is another major cause of immunoresistance. The contemporary immunotherapies may exploit gene amplification of PD-L1 but if the affected chromosome is damaged IFN activation can be lost, again causing immunoresistance. Since these genetic changes can be generated continuously during tumor progression (the entire metastatic process), it would be necessary to monitor them continuously. On the other hand, since tumors are genetically and phenotypically heterogeneous, multiple sampling would be necessary to obtain a more realistic picture of biomarker expressions.


Subject(s)
Disease Progression , Mutation , Neoplasms/genetics , Neoplasms/immunology , Humans , Immunotherapy , Neoplasms/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...