19 research outputs found
Neuroendocrine Tumours: a Literature Review
Neuroendocrine tumours (NETs) are a heterogeneous group of malignant neoplasms with diverse morphology and nomenclature. Well-differentiated NETs were historically termed carcinoid tumours, which entailed abundant confusion and misclassification. Cross body-localised NETs have been described from the central nervous system, respiratory and gastrointestinal tracts, larynx, thyroid, skin, breast and urogenital system. The evidence on NET prevalence is diverse, with selected sources estimating a 0.5% rate among total malignancies diagnosed. Carcinoid syndrome is a known important associate of NETs. Its presence resulting from the amine and peptide hypersecretion often facilitates the NET diagnosis, and curative surgery becomes a treatment of choice, if technically feasible. Adjuvant therapy is ambiguous. When surgery is impractical due to a usually advanced NET at diagnosis, drug therapy is adopted to relief symptoms and control the disease
Hepatocellular Carcinoma: Aetiology and Mechanisms of Development. A Literature Review
Liver cancer remains a global challenge of healthcare, with the incidence growing worldwide. According to various authors, over 1 million patients will be diagnosed with liver cancer each year by 2025. The molecular pathogenesis of HCC varies with respect to genotoxic lesions and aetiologies. Although our understanding of the HCC pathophysiology and drivers tends to improve, it is still distant from translation into clinical practice. About 25 % of HCC cases are associated with variant mutations. HCC pathophysiology is a complex multi-step process. The interaction of various factors underlies the early stages of malignant hepatocyte transformation towards the development of HCC. Overall, about 20β25 % of HCC patients have at least one potential driver mutation. Obesity should also be noted as being associated with a higher risk of HCC and various other cancers. Despite many issues in the HCC pathogenesis being already known, the unresolved questions remain. Modern molecular genetic diagnostics and animal modelling of malignant tumours are expanding our horizons of knowledge in this field
ΠΡΠ΅Π΄ΠΈΠΊΡΠΎΡΡ ΠΏΠ΅ΡΠ²ΠΈΡΠ½ΠΎ-ΠΌΠ½ΠΎΠΆΠ΅ΡΡΠ²Π΅Π½Π½ΡΡ Π·Π»ΠΎΠΊΠ°ΡΠ΅ΡΡΠ²Π΅Π½Π½ΡΡ Π½ΠΎΠ²ΠΎΠΎΠ±ΡΠ°Π·ΠΎΠ²Π°Π½ΠΈΠΉ. ΠΠ±Π·ΠΎΡ Π»ΠΈΡΠ΅ΡΠ°ΡΡΡΡ
The present paper considers the predictors of multiple primary malignancies. The multiple primary malignancies are often induced by genetic predisposition and familial cancer syndromes, environmental carcinogens and bad habits (e.g. tobacco and alcohol abuse), immunodeficiency and infectious diseases, carcinogens occurring as a result of various treatments, etc. Germinal mutations are considered to be one of the causes of primary cancer β they increase the risk of various multiple primary malignancies. Hereditary cancers are characterized by a strong family history, early age of onset and occurrence of multiple primary malignancies. The paper considers various mechanisms of multiple primary malignancies with an emphasis on the effect of carcinogens in carriers of pathogenic genes on cancer development. In proven cases, when patients are exposed to multiple carcinogens, they should be aware of possible late and long-term effects of treatment and their symptoms, as well as secondary malignancies that might occur. When the main predictors of carcinogenesis are manageable, and primary preventive measures can be taken, the hereditary malignancies require postexposure prophylaxis. In addition to specific recommendations for the prevention of primary cancers, it is essential to follow recommendations for the prevention of second primary cancers.Π ΡΡΠ°ΡΡΠ΅ ΡΠ°ΡΡΠΌΠΎΡΡΠ΅Π½Ρ ΠΏΡΠ΅Π΄ΠΈΠΊΡΠΎΡΡ ΠΏΠ΅ΡΠ²ΠΈΡΠ½ΠΎ-ΠΌΠ½ΠΎΠΆΠ΅ΡΡΠ²Π΅Π½Π½ΡΡ
Π·Π»ΠΎΠΊΠ°ΡΠ΅ΡΡΠ²Π΅Π½Π½ΡΡ
Π½ΠΎΠ²ΠΎΠΎΠ±ΡΠ°Π·ΠΎΠ²Π°Π½ΠΈΠΉ. ΠΡΠΈΡΠΈΠ½Π°ΠΌΠΈ ΠΏΠ΅ΡΠ²ΠΈΡΠ½ΠΎ-ΠΌΠ½ΠΎΠΆΠ΅ΡΡΠ²Π΅Π½Π½ΡΡ
Π·Π»ΠΎΠΊΠ°ΡΠ΅ΡΡΠ²Π΅Π½Π½ΡΡ
Π½ΠΎΠ²ΠΎΠΎΠ±ΡΠ°Π·ΠΎΠ²Π°Π½ΠΈΠΉ ΡΠ°ΡΡΠΎ ΡΠ²Π»ΡΡΡΡΡ Π½Π°Π»ΠΈΡΠΈΠ΅ Π³Π΅Π½Π΅ΡΠΈΡΠ΅ΡΠΊΠΎΠΉ ΠΏΡΠ΅Π΄ΡΠ°ΡΠΏΠΎΠ»ΠΎΠΆΠ΅Π½Π½ΠΎΡΡΠΈ ΠΈ ΡΠ΅ΠΌΠ΅ΠΉΠ½ΡΠ΅ ΡΠ°ΠΊΠΎΠ²ΡΠ΅ ΡΠΈΠ½Π΄ΡΠΎΠΌΡ, ΠΊΠ°Π½ΡΠ΅ΡΠΎΠ³Π΅Π½Ρ ΠΎΠΊΡΡΠΆΠ°ΡΡΠ΅ΠΉ ΡΡΠ΅Π΄Ρ ΠΈ Π²ΡΠ΅Π΄Π½ΡΠ΅ ΠΏΡΠΈΠ²ΡΡΠΊΠΈ (Π½Π°ΠΏΡΠΈΠΌΠ΅Ρ, ΡΠ°Π±Π°ΠΊ, ΡΠΏΠΎΡΡΠ΅Π±Π»Π΅Π½ΠΈΠ΅ Π°Π»ΠΊΠΎΠ³ΠΎΠ»Ρ), Π½Π°Π»ΠΈΡΠΈΠ΅ ΠΈΠΌΠΌΡΠ½ΠΎΠ΄Π΅ΡΠΈΡΠΈΡΠ° ΠΈ ΠΈΠ½ΡΠ΅ΠΊΡΠΈΠΎΠ½Π½ΡΠ΅ Π·Π°Π±ΠΎΠ»Π΅Π²Π°Π½ΠΈΡ, ΠΊΠ°Π½ΡΠ΅ΡΠΎΠ³Π΅Π½Ρ, ΡΠΎΡΠΌΠΈΡΡΡΡΠΈΠ΅ΡΡ ΠΏΡΠΈ ΠΏΡΠΈΠΌΠ΅Π½Π΅Π½ΠΈΠΈ ΡΠ°Π·Π»ΠΈΡΠ½ΡΡ
ΠΌΠ΅ΡΠΎΠ΄ΠΎΠ² Π»Π΅ΡΠ΅Π½ΠΈΡ ΠΈ Ρ.Π΄. ΠΠ΄Π½ΠΎΠΉ ΠΈΠ· ΠΏΡΠΈΡΠΈΠ½ Π²ΠΎΠ·Π½ΠΈΠΊΠ½ΠΎΠ²Π΅Π½ΠΈΡ ΠΏΠ΅ΡΠ²ΠΈΡΠ½ΠΎΠ³ΠΎ ΡΠ°ΠΊΠ° ΡΠ²Π»ΡΡΡΡΡ Π³Π΅ΡΠΌΠΈΠ½Π°Π»ΡΠ½ΡΠ΅ ΠΌΡΡΠ°ΡΠΈΠΈ, ΠΊΠΎΡΠΎΡΡΠ΅ ΠΏΠΎΠ²ΡΡΠ°ΡΡ ΡΠΈΡΠΊ ΡΠ°Π·Π²ΠΈΡΠΈΡ ΡΠ°Π·Π»ΠΈΡΠ½ΡΡ
ΠΏΠ΅ΡΠ²ΠΈΡΠ½ΠΎ-ΠΌΠ½ΠΎΠΆΠ΅ΡΡΠ²Π΅Π½Π½ΡΡ
Π·Π»ΠΎΠΊΠ°ΡΠ΅ΡΡΠ²Π΅Π½Π½ΡΡ
Π½ΠΎΠ²ΠΎΠΎΠ±ΡΠ°Π·ΠΎΠ²Π°Π½ΠΈΠΉ. ΠΠ»Ρ Π½Π°ΡΠ»Π΅Π΄ΡΡΠ²Π΅Π½Π½ΠΎΠ³ΠΎ ΡΠ°ΠΊΠ° Ρ
Π°ΡΠ°ΠΊΡΠ΅ΡΠ΅Π½ ΠΎΡΡΠ³ΠΎΡΠ΅Π½Π½ΡΠΉ ΡΠ΅ΠΌΠ΅ΠΉΠ½ΡΠΉ Π°Π½Π°ΠΌΠ½Π΅Π·, ΡΠ°Π½Π½ΠΈΠΉ Π²ΠΎΠ·ΡΠ°ΡΡ Π½Π°ΡΠ°Π»Π° ΠΈ ΠΏΠΎΡΠ²Π»Π΅Π½ΠΈΠ΅ ΠΏΠ΅ΡΠ²ΠΈΡΠ½ΠΎ-ΠΌΠ½ΠΎΠΆΠ΅ΡΡΠ²Π΅Π½Π½ΡΡ
Π·Π»ΠΎΠΊΠ°ΡΠ΅ΡΡΠ²Π΅Π½Π½ΡΡ
Π½ΠΎΠ²ΠΎΠΎΠ±ΡΠ°Π·ΠΎΠ²Π°Π½ΠΈΠΉ. Π ΡΡΠ°ΡΡΠ΅ ΡΠ°ΡΡΠΌΠΎΡΡΠ΅Π½Ρ ΡΠ°Π·Π»ΠΈΡΠ½ΡΠ΅ ΠΌΠ΅Ρ
Π°Π½ΠΈΠ·ΠΌΡ ΡΠ°Π·Π²ΠΈΡΠΈΡ ΠΏΠ΅ΡΠ²ΠΈΡΠ½ΠΎ-ΠΌΠ½ΠΎΠΆΠ΅ΡΡΠ²Π΅Π½Π½ΡΡ
Π·Π»ΠΎΠΊΠ°ΡΠ΅ΡΡΠ²Π΅Π½Π½ΡΡ
Π½ΠΎΠ²ΠΎΠΎΠ±ΡΠ°Π·ΠΎΠ²Π°Π½ΠΈΠΉ, ΠΎΡΠΎΠ±ΡΠΉ Π°ΠΊΡΠ΅Π½Ρ ΡΠ΄Π΅Π»Π°Π½ Π½Π° Π²Π»ΠΈΡΠ½ΠΈΠ΅ ΠΊΠ°Π½ΡΠ΅ΡΠΎΠ³Π΅Π½ΠΎΠ² Ρ Π½ΠΎΡΠΈΡΠ΅Π»Π΅ΠΉ ΠΏΠ°ΡΠΎΠ³Π΅Π½Π½ΡΡ
Π³Π΅Π½ΠΎΠ², ΡΠΏΠΎΡΠΎΠ±ΡΡΠ²ΡΡΡΠΈΡ
ΡΠ°Π·Π²ΠΈΡΠΈΡ ΡΠ°ΠΊΠ°. ΠΡΠΈ Π²ΠΎΠ·Π΄Π΅ΠΉΡΡΠ²ΠΈΠΈ Π½Π° ΠΏΠ°ΡΠΈΠ΅Π½ΡΠ° ΠΌΠ½ΠΎΠΆΠ΅ΡΡΠ²Π° ΠΊΠ°Π½ΡΠ΅ΡΠΎΠ³Π΅Π½ΠΎΠ² Π²Π°ΠΆΠ½ΠΎ, ΡΡΠΎΠ±Ρ ΠΏΠ°ΡΠΈΠ΅Π½ΡΡ Ρ ΡΠΆΠ΅ ΡΡΡΠ°Π½ΠΎΠ²Π»Π΅Π½Π½ΡΠΌ Π΄ΠΈΠ°Π³Π½ΠΎΠ·ΠΎΠΌ ΠΈΠΌΠ΅Π»ΠΈ ΠΈΠ½ΡΠΎΡΠΌΠ°ΡΠΈΡ ΠΎ Π²ΠΎΠ·ΠΌΠΎΠΆΠ½ΡΡ
ΠΏΠΎΠ·Π΄Π½ΠΈΡ
ΠΈ Π΄ΠΎΠ»Π³ΠΎΡΡΠΎΡΠ½ΡΡ
ΠΏΠΎΡΠ»Π΅Π΄ΡΡΠ²ΠΈΡΡ
Π»Π΅ΡΠ΅Π½ΠΈΡ ΠΈ ΠΈΡ
ΡΠΈΠΌΠΏΡΠΎΠΌΠ°Ρ
, Π° ΡΠ°ΠΊΠΆΠ΅ ΠΎ Π²ΠΎΠ·ΠΌΠΎΠΆΠ½ΠΎΠΌ Π²ΠΎΠ·Π½ΠΈΠΊΠ½ΠΎΠ²Π΅Π½ΠΈΠΈ Π²ΡΠΎΡΡΡ
Π·Π»ΠΎΠΊΠ°ΡΠ΅ΡΡΠ²Π΅Π½Π½ΡΡ
Π½ΠΎΠ²ΠΎΠΎΠ±ΡΠ°Π·ΠΎΠ²Π°Π½ΠΈΠΉ. ΠΡΠ»ΠΈ ΠΎΡΠ½ΠΎΠ²Π½ΡΠ΅ ΠΏΡΠ΅Π΄ΠΈΠΊΡΠΎΡΡ ΠΊΠ°Π½ΡΠ΅ΡΠΎΠ³Π΅Π½Π΅Π·Π° ΡΠΏΡΠ°Π²Π»ΡΠ΅ΠΌΡ ΠΈ ΠΌΠΎΠΆΠ½ΠΎ ΠΏΡΠΎΠ²ΠΎΠ΄ΠΈΡΡ ΠΏΠ΅ΡΠ²ΠΈΡΠ½ΡΠ΅ ΠΏΡΠΎΡΠΈΠ»Π°ΠΊΡΠΈΡΠ΅ΡΠΊΠΈΠ΅ ΠΌΠ΅ΡΠΎΠΏΡΠΈΡΡΠΈΡ, ΡΠΎ ΠΏΠΎ ΠΎΡΠ½ΠΎΡΠ΅Π½ΠΈΡ ΠΊ Π½Π°ΡΠ»Π΅Π΄ΡΡΠ²Π΅Π½Π½ΡΠΌ Π·Π»ΠΎΠΊΠ°ΡΠ΅ΡΡΠ²Π΅Π½Π½ΡΠΌ Π½ΠΎΠ²ΠΎΠΎΠ±ΡΠ°Π·ΠΎΠ²Π°Π½ΠΈΡΠΌ Π²Π°ΠΆΠ½ΠΎΠ΅ Π·Π½Π°ΡΠ΅Π½ΠΈΠ΅ ΠΈΠΌΠ΅Π΅Ρ Π²ΡΠΎΡΠΈΡΠ½Π°Ρ ΠΏΡΠΎΡΠΈΠ»Π°ΠΊΡΠΈΠΊΠ°. Π Π΄ΠΎΠΏΠΎΠ»Π½Π΅Π½ΠΈΠ΅ ΠΊ ΡΠ΅ΠΊΠΎΠΌΠ΅Π½Π΄Π°ΡΠΈΡΠΌ, ΠΊΠΎΡΠΎΡΡΠ΅ ΡΠΏΠ΅ΡΠΈΡΠΈΡΠ½Ρ Π΄Π»Ρ ΠΏΡΠΎΡΠΈΠ»Π°ΠΊΡΠΈΠΊΠΈ ΠΏΠ΅ΡΠ²ΠΈΡΠ½ΠΎΠ³ΠΎ ΡΠ°ΠΊΠ°, Π²Π°ΠΆΠ½ΠΎ ΡΠ»Π΅Π΄ΠΎΠ²Π°ΡΡ ΡΠ΅ΠΊΠΎΠΌΠ΅Π½Π΄Π°ΡΠΈΡΠΌ ΠΏΠΎ ΠΏΡΠΎΡΠΈΠ»Π°ΠΊΡΠΈΠΊΠ΅ Π²ΡΠΎΡΠΎΠ³ΠΎ ΠΏΠ΅ΡΠ²ΠΈΡΠ½ΠΎΠ³ΠΎ ΡΠ°ΠΊΠ°
ΠΠ½Π³ΠΈΠ±ΠΈΡΠΎΡΡ ΠΈΠΌΠΌΡΠ½Π½ΡΡ ΠΊΠΎΠ½ΡΡΠΎΠ»ΡΠ½ΡΡ ΡΠΎΡΠ΅ΠΊ ΠΏΡΠΈ ΡΡΠΎΡΠ΅Π»ΠΈΠ°Π»ΡΠ½ΠΎΠΉ ΠΊΠ°ΡΡΠΈΠ½ΠΎΠΌΠ΅ (ΠΎΠ±Π·ΠΎΡ Π»ΠΈΡΠ΅ΡΠ°ΡΡΡΡ)
Bladder cancer is globally considered as one of the most aggressive neoplasms. Traditionally, first-line therapy for metastatic urothelial carcinoma has remained unchanged over the past decades and has been based on combinations of cisplatin. Unfortunately, almost all patients eventually progress and die from bladder cancer, despite the initial response associated with cisplatin-based combinations. Immune checkpoint inhibitors are becoming an increasingly widely used therapeutic option in many solid tumors. In bladder cancer, a high level of programmed death-ligand is determined by rapidly progressive and aggressive tumors and unsatisfactory survival rates. Although checkpoint inhibitors are effective in metastatic urothelial bladder cancer, only a small proportion of treated patients receive a clear benefit, while a large number of patients experience significant side effects and toxicity without improving quality of life or surviving. None of the available biomarkers at this point was associated with response rates. There is evidence of an correlation between PD-L1 expression, the efficacy of immune checkpoint inhibitors, and treatment outcomes in patients with bladder cancer. A major paradigm shift in bladder cancer medicine has followed the FDA approval of avelumab, pembrolizumab, durvalumab, atezolizumab, and nivolumab for the treatment of patients with metastatic urothelial carcinoma previously treated with chemotherapy. Combining classical clinicopathological parameters with data obtained via information technology, together with genomic profiling, could be the future of personalized therapy for bladder cancer.Π Π°ΠΊ ΠΌΠΎΡΠ΅Π²ΠΎΠ³ΠΎ ΠΏΡΠ·ΡΡΡ ΡΡΠΈΡΠ°Π΅ΡΡΡ ΠΎΠ΄Π½ΠΈΠΌ ΠΈΠ· ΡΠ°ΠΌΡΡ
Π°Π³ΡΠ΅ΡΡΠΈΠ²Π½ΡΡ
Π½ΠΎΠ²ΠΎΠΎΠ±ΡΠ°Π·ΠΎΠ²Π°Π½ΠΈΠΉ Π²ΠΎ Π²ΡΠ΅ΠΌ ΠΌΠΈΡΠ΅. Π’Π΅ΡΠ°ΠΏΠΈΡ ΠΏΠ΅ΡΠ²ΠΎΠΉ Π»ΠΈΠ½ΠΈΠΈ ΠΌΠ΅ΡΠ°ΡΡΠ°ΡΠΈΡΠ΅ΡΠΊΠΎΠΉ ΡΡΠΎΡΠ΅Π»ΠΈΠ°Π»ΡΠ½ΠΎΠΉ ΠΊΠ°ΡΡΠΈΠ½ΠΎΠΌΡ ΠΎΡΡΠ°Π²Π°Π»Π°ΡΡ Π½Π΅ΠΈΠ·ΠΌΠ΅Π½Π½ΠΎΠΉ Π½Π° ΠΏΡΠΎΡΡΠΆΠ΅Π½ΠΈΠΈ ΠΏΠΎΡΠ»Π΅Π΄Π½ΠΈΡ
Π΄Π΅ΡΡΡΠΈΠ»Π΅ΡΠΈΠΉ ΠΈ ΠΎΡΠ½ΠΎΠ²ΡΠ²Π°Π»Π°ΡΡ Π½Π° ΠΊΠΎΠΌΠ±ΠΈΠ½Π°ΡΠΈΡΡ
ΡΠΈΡΠΏΠ»Π°ΡΠΈΠ½Π°. Π ΡΠΎΠΆΠ°Π»Π΅Π½ΠΈΡ, ΠΏΠΎΡΡΠΈ Π²ΡΠ΅ ΠΏΠ°ΡΠΈΠ΅Π½ΡΡ Π² ΠΊΠΎΠ½Π΅ΡΠ½ΠΎΠΌ ΡΡΠ΅ΡΠ΅ ΠΏΡΠΎΠ³ΡΠ΅ΡΡΠΈΡΡΡΡ ΠΈ ΡΠΌΠΈΡΠ°ΡΡ ΠΎΡ ΡΠ°ΠΊΠ° ΠΌΠΎΡΠ΅Π²ΠΎΠ³ΠΎ ΠΏΡΠ·ΡΡΡ, Π½Π΅ΡΠΌΠΎΡΡΡ Π½Π° ΠΏΠ΅ΡΠ²ΠΎΠ½Π°ΡΠ°Π»ΡΠ½ΡΠΉ ΠΎΡΠ²Π΅Ρ, ΡΠ²ΡΠ·Π°Π½Π½ΡΠΉ Ρ ΠΊΠΎΠΌΠ±ΠΈΠ½Π°ΡΠΈΡΠΌΠΈ Π½Π° ΠΎΡΠ½ΠΎΠ²Π΅ ΡΠΈΡΠΏΠ»Π°ΡΠΈΠ½Π°. ΠΠ½Π³ΠΈΠ±ΠΈΡΠΎΡΡ ΠΈΠΌΠΌΡΠ½Π½ΡΡ
ΠΊΠΎΠ½ΡΡΠΎΠ»ΡΠ½ΡΡ
ΡΠΎΡΠ΅ΠΊ ΡΡΠ°Π½ΠΎΠ²ΡΡΡΡ Π²ΡΠ΅ Π±ΠΎΠ»Π΅Π΅ ΡΠΈΡΠΎΠΊΠΎ ΠΈΡΠΏΠΎΠ»ΡΠ·ΡΠ΅ΠΌΡΠΌ ΡΠ΅ΡΠ°ΠΏΠ΅Π²ΡΠΈΡΠ΅ΡΠΊΠΈΠΌ Π²Π°ΡΠΈΠ°Π½ΡΠΎΠΌ ΠΏΡΠΈ ΠΌΠ½ΠΎΠ³ΠΈΡ
ΡΠΎΠ»ΠΈΠ΄Π½ΡΡ
ΠΎΠΏΡΡ
ΠΎΠ»ΡΡ
. ΠΡΠΈ ΡΠ°ΠΊΠ΅ ΠΌΠΎΡΠ΅Π²ΠΎΠ³ΠΎ ΠΏΡΠ·ΡΡΡ Π²ΡΡΠΎΠΊΠΈΠΉ ΡΡΠΎΠ²Π΅Π½Ρ Π»ΠΈΠ³Π°Π½Π΄Π° Π·Π°ΠΏΡΠΎΠ³ΡΠ°ΠΌΠΌΠΈΡΠΎΠ²Π°Π½Π½ΠΎΠΉ ΡΠΌΠ΅ΡΡΠΈ ΡΠ²ΡΠ·Π°Π½ Ρ Π±ΡΡΡΡΠΎ ΠΏΡΠΎΠ³ΡΠ΅ΡΡΠΈΡΡΡΡΠΈΠΌΠΈ ΠΈ Π°Π³ΡΠ΅ΡΡΠΈΠ²Π½ΡΠΌΠΈ ΠΎΠΏΡΡ
ΠΎΠ»ΡΠΌΠΈ Ρ Π½Π΅ΡΠ΄ΠΎΠ²Π»Π΅ΡΠ²ΠΎΡΠΈΡΠ΅Π»ΡΠ½ΡΠΌΠΈ ΠΏΠΎΠΊΠ°Π·Π°ΡΠ΅Π»ΡΠΌΠΈ Π²ΡΠΆΠΈΠ²Π°Π΅ΠΌΠΎΡΡΠΈ. Π₯ΠΎΡΡ ΠΈΠ½Π³ΠΈΠ±ΠΈΡΠΎΡΡ ΠΊΠΎΠ½ΡΡΠΎΠ»ΡΠ½ΡΡ
ΡΠΎΡΠ΅ΠΊ ΡΡΡΠ΅ΠΊΡΠΈΠ²Π½Ρ ΠΏΡΠΈ ΠΌΠ΅ΡΠ°ΡΡΠ°ΡΠΈΡΠ΅ΡΠΊΠΎΠΌ ΡΡΠΎΡΠ΅Π»ΠΈΠ°Π»ΡΠ½ΠΎΠΌ ΡΠ°ΠΊΠ΅ ΠΌΠΎΡΠ΅Π²ΠΎΠ³ΠΎ ΠΏΡΠ·ΡΡΡ, Π»ΠΈΡΡ Π½Π΅Π±ΠΎΠ»ΡΡΠ°Ρ ΡΠ°ΡΡΡ ΠΏΡΠΎΠ»Π΅ΡΠ΅Π½Π½ΡΡ
ΠΏΠ°ΡΠΈΠ΅Π½ΡΠΎΠ² ΠΏΠΎΠ»ΡΡΠ°Π΅Ρ ΡΠ²Π½ΡΡ ΠΏΠΎΠ»ΡΠ·Ρ, Π² ΡΠΎ Π²ΡΠ΅ΠΌΡ ΠΊΠ°ΠΊ Π±ΠΎΠ»ΡΡΠΎΠ΅ ΡΠΈΡΠ»ΠΎ ΠΏΠ°ΡΠΈΠ΅Π½ΡΠΎΠ² ΠΏΠΎΠ»ΡΡΠ°ΡΡ Π·Π½Π°ΡΠΈΡΠ΅Π»ΡΠ½ΡΠ΅ ΠΏΠΎΠ±ΠΎΡΠ½ΡΠ΅ ΡΡΡΠ΅ΠΊΡΡ ΠΈ ΡΠΎΠΊΡΠΈΡΠ½ΠΎΡΡΡ Π±Π΅Π· ΡΠ»ΡΡΡΠ΅Π½ΠΈΡ ΠΊΠ°ΡΠ΅ΡΡΠ²Π° ΠΆΠΈΠ·Π½ΠΈ ΠΈΠ»ΠΈ Π²ΡΠΆΠΈΠ²Π°Π΅ΠΌΠΎΡΡΠΈ. ΠΠΈ ΠΎΠ΄ΠΈΠ½ ΠΈΠΌΠ΅ΡΡΠΈΠΉΡΡ Π±ΠΈΠΎΠΌΠ°ΡΠΊΠ΅Ρ Π½Π° ΡΡΠΎΡ ΠΌΠΎΠΌΠ΅Π½Ρ Π½Π΅ Π±ΡΠ» ΡΠ²ΡΠ·Π°Π½ Ρ ΡΠ°ΡΡΠΎΡΠΎΠΉ ΠΎΡΠ²Π΅ΡΠΎΠ². ΠΠΌΠ΅ΡΡΡΡ Π΄Π°Π½Π½ΡΠ΅ ΠΎ ΡΠ²ΡΠ·ΠΈ ΠΌΠ΅ΠΆΠ΄Ρ ΡΠΊΡΠΏΡΠ΅ΡΡΠΈΠ΅ΠΉ PD-L1, ΡΡΡΠ΅ΠΊΡΠΈΠ²Π½ΠΎΡΡΡΡ ΠΈΠ½Π³ΠΈΠ±ΠΈΡΠΎΡΠΎΠ² ΠΈΠΌΠΌΡΠ½Π½ΡΡ
ΠΊΠΎΠ½ΡΡΠΎΠ»ΡΠ½ΡΡ
ΡΠΎΡΠ΅ΠΊ ΠΈ ΡΠ΅Π·ΡΠ»ΡΡΠ°ΡΠ°ΠΌΠΈ Π»Π΅ΡΠ΅Π½ΠΈΡ ΠΏΠ°ΡΠΈΠ΅Π½ΡΠΎΠ² Ρ ΡΠ°ΠΊΠΎΠΌ ΠΌΠΎΡΠ΅Π²ΠΎΠ³ΠΎ ΠΏΡΠ·ΡΡΡ. Π‘Π΅ΡΡΠ΅Π·Π½ΡΠΉ ΡΠ΄Π²ΠΈΠ³ ΠΏΠ°ΡΠ°Π΄ΠΈΠ³ΠΌΡ Π² ΠΌΠ΅Π΄ΠΈΡΠΈΠ½Π΅ ΡΠ°ΠΊΠ° ΠΌΠΎΡΠ΅Π²ΠΎΠ³ΠΎ ΠΏΡΠ·ΡΡΡ Π±ΡΠ» ΡΠ²ΡΠ·Π°Π½ Ρ ΠΎΠ΄ΠΎΠ±ΡΠ΅Π½ΠΈΠ΅ΠΌ FDA Π°Π²Π΅Π»ΡΠΌΠ°Π±Π°, ΠΏΠ΅ΠΌΠ±ΡΠΎΠ»ΠΈΠ·ΡΠΌΠ°Π±Π°, Π΄ΡΡΠ²Π°Π»ΡΠΌΠ°Π±Π°, Π°ΡΠ΅Π·ΠΎΠ»ΠΈΠ·ΡΠΌΠ°Π±Π° ΠΈ Π½ΠΈΠ²ΠΎΠ»ΡΠΌΠ°Π±Π° Π΄Π»Ρ Π»Π΅ΡΠ΅Π½ΠΈΡ ΠΏΠ°ΡΠΈΠ΅Π½ΡΠΎΠ² Ρ ΠΌΠ΅ΡΠ°ΡΡΠ°ΡΠΈΡΠ΅ΡΠΊΠΎΠΉ ΡΡΠΎΡΠ΅Π»ΠΈΠ°Π»ΡΠ½ΠΎΠΉ ΠΊΠ°ΡΡΠΈΠ½ΠΎΠΌΠΎΠΉ, ΡΠ°Π½Π΅Π΅ ΠΏΠΎΠ»ΡΡΠ°Π²ΡΠΈΡ
Ρ
ΠΈΠΌΠΈΠΎΡΠ΅ΡΠ°ΠΏΠΈΡ. Π‘ΠΎΡΠ΅ΡΠ°Π½ΠΈΠ΅ ΠΊΠ»Π°ΡΡΠΈΡΠ΅ΡΠΊΠΈΡ
ΠΊΠ»ΠΈΠ½ΠΈΠΊΠΎ-ΠΏΠ°ΡΠΎΠ»ΠΎΠ³ΠΈΡΠ΅ΡΠΊΠΈΡ
ΠΏΠ°ΡΠ°ΠΌΠ΅ΡΡΠΎΠ² Ρ Π΄Π°Π½Π½ΡΠΌΠΈ, ΠΏΠΎΠ»ΡΡΠ΅Π½Π½ΡΠΌΠΈ Ρ ΠΏΠΎΠΌΠΎΡΡΡ ΠΈΠ½ΡΠΎΡΠΌΠ°ΡΠΈΠΎΠ½Π½ΡΡ
ΡΠ΅Ρ
Π½ΠΎΠ»ΠΎΠ³ΠΈΠΉ, Π²ΠΌΠ΅ΡΡΠ΅ Ρ Π³Π΅Π½ΠΎΠΌΠ½ΡΠΌ ΠΏΡΠΎΡΠΈΠ»ΠΈΡΠΎΠ²Π°Π½ΠΈΠ΅ΠΌ ΠΌΠΎΠΆΠ΅Ρ ΡΡΠ°ΡΡ Π±ΡΠ΄ΡΡΠΈΠΌ ΠΏΠ΅ΡΡΠΎΠ½Π°Π»ΠΈΠ·ΠΈΡΠΎΠ²Π°Π½Π½ΠΎΠΉ ΡΠ΅ΡΠ°ΠΏΠΈΠΈ ΡΠ°ΠΊΠ° ΠΌΠΎΡΠ΅Π²ΠΎΠ³ΠΎ ΠΏΡΠ·ΡΡΡ
ΠΡΠΈΠ±ΡΠ»ΠΈΠ½ Π² ΡΠ΅Π°Π»ΡΠ½ΠΎΠΉ ΠΊΠ»ΠΈΠ½ΠΈΡΠ΅ΡΠΊΠΎΠΉ ΠΏΡΠ°ΠΊΡΠΈΠΊΠ΅ ΠΏΡΠΈ ΠΌΠ΅ΡΠ°ΡΡΠ°ΡΠΈΡΠ΅ΡΠΊΠΎΠΌ ΡΠ°ΠΊΠ΅ ΠΌΠΎΠ»ΠΎΡΠ½ΠΎΠΉ ΠΆΠ΅Π»Π΅Π·Ρ
Breast cancer is the most common cancer in women, both in the Russian Federation and in the United States. Eribulin is a new microtubule inhibitor, used in the USA, Europe and other countries around the world to treat patients with advanced or metastatic breast cancer, resistant to other lines of therapy. Eribulin mesylate is a structurally simplified, synthetic analogue of halichondrin B, a substance derived from a marine spongeΒ Halichondria okadai. The mechanism of action: eribulin inhibits the microtubule dynamics, binds to the plus ends of microtubules and inhibits microtubule growth in interphase cells, without affecting the shortening phase and tubulin sequesters into nonproductive aggregates, leading to G2/M cell-cycle block and eventual apoptotic cell death after prolonged mitotic blockage. A wide variety of patients with a number of comorbidities and lower functional status are treated in actual clinical practice. The reported data suggest that eribulin has demonstrated comparable results to the EMBRACE study for a wider and more diverse cohort of patients. Further studies of eribulin in actual clinical practice will certainly provide new opportunities for the treatment of metastatic breast cancer.Π Π°ΠΊ ΠΌΠΎΠ»ΠΎΡΠ½ΠΎΠΉ ΠΆΠ΅Π»Π΅Π·Ρ ΡΠ²Π»ΡΠ΅ΡΡΡ Π½Π°ΠΈΠ±ΠΎΠ»Π΅Π΅ ΡΠ°ΡΠΏΡΠΎΡΡΡΠ°Π½Π΅Π½Π½ΡΠΌ Π²ΠΈΠ΄ΠΎΠΌ ΡΠ°ΠΊΠ° Ρ ΠΆΠ΅Π½ΡΠΈΠ½ ΠΊΠ°ΠΊ Π² Π ΠΎΡΡΠΈΠΉΡΠΊΠΎΠΉ Π€Π΅Π΄Π΅ΡΠ°ΡΠΈΠΈ, ΡΠ°ΠΊ ΠΈ Π² Π‘Π¨Π. ΠΡΠΈΠ±ΡΠ»ΠΈΠ½, Π½ΠΎΠ²ΡΠΉ ΠΈΠ½Π³ΠΈΠ±ΠΈΡΠΎΡ Π΄ΠΈΠ½Π°ΠΌΠΈΠΊΠΈ ΠΌΠΈΠΊΡΠΎΡΡΡΠ±ΠΎΡΠ΅ΠΊ, ΠΈΡΠΏΠΎΠ»ΡΠ·ΡΠ΅ΡΡΡ Π² Π‘Π¨Π, ΠΠ²ΡΠΎΠΏΠ΅ ΠΈ Π΄ΡΡΠ³ΠΈΡ
ΡΡΡΠ°Π½Π°Ρ
ΠΏΠΎ Π²ΡΠ΅ΠΌΡ ΠΌΠΈΡΡ Π΄Π»Ρ Π»Π΅ΡΠ΅Π½ΠΈΡ ΠΏΠ°ΡΠΈΠ΅Π½ΡΠΎΠΊ Ρ ΠΏΡΠΎΠ³ΡΠ΅ΡΡΠΈΡΡΡΡΠΈΠΌ ΠΈΠ»ΠΈ ΠΌΠ ΠΠ, ΠΊΠΎΡΠΎΡΡΠ΅ ΡΠ΅Π·ΠΈΡΡΠ΅Π½ΡΠ½Ρ Π΄Π»Ρ Π΄ΡΡΠ³ΠΈΡ
Π»ΠΈΠ½ΠΈΠΉ ΡΠ΅ΡΠ°ΠΏΠΈΠΈ. ΠΠ΅Π·ΠΈΠ»Π°Ρ ΡΡΠΈΠ±ΡΠ»ΠΈΠ½Π° ΠΏΡΠ΅Π΄ΡΡΠ°Π²Π»ΡΠ΅Ρ ΡΠΎΠ±ΠΎΠΉ ΡΡΡΡΠΊΡΡΡΠ½ΠΎ ΡΠΏΡΠΎΡΠ΅Π½Π½ΡΠΉ ΡΠΈΠ½ΡΠ΅ΡΠΈΡΠ΅ΡΠΊΠΈΠΉ Π°Π½Π°Π»ΠΎΠ³ Π³Π°Π»ΠΈΡ
ΠΎΠ½Π΄ΡΠΈΠ½Π° Π, Π½Π°ΡΡΡΠ°Π»ΡΠ½ΠΎΠ³ΠΎ ΠΏΡΠΎΠ΄ΡΠΊΡΠ°, Π²ΡΠ΄Π΅Π»Π΅Π½Π½ΠΎΠ³ΠΎ ΠΈΠ· ΠΌΠΎΡΡΠΊΠΎΠΉ Π³ΡΠ±ΠΊΠΈ Halichondria okadai. ΠΠ΅Ρ
Π°Π½ΠΈΠ·ΠΌ Π΄Π΅ΠΉΡΡΠ²ΠΈΡ ΡΡΠΈΠ±ΡΠ»ΠΈΠ½Π° β ΠΏΠΎΠ΄Π°Π²Π»Π΅Π½ΠΈΠ΅ Π΄ΠΈΠ½Π°ΠΌΠΈΠΊΠΈ ΠΌΠΈΠΊΡΠΎΡΡΡΠ±ΠΎΡΠ΅ΠΊ. ΠΠ½ ΡΠ²ΡΠ·ΡΠ²Π°Π΅ΡΡΡ Ρ ΠΏΠ»ΡΡΠΎΠ²ΡΠΌΠΈ ΠΊΠΎΠ½ΡΠ°ΠΌΠΈ ΠΌΠΈΠΊΡΠΎΡΡΡΠ±ΠΎΡΠ΅ΠΊ ΠΈ ΠΏΠΎΠ΄Π°Π²Π»ΡΠ΅Ρ ΡΠΎΡΡ ΠΌΠΈΠΊΡΠΎΡΡΡΠ±ΠΎΡΠ΅ΠΊ Π² ΠΌΠ΅ΠΆΡΠ°Π·Π½ΡΡ
ΠΊΠ»Π΅ΡΠΊΠ°Ρ
, Π½Π΅ Π²Π»ΠΈΡΡ Π½Π° ΡΠ°Π·Ρ ΡΠΊΠΎΡΠΎΡΠ΅Π½ΠΈΡ ΠΈ ΡΠ΅ΠΊΠ²Π΅ΡΡΡΡ ΡΡΠ±ΡΠ»ΠΈΠ½Π° Π² Π½Π΅ΠΏΡΠΎΠ΄ΡΠΊΡΠΈΠ²Π½ΡΠ΅ Π°Π³ΡΠ΅Π³Π°ΡΡ, ΠΏΡΠΈΠ²ΠΎΠ΄ΡΡΠΈΠ΅ ΠΊ G2/M Π±Π»ΠΎΠΊΠΈΡΠΎΠ²ΠΊΠ΅ ΠΊΠ»Π΅ΡΠΎΡΠ½ΠΎΠ³ΠΎ ΡΠΈΠΊΠ»Π° ΠΈ, Π² ΠΊΠΎΠ½Π΅ΡΠ½ΠΎΠΌ ΡΡΠ΅ΡΠ΅, Π°ΠΏΠΎΠΏΡΠΎΠ·Ρ ΠΏΠΎΡΠ»Π΅ Π΄Π»ΠΈΡΠ΅Π»ΡΠ½ΠΎΠΉ ΠΌΠΈΡΠΎΡΠΈΡΠ΅ΡΠΊΠΎΠΉ Π±Π»ΠΎΠΊΠΈΡΠΎΠ²ΠΊΠΈ. Π ΡΠ΅Π°Π»ΡΠ½ΠΎΠΉ ΠΊΠ»ΠΈΠ½ΠΈΡΠ΅ΡΠΊΠΎΠΉ ΠΏΡΠ°ΠΊΡΠΈΠΊΠ΅ Π»Π΅ΡΠ΅Π½ΠΈΠ΅ ΠΏΠΎΠ»ΡΡΠ°ΡΡ ΡΠ°ΠΌΡΠ΅ ΡΠ°Π·Π½ΠΎΠΎΠ±ΡΠ°Π·Π½ΡΠ΅ ΠΏΠ°ΡΠΈΠ΅Π½ΡΠΊΠΈ Ρ ΠΌΠ°ΡΡΠΎΠΉ ΡΠΎΠΏΡΡΡΡΠ²ΡΡΡΠ΅ΠΉ ΠΏΠ°ΡΠΎΠ»ΠΎΠ³ΠΈΠΈ ΠΈ Π±ΠΎΠ»Π΅Π΅ Π½ΠΈΠ·ΠΊΠΈΠΌ ΡΡΠ½ΠΊΡΠΈΠΎΠ½Π°Π»ΡΠ½ΡΠΌ ΡΡΠ°ΡΡΡΠΎΠΌ. ΠΡΠΈΠ²Π΅Π΄Π΅Π½Π½ΡΠ΅ Π΄Π°Π½Π½ΡΠ΅ ΡΠ²ΠΈΠ΄Π΅ΡΠ΅Π»ΡΡΡΠ²ΡΡΡ, ΡΡΠΎ ΡΡΠΈΠ±ΡΠ»ΠΈΠ½ ΠΏΡΠΎΠ΄Π΅ΠΌΠΎΠ½ΡΡΡΠΈΡΠΎΠ²Π°Π» ΡΠΎΠΏΠΎΡΡΠ°Π²ΠΈΠΌΡΠ΅ ΡΠ΅Π·ΡΠ»ΡΡΠ°ΡΡ Ρ ΠΈΡΡΠ»Π΅Π΄ΠΎΠ²Π°Π½ΠΈΠ΅ΠΌ EMBRACE Π΄Π»Ρ Π±ΠΎΠ»Π΅Π΅ ΡΠΈΡΠΎΠΊΠΎΠΉ ΠΈ ΡΠ°Π·Π½ΠΎΠΎΠ±ΡΠ°Π·Π½ΠΎΠΉ ΠΊΠΎΠ³ΠΎΡΡΡ ΠΏΠ°ΡΠΈΠ΅Π½ΡΠΎΠ². ΠΠ°Π»ΡΠ½Π΅ΠΉΡΠΈΠ΅ ΠΈΡΡΠ»Π΅Π΄ΠΎΠ²Π°Π½ΠΈΡ ΡΡΠΈΠ±ΡΠ»ΠΈΠ½Π° Π² ΡΠ΅Π°Π»ΡΠ½ΠΎΠΉ ΠΊΠ»ΠΈΠ½ΠΈΡΠ΅ΡΠΊΠΎΠΉ ΠΏΡΠ°ΠΊΡΠΈΠΊΠ΅, Π±Π΅Π·ΡΡΠ»ΠΎΠ²Π½ΠΎ, ΠΎΡΠΊΡΠΎΡΡ Π½ΠΎΠ²ΡΠ΅ Π²ΠΎΠ·ΠΌΠΎΠΆΠ½ΠΎΡΡΠΈ Π² ΡΠ΅ΡΠ°ΠΏΠΈΠΈ ΠΌΠ΅ΡΠ°ΡΡΠ°ΡΠΈΡΠ΅ΡΠΊΠΎΠ³ΠΎ Π ΠΠ.
ΠΡΠ΅Π·ΠΎΠ»ΠΈΠ·ΡΠΌΠ°Π± ΠΈ Π±Π΅Π²Π°ΡΠΈΠ·ΡΠΌΠ°Π± Ρ ΠΏΠ°ΡΠΈΠ΅Π½ΡΠΎΠ² ΡΒ Π³Π΅ΠΏΠ°ΡΠΎΡΠ΅Π»Π»ΡΠ»ΡΡΠ½ΠΎΠΉ ΠΊΠ°ΡΡΠΈΠ½ΠΎΠΌΠΎΠΉ Π² ΡΠ΅Π°Π»ΡΠ½ΠΎΠΉ ΠΊΠ»ΠΈΠ½ΠΈΡΠ΅ΡΠΊΠΎΠΉ ΠΏΡΠ°ΠΊΡΠΈΠΊΠ΅
Β Β Randomized clinical trials and actual clinical practice differsignificantly. Evidence-based medicine develops new agents referring to, primarily, pharmaceutical findings, preclinical studies and, most importantly, randomized clinical trials. Hepatocellular carcinoma is the most common primary malignancy of the liver, and one of the main causes of fatal outcomes among cancer patients worldwide, including in the Asia-Pacific region, with an estimated 800,000 deaths annually. For more than 10 years, sorafenib, a tyrosine kinase inhibitor, was the only authorized treatment for advanced hepatocellular carcinoma. The next stage in the development of drug therapy for hepatocellular carcinoma involved immune checkpoint inhibitors. The combination of atezolizumab with bevacizumab in the phase III trial (IMbrave150) improved outcomes of advanced hepatocellular carcinoma, such as overall survival and progression-free survival (6.8 versus 4.3). The paper presents the trials of atezolizumab and bevacizumab combination, demonstrates comparable data on the treatment of patients with HCC in real clinical practice and data on the phase III IMbrave150. To further analyze the efficacy of the combination of atezolizumab and bevacizumab, prospective clinical trials should include heterogeneous patient groups.Β Β Π Π°Π½Π΄ΠΎΠΌΠΈΠ·ΠΈΡΠΎΠ²Π°Π½Π½ΡΠ΅ ΠΊΠ»ΠΈΠ½ΠΈΡΠ΅ΡΠΊΠΈΠ΅ ΠΈΡΡΠ»Π΅Π΄ΠΎΠ²Π°Π½ΠΈΡ ΠΈ ΡΠ΅Π°Π»ΡΠ½Π°Ρ ΠΊΠ»ΠΈΠ½ΠΈΡΠ΅ΡΠΊΠ°Ρ ΠΏΡΠ°ΠΊΡΠΈΠΊΠ° ΠΈΠΌΠ΅ΡΡ ΠΌΠ½ΠΎΠ³ΠΎ ΡΠ°Π·Π»ΠΈΡΠΈΠΉ. Π‘ΠΎΠ·Π΄Π°Π½ΠΈΠ΅ Π½ΠΎΠ²ΡΡ
Π»Π΅ΠΊΠ°ΡΡΡΠ²Π΅Π½Π½ΡΡ
ΡΡΠ΅Π΄ΡΡΠ² Π²ΠΎ Π²ΡΠ΅ΠΌΡ Π΄ΠΎΠΊΠ°Π·Π°ΡΠ΅Π»ΡΠ½ΠΎΠΉ ΠΌΠ΅Π΄ΠΈΡΠΈΠ½Ρ ΠΎΡΠ½ΠΎΠ²Π°Π½ΠΎ ΠΏΡΠ΅ΠΆΠ΄Π΅ Π²ΡΠ΅Π³ΠΎ Π½Π° ΡΠ°ΡΠΌΠ°ΡΠ΅Π²ΡΠΈΡΠ΅ΡΠΊΠΈΡ
ΡΠ°Π·ΡΠ°Π±ΠΎΡΠΊΠ°Ρ
, ΠΏΡΠΎΠ²Π΅Π΄Π΅Π½ΠΈΠΈ Π΄ΠΎΠΊΠ»ΠΈΠ½ΠΈΡΠ΅ΡΠΊΠΈΡ
ΠΈΡΡΠ»Π΅Π΄ΠΎΠ²Π°Π½ΠΈΠΉ ΠΈ, ΡΠ°ΠΌΠΎΠ΅ ΠΎΡΠ½ΠΎΠ²Π½ΠΎΠ΅, ΡΠ°Π½Π΄ΠΎΠΌΠΈΠ·ΠΈΡΠΎΠ²Π°Π½Π½ΡΡ
ΠΊΠ»ΠΈΠ½ΠΈΡΠ΅ΡΠΊΠΈΡ
ΠΈΡΡΠ»Π΅Π΄ΠΎΠ²Π°Π½ΠΈΡΡ
. ΠΠ΅ΠΏΠ°ΡΠΎΡΠ΅Π»Π»ΡΠ»ΡΡΠ½Π°Ρ ΠΊΠ°ΡΡΠΈΠ½ΠΎΠΌΠ° ΡΠ²Π»ΡΠ΅ΡΡΡ Π½Π°ΠΈΠ±ΠΎΠ»Π΅Π΅ ΡΠ°ΡΠΏΡΠΎΡΡΡΠ°Π½Π΅Π½Π½ΠΎΠΉ ΠΏΠ΅ΡΠ²ΠΈΡΠ½ΠΎΠΉ Π·Π»ΠΎΠΊΠ°ΡΠ΅ΡΡΠ²Π΅Π½Π½ΠΎΠΉ ΠΎΠΏΡΡ
ΠΎΠ»ΡΡ ΠΏΠ΅ΡΠ΅Π½ΠΈ ΠΈ ΠΎΠ΄Π½ΠΎΠΉ ΠΈΠ· Π²Π΅Π΄ΡΡΠΈΡ
ΠΏΡΠΈΡΠΈΠ½ ΡΠΌΠ΅ΡΡΠ½ΠΎΡΡΠΈ ΡΡΠ΅Π΄ΠΈ ΠΎΠ½ΠΊΠΎΠ»ΠΎΠ³ΠΈΡΠ΅ΡΠΊΠΈΡ
ΠΏΠ°ΡΠΈΠ΅Π½ΡΠΎΠ² Π²ΠΎ Π²ΡΠ΅ΠΌ ΠΌΠΈΡΠ΅ Ρ ΠΏΡΠΈΠΌΠ΅ΡΠ½ΡΠΌ ΠΊΠΎΠ»ΠΈΡΠ΅ΡΡΠ²ΠΎΠΌ 800 000 ΡΠΌΠ΅ΡΡΠ΅ΠΉ Π΅ΠΆΠ΅Π³ΠΎΠ΄Π½ΠΎ. ΠΠΎΠ»Π΅Π΅ 10 Π»Π΅Ρ ΡΠΈΡΠΎΠ·ΠΈΠ½ΠΊΠΈΠ½Π°Π·Π½ΡΠΉ ΠΈΠ½Π³ΠΈΠ±ΠΈΡΠΎΡ ΡΠΎΡΠ°ΡΠ΅Π½ΠΈΠ± Π±ΡΠ» Π΅Π΄ΠΈΠ½ΡΡΠ²Π΅Π½Π½ΡΠΌ Π·Π°ΡΠ΅Π³ΠΈΡΡΡΠΈΡΠΎΠ²Π°Π½Π½ΡΠΌ ΡΡΠ΅Π΄ΡΡΠ²ΠΎΠΌ Π»Π΅ΡΠ΅Π½ΠΈΡ ΡΠ°ΡΠΏΡΠΎΡΡΡΠ°Π½Π΅Π½Π½ΠΎΠΉ Π³Π΅ΠΏΠ°ΡΠΎΡΠ΅Π»Π»ΡΠ»ΡΡΠ½ΠΎΠΉ ΠΊΠ°ΡΡΠΈΠ½ΠΎΠΌΡ. Π‘Π»Π΅Π΄ΡΡΡΠΈΠΌ ΡΡΠ°ΠΏΠΎΠΌ ΡΠ°Π·Π²ΠΈΡΠΈΡ Π»Π΅ΠΊΠ°ΡΡΡΠ²Π΅Π½Π½ΠΎΠΉ ΡΠ΅ΡΠ°ΠΏΠΈΠΈ Π³Π΅ΠΏΠ°ΡΠΎΡΠ΅Π»Π»ΡΠ»ΡΡΠ½ΠΎΠΉ ΠΊΠ°ΡΡΠΈΠ½ΠΎΠΌΡ ΡΠ²ΠΈΠ»Π°ΡΡ ΡΠ΅ΡΠ°ΠΏΠΈΡ ΠΈΠ½Π³ΠΈΠ±ΠΈΡΠΎΡΠ°ΠΌΠΈ ΠΊΠΎΠ½ΡΡΠΎΠ»ΡΠ½ΡΡ
ΡΠΎΡΠ΅ΠΊ ΠΈΠΌΠΌΡΠ½ΠΈΡΠ΅ΡΠ°. ΠΠΎΠΌΠ±ΠΈΠ½Π°ΡΠΈΡ Π°ΡΠ΅Π·ΠΎΠ»ΠΈΠ·ΡΠΌΠ°Π±Π° Ρ Π±Π΅Π²Π°ΡΠΈΠ·ΡΠΌΠ°Π±ΠΎΠΌ Π² ΠΈΡΡΠ»Π΅Π΄ΠΎΠ²Π°Π½ΠΈΠΈ III ΡΠ°Π·Ρ (IMbrave150) ΡΠ»ΡΡΡΠΈΠ»Π° ΡΠ΅Π·ΡΠ»ΡΡΠ°ΡΡ Π»Π΅ΡΠ΅Π½ΠΈΡ ΡΠ°ΡΠΏΡΠΎΡΡΡΠ°Π½Π΅Π½Π½ΠΎΠΉ Π³Π΅ΠΏΠ°ΡΠΎΡΠ΅Π»Π»ΡΠ»ΡΡΠ½ΠΎΠΉ ΠΊΠ°ΡΡΠΈΠ½ΠΎΠΌΡ, ΡΠ°ΠΊΠΈΠ΅ ΠΊΠ°ΠΊ ΠΎΠ±ΡΠ°Ρ Π²ΡΠΆΠΈΠ²Π°Π΅ΠΌΠΎΡΡΡ ΠΈ Π²ΡΠΆΠΈΠ²Π°Π΅ΠΌΠΎΡΡΡ Π±Π΅Π· ΠΏΡΠΎΠ³ΡΠ΅ΡΡΠΈΡΠΎΠ²Π°Π½ΠΈΡ (6,8 ΠΏΡΠΎΡΠΈΠ² 4,3). ΠΡΠΈΠ²Π΅Π΄Π΅Π½Π½ΡΠ΅ Π² ΡΡΠ°ΡΡΠ΅ ΠΈΡΡΠ»Π΅Π΄ΠΎΠ²Π°Π½ΠΈΡ ΠΊΠΎΠΌΠ±ΠΈΠ½Π°ΡΠΈΠΈ Π°ΡΠ΅Π·ΠΎΠ»ΠΈΠ·ΡΠΌΠ°Π±Π° ΠΈ Π±Π΅Π²Π°ΡΠΈΠ·ΡΠΌΠ°Π±Π° Π΄Π΅ΠΌΠΎΠ½ΡΡΡΠΈΡΡΡΡ ΡΠΎΠΏΠΎΡΡΠ°Π²ΠΈΠΌΡΠ΅ Π΄Π°Π½Π½ΡΠ΅ ΠΏΡΠΈ Π»Π΅ΡΠ΅Π½ΠΈΠΈ ΠΏΠ°ΡΠΈΠ΅Π½ΡΠΎΠ² Ρ ΠΠ¦Π Π² ΡΠ΅Π°Π»ΡΠ½ΠΎΠΉ ΠΊΠ»ΠΈΠ½ΠΈΡΠ΅ΡΠΊΠΎΠΉ ΠΏΡΠ°ΠΊΡΠΈΠΊΠ΅ ΠΈ Π² ΠΈΡΡΠ»Π΅Π΄ΠΎΠ²Π°Π½ΠΈΠΈ III ΡΠ°Π·Ρ IMbrave150. ΠΡΠΎΡΠΏΠ΅ΠΊΡΠΈΠ²Π½ΡΠ΅ ΠΊΠ»ΠΈΠ½ΠΈΡΠ΅ΡΠΊΠΈΠ΅ ΠΈΡΠΏΡΡΠ°Π½ΠΈΡ, ΠΊΠΎΡΠΎΡΡΠ΅ Π²ΠΊΠ»ΡΡΠ°ΡΡ ΡΠ°Π·Π½ΠΎΡΠΎΠ΄Π½ΡΠ΅ Π³ΡΡΠΏΠΏΡ ΠΏΠ°ΡΠΈΠ΅Π½ΡΠΎΠ², Π½Π΅ΠΎΠ±Ρ
ΠΎΠ΄ΠΈΠΌΡ Π΄Π»Ρ Π΄Π°Π»ΡΠ½Π΅ΠΉΡΠ΅Π³ΠΎ Π°Π½Π°Π»ΠΈΠ·Π° ΡΡΡΠ΅ΠΊΡΠΈΠ²Π½ΠΎΡΡΠΈ ΠΊΠΎΠΌΠ±ΠΈΠ½Π°ΡΠΈΠΈ Π°ΡΠ΅Π·ΠΎΠ»ΠΈΠ·ΡΠΌΠ°Π±Π° ΠΈ Π±Π΅Π²Π°ΡΠΈΠ·ΡΠΌΠ°Π±Π°
ΠΠ»ΠΈΠ½ΠΈΡΠ΅ΡΠΊΠΈΠΉ ΡΠ»ΡΡΠ°ΠΉ Π»Π΅ΡΠ΅Π½ΠΈΡ ΠΌΠ΅ΡΠ°ΡΡΠ°ΡΠΈΡΠ΅ΡΠΊΠΎΠ³ΠΎ ΠΏΠΎΡΠ΅ΡΠ½ΠΎΠΊΠ»Π΅ΡΠΎΡΠ½ΠΎΠ³ΠΎ ΡΠ°ΠΊΠ° ΠΊΠΎΠΌΠ±ΠΈΠ½Π°ΡΠΈΠ΅ΠΉ Π½ΠΈΠ²ΠΎΠ»ΡΠΌΠ°Π±Π° ΡΒ ΠΊΠ°Π±ΠΎΠ·Π°Π½ΡΠΈΠ½ΠΈΠ±ΠΎΠΌ Π²Β ΡΡΡΠΈΠ½Π½ΠΎΠΉ ΠΊΠ»ΠΈΠ½ΠΈΡΠ΅ΡΠΊΠΎΠΉ ΠΏΡΠ°ΠΊΡΠΈΠΊΠ΅
According to GLOBOCAN, there were about 18 million new cases of cancer and 9.6 million deaths from malignancies worldwide in 2018. Renal cell carcinoma is a malignant tumor characterized by the loss of the VHL gene, which leads to increased angiogenesis. The potential of immuno-oncology and anti-angiogenic drugs has significantly improved outcomes for patients with metastatic renal cell carcinoma. The phase III CheckMate 9ER study compared the efficacy and safety of nivolumab plus cabozantinib versus sunitinib in the first-line treatment of patients with metastatic clear cell renal cell carcinoma. The advantages of nivolumab plus cabozantinib over sunitinib in terms of progression-free survival, overall survival, and objective response rate were generally similar across subgroups based on IMDC risk, PD-L1 expression, and the presence or absence of bone metastases. We present a case report of metastatic renal cell carcinoma. The patient has been on cabozantinib plus nivolumab therapy for 12 months, with a partial response achieved. Treatment was well tolerated; the profile of adverse events was consistent with that in the clinical study.ΠΠΎ Π΄Π°Π½Π½ΡΠΌ GLOBOCAN, Π² 2018 Π³ΠΎΠ΄Ρ Π·Π°ΡΠ΅Π³ΠΈΡΡΡΠΈΡΠΎΠ²Π°Π½ΠΎ ΠΎΠΊΠΎΠ»ΠΎ 18 ΠΌΠΈΠ»Π»ΠΈΠΎΠ½ΠΎΠ² Π½ΠΎΠ²ΡΡ
ΡΠ»ΡΡΠ°Π΅Π² ΡΠ°ΠΊΠ° ΠΈ 9,6 ΠΌΠΈΠ»Π»ΠΈΠΎΠ½ΠΎΠ² ΡΠΌΠ΅ΡΡΠ΅ΠΉ ΠΎΡ Π·Π»ΠΎΠΊΠ°ΡΠ΅ΡΡΠ²Π΅Π½Π½ΡΡ
Π½ΠΎΠ²ΠΎΠΎΠ±ΡΠ°Π·ΠΎΠ²Π°Π½ΠΈΠΉ Π²ΠΎ Π²ΡΠ΅ΠΌ ΠΌΠΈΡΠ΅. ΠΠΎΡΠ΅ΡΠ½ΠΎ-ΠΊΠ»Π΅ΡΠΎΡΠ½ΡΠΉ ΡΠ°ΠΊ ΠΏΡΠ΅Π΄ΡΡΠ°Π²Π»ΡΠ΅Ρ ΡΠΎΠ±ΠΎΠΉ ΠΎΠΏΡΡ
ΠΎΠ»Ρ, Ρ
Π°ΡΠ°ΠΊΡΠ΅ΡΠΈΠ·ΡΡΡΡΡΡΡΡ ΠΏΠΎΡΠ΅ΡΠ΅ΠΉ Π³Π΅Π½Π° VHL, ΠΈ ΡΡΠ° ΠΏΠΎΡΠ΅ΡΡ ΠΏΡΠΈΠ²ΠΎΠ΄ΠΈΡ ΠΊ ΡΡΠΈΠ»Π΅Π½ΠΈΡ Π°Π½Π³ΠΈΠΎΠ³Π΅Π½Π΅Π·Π°. ΠΠΎΠ·ΠΌΠΎΠΆΠ½ΠΎΡΡΠΈ ΠΏΡΠΈΠΌΠ΅Π½Π΅Π½ΠΈΡ ΠΈΠΌΠΌΡΠ½ΠΎΠΎΠ½ΠΊΠΎΠ»ΠΎΠ³ΠΈΡΠ΅ΡΠΊΠΈΡ
ΠΈ Π°Π½ΡΠΈΠ°Π½Π³ΠΈΠΎΠ³Π΅Π½Π½ΡΡ
ΠΏΡΠ΅ΠΏΠ°ΡΠ°ΡΠΎΠ² Π·Π½Π°ΡΠΈΡΠ΅Π»ΡΠ½ΠΎ ΡΠ»ΡΡΡΠΈΠ»ΠΈ ΡΠ΅Π·ΡΠ»ΡΡΠ°ΡΡ Π»Π΅ΡΠ΅Π½ΠΈΡ ΠΏΠ°ΡΠΈΠ΅Π½ΡΠΎΠ² Ρ ΠΌΠ΅ΡΠ°ΡΡΠ°ΡΠΈΡΠ΅ΡΠΊΠΈΠΌ ΠΏΠΎΡΠ΅ΡΠ½ΠΎ-ΠΊΠ»Π΅ΡΠΎΡΠ½ΡΠΉ ΡΠ°ΠΊΠΎΠΌ. ΠΡΡΠ»Π΅Π΄ΠΎΠ²Π°Π½ΠΈΠ΅ III ΡΠ°Π·Ρ CheckMate 9ER ΠΏΠΎΡΠ²ΡΡΠ΅Π½ΠΎ ΡΡΠ°Π²Π½Π΅Π½ΠΈΡ ΡΡΡΠ΅ΠΊΡΠΈΠ²Π½ΠΎΡΡΠΈ ΠΈ Π±Π΅Π·ΠΎΠΏΠ°ΡΠ½ΠΎΡΡΠΈ ΠΊΠΎΠΌΠ±ΠΈΠ½Π°ΡΠΈΠΈ Π½ΠΈΠ²ΠΎΠ»ΡΠΌΠ°Π±Π° Ρ ΠΊΠ°Π±ΠΎΠ·Π°Π½ΡΠΈΠ½ΠΈΠ±ΠΎΠΌ ΠΈ ΡΡΠ½ΠΈΡΠΈΠ½ΠΈΠ±Π° Π² ΡΠ΅ΡΠ°ΠΏΠΈΠΈ ΠΏΠ΅ΡΠ²ΠΎΠΉ Π»ΠΈΠ½ΠΈΠΈ ΠΏΠ°ΡΠΈΠ΅Π½ΡΠΎΠ² ΡΠΎ ΡΠ²Π΅ΡΠ»ΠΎΠΊΠ»Π΅ΡΠΎΡΠ½ΡΠΌ ΠΌΠ΅ΡΠ°ΡΡΠ°ΡΠΈΡΠ΅ΡΠΊΠΈΠΌ ΠΏΠΎΡΠ΅ΡΠ½ΠΎ-ΠΊΠ»Π΅ΡΠΎΡΠ½ΡΠΌ ΡΠ°ΠΊΠΎΠΌ. ΠΡΠ΅ΠΈΠΌΡΡΠ΅ΡΡΠ²Π° ΠΊΠΎΠΌΠ±ΠΈΠ½Π°ΡΠΈΠΈ Π½ΠΈΠ²ΠΎΠ»ΡΠΌΠ°Π±Π° ΠΈ ΠΊΠ°Π±ΠΎΠ·Π°Π½ΡΠΈΠ½ΠΈΠ±Π° Π² ΡΡΠ°Π²Π½Π΅Π½ΠΈΠΈ Ρ ΡΡΠ½ΠΈΡΠΈΠ½ΠΈΠ±ΠΎΠΌ Π² ΠΎΡΠ½ΠΎΡΠ΅Π½ΠΈΠΈ ΠΎΡΡΡΡΡΡΠ²ΠΈΡ ΠΏΡΠΎΠ³ΡΠ΅ΡΡΠΈΡΠΎΠ²Π°Π½ΠΈΡ, ΠΎΠ±ΡΠ΅ΠΉ Π²ΡΠΆΠΈΠ²Π°Π΅ΠΌΠΎΡΡΠΈ ΠΈ ΡΠ°ΡΡΠΎΡΡ ΠΎΠ±ΡΠ΅ΠΊΡΠΈΠ²Π½ΡΡ
ΠΎΡΠ²Π΅ΡΠΎΠ² Π² ΡΠ΅Π»ΠΎΠΌ Π±ΡΠ»ΠΈ ΠΎΠ΄ΠΈΠ½Π°ΠΊΠΎΠ²ΡΠΌΠΈ Π² ΠΏΠΎΠ΄Π³ΡΡΠΏΠΏΠ°Ρ
, Π²ΠΊΠ»ΡΡΠ°Ρ Π³ΡΡΠΏΠΏΡ ΡΠΈΡΠΊΠ° ΠΏΠΎ IMDC, ΡΠΊΡΠΏΡΠ΅ΡΡΠΈΡ ΠΎΠΏΡΡ
ΠΎΠ»Π΅Π²ΠΎΠ³ΠΎ PD-L1 ΠΈ Π½Π°Π»ΠΈΡΠΈΠ΅ ΠΈΠ»ΠΈ ΠΎΡΡΡΡΡΡΠ²ΠΈΠ΅ ΠΌΠ΅ΡΠ°ΡΡΠ°ΡΠΈΡΠ΅ΡΠΊΠΎΠ³ΠΎ ΠΏΠΎΡΠ°ΠΆΠ΅Π½ΠΈΡ ΠΊΠΎΡΡΠ½ΠΎΠΉ ΡΠΊΠ°Π½ΠΈ. ΠΡΠΈΠ²Π΅Π΄Π΅Π½ΠΎ ΠΊΠ»ΠΈΠ½ΠΈΡΠ΅ΡΠΊΠΎΠ΅ Π½Π°Π±Π»ΡΠ΄Π΅Π½ΠΈΠ΅ Π»Π΅ΡΠ΅Π½ΠΈΡ ΠΏΠ°ΡΠΈΠ΅Π½ΡΠ° Ρ ΠΌΠ΅ΡΠ°ΡΡΠ°ΡΠΈΡΠ΅ΡΠΊΠΈΠΌ ΠΏΠΎΡΠ΅ΡΠ½ΠΎ-ΠΊΠ»Π΅ΡΠΎΡΠ½ΡΠΌ ΡΠ°ΠΊΠΎΠΌ. Π Π½Π°ΡΡΠΎΡΡΠ΅Π΅ Π²ΡΠ΅ΠΌΡ ΠΏΠ°ΡΠΈΠ΅Π½ΡΡ Π½Π° ΠΏΡΠΎΡΡΠΆΠ΅Π½ΠΈΠΈ 12 ΠΌΠ΅ΡΡΡΠ΅Π² ΠΏΡΠΎΠ΄ΠΎΠ»ΠΆΠ°Π΅ΡΡΡ ΡΠ΅ΡΠ°ΠΏΠΈΡ ΠΊΠ°Π±ΠΎΠ·Π°Π½ΡΠΈΠ½ΠΈΠ±ΠΎΠΌ ΠΈ Π½ΠΈΠ²ΠΎΠ»ΡΠΌΠ°Π±ΠΎΠΌ, ΡΠΎΡ
ΡΠ°Π½ΡΠ΅ΡΡΡ ΡΠ°ΡΡΠΈΡΠ½ΡΠΉ ΠΎΡΠ²Π΅Ρ. ΠΠ΅ΡΠ΅Π½ΠΎΡΠΈΠΌΠΎΡΡΡ ΡΠ΅ΡΠ°ΠΏΠΈΠΈ ΡΠ΄ΠΎΠ²Π»Π΅ΡΠ²ΠΎΡΠΈΡΠ΅Π»ΡΠ½Π°Ρ, ΡΠΏΠ΅ΠΊΡΡ Π½Π΅ΠΆΠ΅Π»Π°ΡΠ΅Π»ΡΠ½ΡΡ
ΡΠ²Π»Π΅Π½ΠΈΠΉ ΡΠΎΠΎΡΠ²Π΅ΡΡΡΠ²ΡΠ΅Ρ ΡΠ°ΠΊΠΎΠ²ΠΎΠΌΡ Π² ΠΊΠ»ΠΈΠ½ΠΈΡΠ΅ΡΠΊΠΎΠΌ ΠΈΡΡΠ»Π΅Π΄ΠΎΠ²Π°Π½ΠΈΠΈ
Π‘Π»ΡΡΠ°ΠΉ ΠΏΠ΅ΡΠ²ΠΈΡΠ½ΠΎ ΠΌΠ½ΠΎΠΆΠ΅ΡΡΠ²Π΅Π½Π½ΠΎΠ³ΠΎ ΡΠΈΠ½Ρ ΡΠΎΠ½Π½ΠΎΠ³ΠΎ ΡΠ°Π΄ΠΈΠΎΠΈΠ½Π΄ΡΡΠΈΡΠΎΠ²Π°Π½Π½ΠΎΠ³ΠΎ ΡΠ°ΠΊΠ° Π² ΠΊΠ»ΠΈΠ½ΠΈΡΠ΅ΡΠΊΠΎΠΉ ΠΏΡΠ°ΠΊΡΠΈΠΊΠ΅
There is an increase in the number of patients with secondary malignant neoplasms. In addition, in some subgroups after radical cancer treatment, the lifetime risk of developing secondary malignant neoplasms can be as high as 33β%. Secondary malignancies remain an important cause of death in patients who have received radical cancer treatment. The presented clinical case demonstrates the risk of developing primary multiple malignant neoplasms after radiation therapy and chemotherapy. A 39-year-old patient with diffuse large B-cell non-Hodgkin lymphoma underwent definitive treatment including chemo-and radiotherapy. Ten years later, the patient developed the induced multiple malignant tumors: a malignant neoplasm of the heart β myofibrosarcoma of the right ventricle with invasion of the anterior wall of the right ventricle Stage IIIB G2T3N0M0, left breast cancer Stage IIIA T3N2M0. The choice of treatment tactics for this category of patients remains particularly difficult.Π ΡΡΡΡΠΊΡΡΡΠ΅ Π·Π°Π±ΠΎΠ»Π΅Π²Π°Π΅ΠΌΠΎΡΡΠΈ Π½Π°Π±Π»ΡΠ΄Π°Π΅ΡΡΡ ΡΠΎΡΡ ΡΠΈΡΠ»Π° ΠΏΠ°ΡΠΈΠ΅Π½ΡΠΎΠ² Ρ Π²ΡΠΎΡΠΈΡΠ½ΡΠΌΠΈ Π·Π»ΠΎΠΊΠ°ΡΠ΅ΡΡΠ²Π΅Π½Π½ΡΠΌΠΈ Π½ΠΎΠ²ΠΎΠΎΠ±ΡΠ°Π·ΠΎΠ²Π°Π½ΠΈΡΠΌΠΈ. ΠΡΠΎΠΌΠ΅ ΡΠΎΠ³ΠΎ, Π² ΠΎΡΠ΄Π΅Π»ΡΠ½ΡΡ
ΠΏΠΎΠ΄Π³ΡΡΠΏΠΏΠ°Ρ
ΠΏΠΎΡΠ»Π΅ ΡΠ°Π΄ΠΈΠΊΠ°Π»ΡΠ½ΠΎΠ³ΠΎ Π»Π΅ΡΠ΅Π½ΠΈΡ ΡΠ°ΠΊΠ° ΠΏΠΎΠΆΠΈΠ·Π½Π΅Π½Π½ΡΠΉ ΡΠΈΡΠΊ ΡΠ°Π·Π²ΠΈΡΠΈΡ Π²ΡΠΎΡΠΈΡΠ½ΡΡ
Π·Π»ΠΎΠΊΠ°ΡΠ΅ΡΡΠ²Π΅Π½Π½ΡΡ
Π½ΠΎΠ²ΠΎΠΎΠ±ΡΠ°Π·ΠΎΠ²Π°Π½ΠΈΠΉ ΠΌΠΎΠΆΠ΅Ρ Π΄ΠΎΡΡΠΈΠ³Π°ΡΡ 33 %. ΠΡΠΎΡΠΈΡΠ½ΡΠ΅ Π·Π»ΠΎΠΊΠ°ΡΠ΅ΡΡΠ²Π΅Π½Π½ΡΠ΅ Π½ΠΎΠ²ΠΎΠΎΠ±ΡΠ°Π·ΠΎΠ²Π°Π½ΠΈΡ ΠΎΡΡΠ°ΡΡΡΡ Π²Π°ΠΆΠ½ΠΎΠΉ ΠΏΡΠΈΡΠΈΠ½ΠΎΠΉ ΡΠΌΠ΅ΡΡΠΈ ΠΏΠ°ΡΠΈΠ΅Π½ΡΠΎΠ², ΠΏΠΎΠ»ΡΡΠΈΠ²ΡΠΈΡ
ΡΠ°Π΄ΠΈΠΊΠ°Π»ΡΠ½ΠΎΠ΅ Π»Π΅ΡΠ΅Π½ΠΈΠ΅ ΠΏΠΎ ΠΏΠΎΠ²ΠΎΠ΄Ρ ΡΠ°ΠΊΠ°. ΠΡΠ΅Π΄ΡΡΠ°Π²Π»Π΅Π½Π½ΡΠΉ ΠΊΠ»ΠΈΠ½ΠΈΡΠ΅ΡΠΊΠΈΠΉ ΡΠ»ΡΡΠ°ΠΉ Π΄Π΅ΠΌΠΎΠ½ΡΡΡΠΈΡΡΠ΅Ρ ΡΠ°Π·Π²ΠΈΡΠΈΠ΅ ΠΏΠ΅ΡΠ²ΠΈΡΠ½ΠΎ-ΠΌΠ½ΠΎΠΆΠ΅ΡΡΠ²Π΅Π½Π½ΡΡ
Π·Π»ΠΎΠΊΠ°ΡΠ΅ΡΡΠ²Π΅Π½Π½ΡΡ
Π½ΠΎΠ²ΠΎΠΎΠ±ΡΠ°Π·ΠΎΠ²Π°Π½ΠΈΠΉ ΠΏΠΎΡΠ»Π΅ Π»ΡΡΠ΅Π²ΠΎΠΉ ΡΠ΅ΡΠ°ΠΏΠΈΠΈ ΠΈ Ρ
ΠΈΠΌΠΈΠΎΡΠ΅ΡΠ°ΠΏΠΈΠΈ. ΠΠ°ΡΠΈΠ΅Π½ΡΠΊΠ° Π² 39 Π»Π΅Ρ ΠΏΠΎ ΠΏΠΎΠ²ΠΎΠ΄Ρ Π΄ΠΈΡΡΡΠ·Π½ΠΎΠΉ ΠΊΡΡΠΏΠ½ΠΎΠΊΠ»Π΅ΡΠΎΡΠ½ΠΎΠΉ Π-ΠΊΠ»Π΅ΡΠΎΡΠ½ΠΎΠΉ Π½Π΅Ρ
ΠΎΠ΄ΠΆΡΠΊΠΈΠ½ΡΠΊΠΎΠΉ Π»ΠΈΠΌΡΠΎΠΌΡ ΠΏΡΠΎΡΠ»Π° ΡΠ°Π΄ΠΈΠΊΠ°Π»ΡΠ½ΡΠΉ ΠΊΡΡΡ Π»Π΅ΡΠ΅Π½ΠΈΡ. Π§Π΅ΡΠ΅Π· 10 Π»Π΅Ρ Ρ ΠΏΠ°ΡΠΈΠ΅Π½ΡΠΊΠΈ Π±ΡΠ»ΠΈ Π²ΡΡΠ²Π»Π΅Π½Ρ ΡΠ°Π΄ΠΈΠΎΠΈΠ½Π΄ΡΡΠΈΡΠΎΠ²Π°Π½Π½ΡΠ΅ ΠΏΠ΅ΡΠ²ΠΈΡΠ½ΠΎ ΠΌΠ½ΠΎΠΆΠ΅ΡΡΠ²Π΅Π½Π½ΡΠ΅ ΠΎΠΏΡΡ
ΠΎΠ»ΠΈ: Π·Π»ΠΎΠΊΠ°ΡΠ΅ΡΡΠ²Π΅Π½Π½Π°Ρ ΠΌΠΈΠΎΡΠΈΠ±ΡΠΎΡΠ°ΡΠΊΠΎΠΌΠ° ΠΏΡΠ°Π²ΠΎΠ³ΠΎ ΠΆΠ΅Π»ΡΠ΄ΠΎΡΠΊΠ° ΡΠ΅ΡΠ΄ΡΠ° IIIB ΡΡΠ°Π΄ΠΈΠΈ β G2T3N0M0, ΡΠ°ΠΊ Π»Π΅Π²ΠΎΠΉ ΠΌΠΎΠ»ΠΎΡΠ½ΠΎΠΉ ΠΆΠ΅Π»Π΅Π·Ρ IIIA ΡΡΠ°Π΄ΠΈΠΈ β T3N2M0. ΠΠΏΡΠ΅Π΄Π΅Π»Π΅Π½ΠΈΠ΅ ΡΠ°ΠΊΡΠΈΠΊΠΈ Π»Π΅ΡΠ΅Π½ΠΈΡ Π΄Π»Ρ Π΄Π°Π½Π½ΠΎΠΉ ΠΊΠ°ΡΠ΅Π³ΠΎΡΠΈΠΈ Π±ΠΎΠ»ΡΠ½ΡΡ
ΠΎΡΡΠ°Π΅ΡΡΡ ΠΎΡΠΎΠ±Π΅Π½Π½ΠΎ ΡΠ»ΠΎΠΆΠ½ΡΠΌ
ΠΠ΅ΡΡΠΏΠ΅ΠΊΡΠΈΠ²Ρ PARP-ΠΈΠ½Π³ΠΈΠ±ΠΈΡΠΎΡΠΎΠ² ΠΏΡΠΈ Π»Π΅ΡΠ΅Π½ΠΈΠΈ BRCA-Π°ΡΡΠΎΡΠΈΠΈΡΠΎΠ²Π°Π½Π½ΠΎΠ³ΠΎ ΡΠ°ΠΊΠ° ΠΏΠΎΠ΄ΠΆΠ΅Π»ΡΠ΄ΠΎΡΠ½ΠΎΠΉ ΠΆΠ΅Π»Π΅Π·Ρ. ΠΠ±Π·ΠΎΡ Π»ΠΈΡΠ΅ΡΠ°ΡΡΡΡ
Pancreatic adenocarcinoma has aΒ 5-year overall survival rate of 9 %, with an outlook of becoming the second leading cause of cancer mortality in the USA by 2030. Familial pancreatic cancer and genetic predisposition syndromes have attracted more interest in the perspective of targeted therapy. Various authors estimate genetic causes to account for 10β15 % of pancreatic cancers. The BRCA gene mutations comprise the todayβs most relevant genetic predisposition syndrome. The frequency of BRCA1/2 and PALB2 germinal mutations in patients with pancreatic adenocarcinoma constitutes about 5β9 %. Over recent years, PARP inhibitors (PARPi) have composed a new targeted therapy class with a significant effect in breast and ovarian cancers. With the mechanism of action of the PARP inhibitor and platinum drugs targeting different DNA repair pathways, their combination therapy has been suggested as promising. We report studies of a combination treatment with veliparib, gemcitabine and cisplatin in germinal BRCA1/2-mutation patients with advanced wild-type pancreatic adenocarcinoma (WT). Recent advances have identified patients with germinal and somatic mutations in the BRCA1/2 and other genes. HRD-targeted therapy, including platinum and PARP inhibitor drugs, can significantly improve survival.Β ΠΠ΄Π΅Π½ΠΎΠΊΠ°ΡΡΠΈΠ½ΠΎΠΌΠ° ΠΏΠΎΠ΄ΠΆΠ΅Π»ΡΠ΄ΠΎΡΠ½ΠΎΠΉ ΠΆΠ΅Π»Π΅Π·Ρ ΠΈΠΌΠ΅Π΅Ρ 5-Π»Π΅ΡΠ½ΡΡ ΠΎΠ±ΡΡΡ Π²ΡΠΆΠΈΠ²Π°Π΅ΠΌΠΎΡΡΡ 9 %, Π° ΠΊ 2030 Π³ΠΎΠ΄Ρ, ΠΏΠΎ ΠΏΡΠΎΠ³Π½ΠΎΠ·Π°ΠΌ, ΡΡΠ° ΠΎΠΏΡΡ
ΠΎΠ»Ρ ΡΡΠ°Π½Π΅Ρ Π²ΡΠΎΡΠΎΠΉ Π²Π΅Π΄ΡΡΠ΅ΠΉ ΠΏΡΠΈΡΠΈΠ½ΠΎΠΉ ΡΠΌΠ΅ΡΡΠ½ΠΎΡΡΠΈ ΠΎΡ ΠΎΠ½ΠΊΠΎΠ»ΠΎΠ³ΠΈΡΠ΅ΡΠΊΠΈΡ
Π·Π°Π±ΠΎΠ»Π΅Π²Π°Π½ΠΈΠΉ Π² Π‘Π¨Π. Π‘Π΅ΠΌΠ΅ΠΉΠ½ΡΠΉ ΡΠ°ΠΊ ΠΏΠΎΠ΄ΠΆΠ΅Π»ΡΠ΄ΠΎΡΠ½ΠΎΠΉ ΠΆΠ΅Π»Π΅Π·Ρ ΠΈ ΡΠΈΠ½Π΄ΡΠΎΠΌΡ Π³Π΅Π½Π΅ΡΠΈΡΠ΅ΡΠΊΠΎΠΉ ΠΏΡΠ΅Π΄ΡΠ°ΡΠΏΠΎΠ»ΠΎΠΆΠ΅Π½Π½ΠΎΡΡΠΈ ΡΡΠ°Π»ΠΈ ΠΎΠ±Π»Π°ΡΡΡΡ ΠΏΠΎΠ²ΡΡΠ΅Π½Π½ΠΎΠ³ΠΎ ΠΈΠ½ΡΠ΅ΡΠ΅ΡΠ° ΠΈΠ·-Π·Π° ΠΏΠΎΡΠ΅Π½ΡΠΈΠ°Π»ΡΠ½ΡΡ
Π²ΠΎΠ·ΠΌΠΎΠΆΠ½ΠΎΡΡΠ΅ΠΉ ΡΠ°ΡΠ³Π΅ΡΠ½ΠΎΠΉ ΡΠ΅ΡΠ°ΠΏΠΈΠΈ. ΠΠΎ ΠΎΡΠ΅Π½ΠΊΠ°ΠΌ ΡΠ°Π·Π»ΠΈΡΠ½ΡΡ
Π°Π²ΡΠΎΡΠΎΠ², ΠΏΡΠΈΠΌΠ΅ΡΠ½ΠΎ ΠΎΡ 10Β Π΄ΠΎ 15 % ΡΠ»ΡΡΠ°Π΅Π² ΡΠ°ΠΊΠ° ΠΏΠΎΠ΄ΠΆΠ΅Π»ΡΠ΄ΠΎΡΠ½ΠΎΠΉ ΠΆΠ΅Π»Π΅Π·Ρ ΠΎΠ±ΡΡΡΠ½ΡΠ΅ΡΡΡ Π³Π΅Π½Π΅ΡΠΈΡΠ΅ΡΠΊΠΎΠΉ ΠΏΡΠΈΡΠΈΠ½ΠΎΠΉ. ΠΠ· ΡΡΠΈΡ
ΡΠΈΠ½Π΄ΡΠΎΠΌΠΎΠ² Π½Π°ΡΠ»Π΅Π΄ΡΡΠ²Π΅Π½Π½ΠΎΠΉ ΠΏΡΠ΅Π΄ΡΠ°ΡΠΏΠΎΠ»ΠΎΠΆΠ΅Π½Π½ΠΎΡΡΠΈ Π½Π°ΠΈΠ±ΠΎΠ»Π΅Π΅ Π°ΠΊΡΡΠ°Π»ΡΠ½Ρ Π½Π° ΡΠ΅Π³ΠΎΠ΄Π½Ρ BRCA-ΠΌΡΡΠ°ΡΠΈΠΈ. Π§Π°ΡΡΠΎΡΠ° Π³Π΅ΡΠΌΠΈΠ½Π°Π»ΡΠ½ΡΡ
ΠΌΡΡΠ°ΡΠΈΠΉ Π²Β Π³Π΅Π½Π°Ρ
BRCA1/2Β ΠΈΒ PALB2Β ΡΒ ΠΏΠ°ΡΠΈΠ΅Π½ΡΠΎΠ² ΡΒ Π°Π΄Π΅Π½ΠΎΠΊΠ°ΡΡΠΈΠ½ΠΎΠΌΠΎΠΉ ΠΏΠΎΠ΄ΠΆΠ΅Π»ΡΠ΄ΠΎΡΠ½ΠΎΠΉ ΠΆΠ΅Π»Π΅Π·Ρ ΡΠΎΡΡΠ°Π²Π»ΡΠ΅Ρ ΠΎΠΊΠΎΠ»ΠΎ 5β9 %. ΠΒ ΠΏΠΎΡΠ»Π΅Π΄Π½ΠΈΠ΅ Π³ΠΎΠ΄Ρ ΠΈΠ½Π³ΠΈΠ±ΠΈΡΠΎΡΡ PARP (PARPi) ΠΏΠΎΡΠ²ΠΈΠ»ΠΈΡΡ ΠΊΠ°ΠΊ Π½ΠΎΠ²ΡΠΉ ΠΊΠ»Π°ΡΡ ΡΠ°ΡΠ³Π΅ΡΠ½ΠΎΠΉ ΡΠ΅ΡΠ°ΠΏΠΈΠΈ ΡΠΎ Π·Π½Π°ΡΠΈΡΠ΅Π»ΡΠ½ΠΎΠΉ Π°ΠΊΡΠΈΠ²Π½ΠΎΡΡΡΡ ΠΏΡΠΈ ΡΠ°ΠΊΠ΅ ΠΌΠΎΠ»ΠΎΡΠ½ΠΎΠΉ ΠΆΠ΅Π»Π΅Π·Ρ, ΡΠΈΡΠ½ΠΈΠΊΠΎΠ². Π£ΡΠΈΡΡΠ²Π°Ρ, ΡΡΠΎ ΠΌΠ΅Ρ
Π°Π½ΠΈΠ·ΠΌ Π΄Π΅ΠΉΡΡΠ²ΠΈΡ PARPΠΈΠ½Π³ΠΈΠ±ΠΈΡΠΎΡΠΎΠ² ΠΈ ΠΏΡΠ΅ΠΏΠ°ΡΠ°ΡΠΎΠ² ΠΏΠ»Π°ΡΠΈΠ½Ρ ΡΠ°Π·Π»ΠΈΡΠ΅Π½, ΠΎΠ½ΠΈ ΠΎΠΊΠ°Π·ΡΠ²Π°ΡΡ ΡΠ²ΠΎΠ΅ Π΄Π΅ΠΉΡΡΠ²ΠΈΠ΅ Π½Π° ΡΠ°Π·Π½ΡΠ΅ ΠΏΡΡΠΈ ΡΠ΅ΠΏΠ°ΡΠ°ΡΠΈΠΈ ΠΠΠ, Π±ΡΠ»ΠΎ Π²ΡΡΠΊΠ°Π·Π°Π½ΠΎ ΠΏΡΠ΅Π΄ΠΏΠΎΠ»ΠΎΠΆΠ΅Π½ΠΈΠ΅, ΡΡΠΎ ΠΊΠΎΠΌΠ±ΠΈΠ½ΠΈΡΠΎΠ²Π°Π½Π½Π°Ρ ΡΠ΅ΡΠ°ΠΏΠΈΡ ΠΌΠΎΠΆΠ΅Ρ ΠΏΡΠ΅Π΄ΡΡΠ°Π²Π»ΡΡΡ ΠΈΠ½ΡΠ΅ΡΠ΅Ρ. ΠΠ²ΡΠΎΡΡ ΡΠΎΠΎΠ±ΡΠΈΠ»ΠΈ ΠΎ ΡΠ΅Π·ΡΠ»ΡΡΠ°ΡΠ°Ρ
ΠΈΡΡΠ»Π΅Π΄ΠΎΠ²Π°Π½ΠΈΡ ΠΊΠΎΠΌΠ±ΠΈΠ½Π°ΡΠΈΠΈ Π²Π΅Π»ΠΈΠΏΠ°ΡΠΈΠ±Π°, Π³Π΅ΠΌΡΠΈΡΠ°Π±ΠΈΠ½Π° ΠΈ ΡΠΈΡΠΏΠ»Π°ΡΠΈΠ½Π° Ρ ΠΏΠ°ΡΠΈΠ΅Π½ΡΠΎΠ² Ρ Π³Π΅ΡΠΌΠΈΠ½Π°Π»ΡΠ½ΠΎΠΉ ΠΌΡΡΠ°ΡΠΈΠ΅ΠΉ BRCA1/2 ΠΈ ΡΠ°ΡΠΏΡΠΎΡΡΡΠ°Π½Π΅Π½Π½ΠΎΠΉ Π°Π΄Π΅Π½ΠΎΠΊΠ°ΡΡΠΈΠ½ΠΎΠΌΠΎΠΉ ΠΏΠΎΠ΄ΠΆΠ΅Π»ΡΠ΄ΠΎΡΠ½ΠΎΠΉ ΠΆΠ΅Π»Π΅Π·Ρ Π΄ΠΈΠΊΠΎΠ³ΠΎ ΡΠΈΠΏΠ° (WT). ΠΠΎΡΠ»Π΅Π΄Π½ΠΈΠ΅ Π΄ΠΎΡΡΠΈΠΆΠ΅Π½ΠΈΡ ΠΏΠΎΠ·Π²ΠΎΠ»ΠΈΠ»ΠΈ Π²ΡΡΠ²ΠΈΡΡ ΠΏΠ°ΡΠΈΠ΅Π½ΡΠΎΠ² Ρ Π³Π΅ΡΠΌΠΈΠ½Π°Π»ΡΠ½ΡΠΌΠΈ ΠΈ ΡΠΎΠΌΠ°ΡΠΈΡΠ΅ΡΠΊΠΈΠΌΠΈ ΠΌΡΡΠ°ΡΠΈΡΠΌΠΈ Π² Π³Π΅Π½Π°Ρ
BRCA1/2 ΠΈ Π΄ΡΡΠ³ΠΈΡ
. Π’Π΅ΡΠ°ΠΏΠΈΡ, Π½Π°ΠΏΡΠ°Π²Π»Π΅Π½Π½Π°Ρ Π½Π° HRD, Π²ΠΊΠ»ΡΡΠ°Ρ ΠΏΡΠ΅ΠΏΠ°ΡΠ°ΡΡ ΠΏΠ»Π°ΡΠΈΠ½Ρ ΠΈ PARP-ΠΈΠ½Π³ΠΈΠ±ΠΈΡΠΎΡΡ, ΠΌΠΎΠΆΠ΅Ρ Π·Π½Π°ΡΠΈΡΠ΅Π»ΡΠ½ΠΎ ΡΠ»ΡΡΡΠΈΡΡ Π²ΡΠΆΠΈΠ²Π°Π΅ΠΌΠΎΡΡΡ.
ΠΠ΅ΠΏΠ°ΡΠΎΡΠ΅Π»Π»ΡΠ»ΡΡΠ½Π°Ρ ΠΊΠ°ΡΡΠΈΠ½ΠΎΠΌΠ°: ΡΡΠΈΠΎΠ»ΠΎΠ³ΠΈΡΠ΅ΡΠΊΠΈΠ΅ ΡΠ°ΠΊΡΠΎΡΡ ΠΈ ΠΌΠ΅Ρ Π°Π½ΠΈΠ·ΠΌΡ ΡΠ°Π·Π²ΠΈΡΠΈΡ. ΠΠ±Π·ΠΎΡ Π»ΠΈΡΠ΅ΡΠ°ΡΡΡΡ
Liver cancer remains a global challenge of healthcare, with the incidence growing worldwide. According to various authors, over 1 million patients will be diagnosed with liver cancer each year by 2025. The molecular pathogenesis of HCC varies with respect to genotoxic lesions and aetiologies. Although our understanding of the HCC pathophysiology and drivers tends to improve, it is still distant from translation into clinical practice. About 25 % of HCC cases are associated with variant mutations. HCC pathophysiology is a complex multi-step process. The interaction of various factors underlies the early stages of malignant hepatocyte transformation towards the development of HCC. Overall, about 20β25 % of HCC patients have at least one potential driver mutation. Obesity should also be noted as being associated with a higher risk of HCC and various other cancers. Despite many issues in the HCC pathogenesis being already known, the unresolved questions remain. Modern molecular genetic diagnostics and animal modelling of malignant tumours are expanding our horizons of knowledge in this field.Π Π°ΠΊ ΠΏΠ΅ΡΠ΅Π½ΠΈ ΠΎΡΡΠ°Π΅ΡΡΡ Π³Π»ΠΎΠ±Π°Π»ΡΠ½ΠΎΠΉ ΠΏΡΠΎΠ±Π»Π΅ΠΌΠΎΠΉ Π·Π΄ΡΠ°Π²ΠΎΠΎΡ
ΡΠ°Π½Π΅Π½ΠΈΡ, ΠΈ Π·Π°Π±ΠΎΠ»Π΅Π²Π°Π΅ΠΌΠΎΡΡΡ ΡΡΠΎΠΉ ΠΏΠ°ΡΠΎΠ»ΠΎΠ³ΠΈΠ΅ΠΉ ΠΈΠΌΠ΅Π΅Ρ ΡΠ΅Π½Π΄Π΅Π½ΡΠΈΡ ΠΊ ΡΠΎΡΡΡ Π²ΠΎ Π²ΡΠ΅ΠΌ ΠΌΠΈΡΠ΅. ΠΠΎ ΠΎΡΠ΅Π½ΠΊΠ°ΠΌ ΡΠ°Π·Π½ΡΡ
Π°Π²ΡΠΎΡΠΎΠ² ΠΊ 2025 Π³ΠΎΠ΄Ρ Π±ΡΠ΄Π΅Ρ Π²ΡΡΠ²Π»ΡΡΡΡΡ Π΅ΠΆΠ΅Π³ΠΎΠ΄Π½ΠΎ Π±ΠΎΠ»Π΅Π΅ 1 ΠΌΠΈΠ»Π»ΠΈΠΎΠ½Π° ΠΏΠ°ΡΠΈΠ΅Π½ΡΠΎΠ² Ρ Π΄ΠΈΠ°Π³Π½ΠΎΠ·ΠΎΠΌ Β«ΡΠ°ΠΊ ΠΏΠ΅ΡΠ΅Π½ΠΈΒ». ΠΠΎΠ»Π΅ΠΊΡΠ»ΡΡΠ½ΡΠΉ ΠΏΠ°ΡΠΎΠ³Π΅Π½Π΅Π· Π³Π΅ΠΏΠ°ΡΠΎΡΠ΅Π»Π»ΡΠ»ΡΡΠ½ΠΎΠΉ ΠΊΠ°ΡΡΠΈΠ½ΠΎΠΌΡ (ΠΠ¦Π) Π²Π°ΡΡΠΈΡΡΠ΅Ρ Π² Π·Π°Π²ΠΈΡΠΈΠΌΠΎΡΡΠΈ ΠΎΡ ΡΠ°Π·Π»ΠΈΡΠ½ΡΡ
Π³Π΅Π½ΠΎΡΠΎΠΊΡΠΈΡΠ΅ΡΠΊΠΈΡ
ΠΏΠΎΡΠ°ΠΆΠ΅Π½ΠΈΠΉ ΠΈ ΡΡΠΈΠΎΠ»ΠΎΠ³ΠΈΠΈ. Π₯ΠΎΡΡ Π½Π°ΡΠΈ ΠΏΡΠ΅Π΄ΡΡΠ°Π²Π»Π΅Π½ΠΈΡ ΠΎ ΠΏΠ°ΡΠΎΡΠΈΠ·ΠΈΠΎΠ»ΠΎΠ³ΠΈΠΈ ΠΈ Π΄ΡΠ°ΠΉΠ²Π΅ΡΠ°Ρ
ΠΠ¦Π ΠΈΠΌΠ΅ΡΡ ΡΠ΅Π½Π΄Π΅Π½ΡΠΈΡ ΠΊ ΡΠ»ΡΡΡΠ΅Π½ΠΈΡ, ΡΡΠΈ ΠΏΠΎΠ½ΠΈΠΌΠ°Π½ΠΈΡ Π΅ΡΠ΅ Π΄Π°Π»Π΅ΠΊΠΈ Π΄Π»Ρ Π²ΠΎΠΏΠ»ΠΎΡΠ΅Π½ΠΈΡ Π² ΠΊΠ»ΠΈΠ½ΠΈΡΠ΅ΡΠΊΡΡ ΠΏΡΠ°ΠΊΡΠΈΠΊΡ. ΠΡΠΈΠΌΠ΅ΡΠ½ΠΎ 25 % ΡΠ»ΡΡΠ°Π΅Π² ΠΠ¦Π ΡΠ²ΡΠ·Π°Π½Ρ Ρ ΡΠ°Π·Π»ΠΈΡΠ½ΡΠΌΠΈ ΠΌΡΡΠ°ΡΠΈΡΠΌΠΈ. ΠΠ°ΡΠΎΡΠΈΠ·ΠΈΠΎΠ»ΠΎΠ³ΠΈΡ ΠΠ¦Π ΠΏΡΠ΅Π΄ΡΡΠ°Π²Π»ΡΠ΅Ρ ΡΠΎΠ±ΠΎΠΉ ΡΠ»ΠΎΠΆΠ½ΡΠΉ ΠΌΠ½ΠΎΠ³ΠΎΡΡΠ°ΠΏΠ½ΡΠΉ ΠΏΡΠΎΡΠ΅ΡΡ. ΠΠ·Π°ΠΈΠΌΠΎΠ΄Π΅ΠΉΡΡΠ²ΠΈΠ΅ ΡΠ°Π·Π»ΠΈΡΠ½ΡΡ
ΡΠ°ΠΊΡΠΎΡΠΎΠ² Π»Π΅ΠΆΠΈΡ Π² ΠΎΡΠ½ΠΎΠ²Π΅ ΡΠ°Π½Π½ΠΈΡ
ΡΡΠ°Π΄ΠΈΠΉ Π·Π»ΠΎΠΊΠ°ΡΠ΅ΡΡΠ²Π΅Π½Π½ΠΎΠΉ ΡΡΠ°Π½ΡΡΠΎΡΠΌΠ°ΡΠΈΠΈ Π³Π΅ΠΏΠ°ΡΠΎΡΠΈΡΠΎΠ² ΠΈ ΡΠ°Π·Π²ΠΈΡΠΈΠΈ ΠΠ¦Π. Π ΡΠ΅Π»ΠΎΠΌ ΡΠΎΠ»ΡΠΊΠΎ ΠΏΡΠΈΠΌΠ΅ΡΠ½ΠΎ 20β25 % ΠΏΠ°ΡΠΈΠ΅Π½ΡΠΎΠ² Ρ ΠΠ¦Π ΠΈΠΌΠ΅ΡΡ ΠΊΠ°ΠΊ ΠΌΠΈΠ½ΠΈΠΌΡΠΌ ΠΎΠ΄Π½Ρ ΠΏΠΎΡΠ΅Π½ΡΠΈΠ°Π»ΡΠ½ΡΡ Π΄ΡΠ°ΠΉΠ²Π΅ΡΠ½ΡΡ ΠΌΡΡΠ°ΡΠΈΡ. Π’Π°ΠΊΠΆΠ΅ ΡΠ»Π΅Π΄ΡΠ΅Ρ ΠΎΡΠΌΠ΅ΡΠΈΡΡ, ΡΡΠΎ ΠΎΠΆΠΈΡΠ΅Π½ΠΈΠ΅ ΡΠ²ΡΠ·Π°Π½ΠΎ Ρ ΠΏΠΎΠ²ΡΡΠ΅Π½Π½ΡΠΌ ΡΠΈΡΠΊΠΎΠΌ ΡΠ°Π·Π²ΠΈΡΠΈΡ ΠΠ¦Π ΠΈ ΡΠ°ΠΊΠ° Π²ΠΎ ΠΌΠ½ΠΎΠ³ΠΈΡ
ΠΎΡΠ³Π°Π½Π°Ρ
. ΠΠ΅ΡΠΌΠΎΡΡΡ Π½Π° ΠΌΠ½ΠΎΠ³ΠΈΠ΅ ΡΠΆΠ΅ ΠΈΠ·Π²Π΅ΡΡΠ½ΡΠ΅ ΠΌΠΎΠΌΠ΅Π½ΡΡ Π² ΠΏΠ°ΡΠΎΠ³Π΅Π½Π΅Π·Π΅ ΡΠ°Π·Π²ΠΈΡΠΈΡ ΠΠ¦Π, Π΅ΡΠ΅ ΠΎΡΡΠ°ΡΡΡΡ Π½Π΅ΡΠ΅ΡΠ΅Π½Π½ΡΠ΅ Π²ΠΎΠΏΡΠΎΡΡ. Π‘ΠΎΠ²ΡΠ΅ΠΌΠ΅Π½Π½ΡΠ΅ Π²ΠΎΠ·ΠΌΠΎΠΆΠ½ΠΎΡΡΠΈ ΠΌΠΎΠ»Π΅ΠΊΡΠ»ΡΡΠ½ΠΎ-Π³Π΅Π½Π΅ΡΠΈΡΠ΅ΡΠΊΠΎΠΉ Π΄ΠΈΠ°Π³Π½ΠΎΡΡΠΈΠΊΠΈ ΠΈ ΠΌΠΎΠ΄Π΅Π»ΠΈΡΠΎΠ²Π°Π½ΠΈΠ΅ Π·Π»ΠΎΠΊΠ°ΡΠ΅ΡΡΠ²Π΅Π½Π½ΡΡ
ΠΎΠΏΡΡ
ΠΎΠ»Π΅ΠΉ Π½Π° ΠΆΠΈΠ²ΠΎΡΠ½ΡΡ
ΠΏΠΎΠ·Π²ΠΎΠ»ΡΡΡ ΡΠ°ΡΡΠΈΡΠΈΡΡ Π³ΠΎΡΠΈΠ·ΠΎΠ½ΡΡ Π·Π½Π°Π½ΠΈΠΉ Π² ΡΡΠΎΠΉ ΠΎΠ±Π»Π°ΡΡΠΈ