-
1Academic Journal
المؤلفون: R. Orlova V., E. Kaledina A., A. Malkova M., N. Zhukova V., P. Naymushina A., N. Beliak P., A. Jukova E., A. Sarmatova S., N. Karaseva A., I. Egorova V., K. Teslenko N., A. Tarasov V., Р. Орлова В., Е. Каледина А., А. Малкова М., Н. Жукова В., П. Наймушина А., Н. Беляк П., А. Жукова Е., А. Сарматова С., Н. Карасева А., И. Егорова В., К. Тесленко Н., А. Тарасов В.
المصدر: Meditsinskiy sovet = Medical Council; № 9 (2021); 108-113 ; Медицинский Совет; № 9 (2021); 108-113 ; 2658-5790 ; 2079-701X
مصطلحات موضوعية: cancer, SARS-CoV-2, COVID-19, immunotherapy, chemotherapy, radiation therapy, злокачественное новообразование, иммунотерапия, химиотерапия, лучевая терапия
وصف الملف: application/pdf
Relation: https://www.med-sovet.pro/jour/article/view/6253/5667; Du Toit A. Outbreak of a Novel Coronavirus. Nat Rev Microbiol. 2020;18:123. https://doi.org/10.1038/s41579-020-0332-0.; Zhou P., Yang X., Wang X., Hu B., Zhang L., Zhang W. et al. A Pneumonia Outbreak Associated with a New Coronavirus of Probable Bat origin. Nature. 2020;579:270–273. https://doi.org/10.1038/s41586-020-2012-7.; Bertuzzi A.F., Marrari A., Gennaro N., Cariboni U., Ciccarelli M., Giordano L. et al. Low Incidence of SARS-CoV-2 in Patients with Solid Tumours on Active Treatment: An Observational Study at a Tertiary Cancer Centre in Lombardy, Italy. Cancers (Basel). 2020;12(9):2352. https://doi.org/10.3390/cancers12092352.; Wang Q., Berger N.A., Xu R. Analyses of Risk, Racial Disparity, and Outcomes Among US Patients With Cancer and COVID-19 Infection. JAMA Oncol. 2021;7(2):220–227. https://doi.org/10.1001/jamaoncol.2020.6178.; Liang W., Guan W., Chen R., Wang W., Li J., Xu K. et al. Cancer patients in SARS-CoV-2 infection: a nationwide analysis in China. Lancet Oncol. 2020;21(3):335–337. https://doi.org/10.1016/S1470-2045(20)30096-6.; Giannakoulis V.G., Papoutsi E., Siempos I.I. Effect of Cancer on Clinical Outcomes of Patients With COVID-19: A Meta-Analysis of Patient Data. JCO Glob Oncol. 2020;6:799–808. https://doi.org/10.1200/GO.20.00225.; Williamson E.J., Walker A.J., Bhaskaran K., Bacon S., Bates C., Morton C.E. et al. Factors associated with COVID-19-related death using OpenSAFELY. Nature. 2020;584:430–436. https://doi.org/10.1038/s41586-020-2521-4.; Docherty A.B., Harrison E.M., Green C.A., Hardwick H.E., Pius R., Norman L. et al. Features of 20 133 UK patients in hospital with covid-19 using the ISARIC WHO Clinical Characterisation Protocol: prospective observational cohort study. BMJ. 2020;369:m1985. https://doi.org/10.1136/bmj.m1985.; Kuderer N.M., Choueiri T.K., Shah D.P., Shyr Y., Rubinstein S.M., Rivera D.R. et al. COVID-19 and Cancer Consortium. Clinical impact of COVID-19 on patients with cancer (CCC19): a cohort study. Lancet. 2020;395(10241):1907– 1918. https://doi.org/10.1016/S0140-6736(20)31187-9.; Mehta V., Goel S., Kabarriti R., Cole D., Goldfinger M., Acuna-Villaorduna A. et al. Case Fatality Rate of Cancer Patients with COVID-19 in a New York Hospital System. Cancer Discov. 2020;10(7):935–941. https://doi.org/10.1158/2159-8290.CD-20-0516.; Lunski M.J., Burton J., Tawagi K., Maslov D., Simenson V., Barr D. et al. Multivariate mortality analyses in COVID-19: Comparing patients with cancer and patients without cancer in Louisiana. Cancer. 2021;127(2):266– 274. https://doi.org/10.1002/cncr.33243.; Derosa L., Melenotte C., Griscelli F., Gachot B., Marabelle A., Kroemer G., Zitvogel L. The immuno-oncological challenge of COVID-19. Nat Cancer. 2020;1:946–964. Available at: https://www.nature.com/articles/s43018-020-00122-3?error=cookies_not_supported&code=66955eff-548b-4bb7-8885-e78f30bb5078.; Murdoch C., Muthana M., Coffelt S.B., Lewis C.E. The role of myeloid cells in the promotion of tumour angiogenesis. Nat Rev Cancer. 2008;8:618– 631. https://doi.org/10.1038/nrc2444.; Hagemann T., Lawrence T., McNeish I., Charles K.A., Kulbe H., Thompson R.G. et al. “Re-educating” tumor-associated macrophages by targeting NF κB. J Exp Med. 2008;205(6):1261–1268. https://doi.org/10.1084/jem.20080108.; Oh H., Grinberg-Bleyer Y., Liao W., Maloney D., Wang P., Wu Z., Ghosh S. An NF κB transcription-factor-dependent lineage-specific transcriptional program promotes regulatory T cell identity and function. Immunity. 2017;47(3):450–465. https://doi.org/10.1016/j.immuni.2017.08.010.; Dai M., Liu D., Liu M., Zhou F., Li G., Chen Z. Patients with cancer appear more vulnerable to SARS-COV-2: a multi-center study during the COVID-19 outbreak. Cancer Discov. 2020;10(6);783–791. https://doi.org/10.1158/2159-8290.CD-20-0422.; Siu K.L., Chan C.P., Kok K.H., Chiu-Yat Woo P., Jin D.Y. Suppression of innate antiviral response by severe acute respiratory syndrome coronavirus M protein is mediated through the first transmembrane domain. Cell Mol Immunol. 2014;11:141–149. https://doi.org/10.1038/cmi.2013.61.; Versteeg G.A., Bredenbeek P.J., van den Worm S.H., Spaan W.J. Group 2 coronaviruses prevent immediate early interferon induction by protection of viral RNA from host cell recognition. Virology. 2007;361(1):18–26. https://doi.org/10.1016/j.virol.2007.01.020.; Sun L., Xing Y., Chen X., Zheng Y., Yang Y., Nichols D.B. et al. Coronavirus papain-like proteases negatively regulate antiviral innate immune response through disruption of STING-mediated signaling. PLoS One. 2012;7(2):e30802. https://doi.org/10.1371/journal.pone.0030802.; Narayanan K., Huang C., Lokugamage K., Kamitani W., Ikegami T., Tseng C.T.K., Makino S. Severe acute respiratory syndrome coronavirus nsp1 suppresses host gene expression, including that of type I interferon, in infected cells. J Virol. 2008;82(9):4471–4479. https://doi.org/10.1128/JVI.02472-07.; Frieman M., Ratia K., Johnston R.E., Mesecar A.D., Baric R.S. Severe acute respiratory syndrome coronavirus papain-like protease ubiquitin-like domain and catalytic domain regulate antagonism of IRF3 and NF-kappaB signaling. J Virol. 2009;83(13):6689–6705. https://doi.org/10.1128/JVI.02220-08.; Qin C., Zhou L., Hu Z., Zhang S., Yang S., Tao Y. et al. Dysregulation of immune response in patients with COVID-19 in Wuhan, China. Clin Infect Dis. 2020;71(15):762–768. https://doi.org/10.1093/cid/ciaa248.; Xie J., Fan H.W., Li T.S., Qiu Z.F., Han Y. Dynamic changes of T lymphocyte subsets in the long-term follow-up of severe acute respiratory syndrome patients. Chinese Academy of Medical Science. 2006;28(2):253–255. Available at: https://pubmed.ncbi.nlm.nih.gov/16733915/.; Li T., Qiu Z., Zhang L., Han Y., He W., Liu Z. et al. Significant Changes of Peripheral T Lymphocyte Subsets in Patients with Severe Acute Respiratory Syndrome. The Journal of Infectious Diseases. 2004;189(4):648– 651. https://doi.org/10.1086/381535.; Wong R.S., Wu A., To K.F., Lee N., Lam C.W., Wong C.K. et al. Haematological manifestations in patients with severe acute respiratory syndrome: retrospective analysis. BMJ. 2003;326:1358–1362. https://doi.org/10.1136/bmj.326.7403.1358.; Cui W., Fan Y., Wu W., Zhang F., Wang J.Y., Ni A.P. Expression of lymphocytes and lymphocyte subsets in patients with severe acute respiratory syndrome. Clinical infectious diseases. 2003;37(6):857–859. https://doi.org/10.1086/378587.; Zheng H.Y., Zhang M., Yang C.X., Zhang N., Wang X.C., Yang X.P. et al. Elevated exhaustion levels and reduced functional diversity of T cells in peripheral blood may predict severe progression in COVID-19 patients. Cell Mol Immunol. 2020;17:541–543. https://doi.org/10.1038/s41423-020-0401-3.; Gu J., Gong E., Zhang B., Zheng J., Gao Z., Zhong Y. et al. Multiple organ infection and the pathogenesis of SARS. J Exp Med. 2005;202(3):415–424. https://doi.org/10.1084/jem.20050828.; Cheung C.Y., Poon L.L., Ng I.H., Luk W., Sia S.F., Wu M.H. et al. Cytokine responses in severe acute respiratory syndrome coronavirus-infected macrophages in vitro: possible relevance to pathogenesis. J Virol. 2005;79(12):7819–7826. https://doi.org/10.1128/JVI.79.12.7819-7826.2005.; Yilla M., Harcourt B.H., Hickman C.J., McGrew M., Tamin A., Goldsmith C.S. et al. SARS-coronavirus replication in human peripheral monocytes/macrophages. Virus research. 2005;107(1):93–101. https://doi.org/10.1016/j.virusres.2004.09.004.; Tseng C.T.K., Perrone L.A., Zhu H., Makino S., Peters C.J. Severe acute respiratory syndrome and the innate immune responses: modulation of effector cell function without productive infection. J Immunol. 2005;174(12):7977–7985. https://doi.org/10.4049/jimmunol.174.12.7977.; Law H.K., Cheung C.Y., Ng H.Y., Sia S.F., Chan Y.O., Luk W. et al. Chemokine up-regulation in SARS-coronavirus–infected, monocyte-derived human dendritic cells. Blood. 2005;106(7):2366–2374. https://doi.org/10.1182/blood-2004-10-4166.; Tavakkoli M., Wilkins C.R., Mones J.V., Mauro M.J. A novel paradigm between leukocytosis, G-CSF secretion, neutrophil-to-lymphocyte ratio, myeloid-derived suppressor cells, and prognosis in non-small cell lung cancer. Front Oncol. 2019;9:295. https://doi.org/10.3389/fonc.2019.00295.; Clift A.K., Coupland C.A., Keogh R.H., Diaz-Ordaz K., Williamson E., Harrison E.M. et al. Living risk prediction algorithm (QCOVID) for risk of hospital admission and mortality from coronavirus 19 in adults: national derivation and validation cohort study. BMJ. 2020;371:m3731. https://doi.org/10.1136/bmj.m3731.; Yekedüz E., Utkan G., Ürün Y. A systematic review and meta-analysis: the effect of active cancer treatment on severity of COVID-19. Eur J Cancer. 2020;141:92–104. https://doi.org/10.1016/j.ejca.2020.09.028.; Lee L.Y., Cazier J.B., Starkey T., Turnbull C.D., Team U.C.C.M.P., Kerr R. et al. COVID-19 mortality in patients with cancer on chemotherapy or other anticancer treatments: a prospective cohort study. Lancet. 2020;395(10241): 1919–1926. https://doi.org/10.1016/S0140-6736(20)31173-9.; Tini G., Sarocchi M., Tocci G., Arboscello E., Ghigliotti G., Novo G. et al. Arterial hypertension in cancer: the elephant in the room. Int J Cardiol. 2019;281:133–139. https://doi.org/10.1016/j.ijcard.2019.01.082.; Jee J., Foote M., Lumish M., Stonestrom A., Wills B., Narendra V. et al. Chemotherapy and COVID-19 Outcomes in Patients With Cancer. J Clin Oncol. 2020;38(30):3538–3546. https://doi.org/10.1200/JCO.20.01307.; Pinato D., Zambelli A., Aguilar-Company J., Bower M., Sng C., Salazar R. et al. Clinical Portrait of the SARS-CoV-2 Epidemic in European Patients with Cancer. Cancer Discov. 2020;10(10):1465–1474. https://doi.org/10.1158/2159-8290.CD-20-0773.; Curigliano G., Banerjee S., Cervantes A., Garassino M., Garrido P., Girard N. et al. Managing cancer patients during the COVID-19 pandemic: an ESMO multidisciplinary expert consensus. Ann Oncol. 2020;31(10):1320–1335. https://doi.org/10.1016/j.annonc.2020.07.010.; Weisberg E., Parent A., Yang P., Sattler M., Liu Q., Liu Q. et al. Repurposing of Kinase Inhibitors for Treatment of COVID-19. Pharm Res. 2020;37:167. https://doi.org/10.1007/s11095-020-02851-7.; Luo J., Rizvi H., Egger J.V., Preeshagul I.R., Wolchok J.D., Hellmann M.D. Impact of PD-1 Blockade on Severity of COVID-19 in Patients with Lung Cancers. Cancer Discov. 2020;10(8):1121–1128. https://doi.org/10.1158/2159-8290.CD-20-0596.; Vardhana S.A., Wolchok J.D. The many faces of the anti-COVID immune response. J Exp Med. 2020;217(6):e20200678. https://doi.org/10.1084/jem.20200678.; Garassino M.C., Whisenant J.G., Huang L.C., Trama A., Torri V., Agustoni F. et al. COVID-19 in patients with thoracic malignancies (TERAVOLT): first results of an international, registry-based, cohort study. Lancet Oncol. 2020;21(7):914–922. https://doi.org/10.1016/S1470-2045(20)30314-4.; Robilotti E.V., Babady N.E., Mead P.A., Rolling T., Perez-Johnston R., Bernardes M. et al. Determinants of COVID-19 disease severity in patients with cancer. Nat Med. 2020;26:1218–1223. https://doi.org/10.1038/s41591-020-0979-0.; Vordermark D. Shift in indications for radiotherapy during the COVID-19 pandemic? A review of organ-specific cancer management recommendations from multidisciplinary and surgical expert groups. Radiat Oncol. 2020;15:140. https://doi.org/10.1186/s13014-020-01579-3.; Joseph N., Choudhury A. Lymphocytopenia and Radiotherapy Treatment Volumes in the Time of COVID-19. Clin Oncol. 2020;32(7):420–422. https://doi.org/10.1016/j.clon.2020.04.011.; Wild A., Herman J., Dholakia A., Moningi S., Lu Y., Rosati L. et al. Lymphocyte-Sparing Effect of Stereotactic Body Radiation Therapy in Patients With Unresectable Pancreatic Cancer. Int J Radiat Oncol. 2016;94(3):571–579. https://doi.org/10.1016/j.ijrobp.2015.11.026.; https://www.med-sovet.pro/jour/article/view/6253
-
2Academic Journal
المؤلفون: N. Besova S., T. Titova A., E. Artamonova V., D. Stroyakovskiy L., E. Perminova V., D. Yukal’chuk Yu., D. Ponomarenko M., N. Belyak P., R. Orlova V., G. Teletaeva M., E. Ratner Yu., A. Mochalova S., O. Gordeeva O., A. Zhabina S., S. Gamayunov V., A. Smolin V., A. Povyshev Yu., M. Andrievskikh I., A. Tryakin A., I. Stilidi S., Н. Бесова С., Т. Титова А., Е. Артамонова В., Д. Строяковский Л., Е. Перминова В., Д. Юкальчук Ю., Д. Пономаренко М., Н. Беляк П., Р. Орлова В., Г. Телетаева М., Е. Ратнер Ю., А. Мочалова С., О. Гордеева О., А. Жабина С., С. Гамаюнов В., А. Смолин В., А. Повышев Ю., М. Андриевских И., А. Трякин А., И. Стилиди С.
المصدر: Meditsinskiy sovet = Medical Council; № 19 (2019); 104-113 ; Медицинский Совет; № 19 (2019); 104-113 ; 2658-5790 ; 2079-701X ; 10.21518/2079-701X-2019-19
مصطلحات موضوعية: ramucirumab, gastric cancer, irinotecan, fluoropyrimidine adenocarcinoma, second line treatment, рамуцирумаб, рак желудка, вторая линия лечения, паклитаксел, иринотекан
وصف الملف: application/pdf
Relation: https://www.med-sovet.pro/jour/article/view/5238/4768; Bray F., Ferlay J., Soerjomataram I., Siegel R.L. Torre LA., Jemal A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2018 Nov;68(6):394-424. doi:10.3322/caac.21492.; Каприн А.Д., Старинский В.В., Петрова Г.В. (ред.). Злокачественные новообразования в России в 2017 году (заболеваемость и смертность). М.: МНИОИ им. П.А. Герцена – филиал ФГБУ «НМИЦ радиологии» Минздрава России; 2018. Режим доступа: http://www.oncology.ru/service/statistics/; Каприн А.Д., Старинский В.В., Петрова Г.В. (ред.). Состояние онкологической помощи населению России в 2018 году. М.: МНИОИ им. П.А. Герцена – филиал ФГБУ «НМИЦ радиологии» Минздрава России; 2019. 236 с. Режим доступа: http://www.oncology.ru/service/statistics.; Vanhoefer U., Rougier P., Wilke H., et al. Final results of a randomized phase III trial of sequential high-dose methotrexate, fluorouracil, and doxorubicin versus etoposide, leucovorin, and fluorouracil versus infusional fluorouracil and cisplatin in advanced gastric cancer: A trial of the European Organization for Research and Treatment of Cancer Gastrointestinal Tract Cancer Co-operative Group. J Clin Oncol. 2000 Jul;18(14):2648-2657. doi:10.1200/JCO.2000.18.14.2648.; Kang Y., Kang W.K., Shin D.B., et al. Randomized phase III trial of capecitabine/cisplatin (XP) vs. continuous infusion of 5-FU/cisplatin (FP) as first-line therapy in patient with advanced gastric cancer: Efficacy and safety results. J Clin Oncol. 2016;24(18_suppl). doi:10.1200/jco.2006.24.18_suppl.lba4018.; Al-Batran S.-E., Hartmann J.T., Probst S., et al. Phase III Trial in Metastatic Gastroesophageal Adenocarcinoma with Fluorouracil, Leucovorin Plus Either Oxaliplatin or Cisplatin: A Study of the Arbeitsgemeinschaft Internistische Onkologie. J Clin Oncol. 2008;(26):1435-1442. doi:10.1200/JCO.2007.13.9378.; Park Y.H., Kim B.S., Ryoo B.Y., Yang S.H. A phase II study of capecitabine plus 3-weekly oxaliplatin as first-line therapy for patients with advanced gastric cancer. Br J Cancer. 2006;94(7):959-963. doi:10.1038/sj.bjc.6603046.; Dank M., Zaluski J., Barone C., et al. Randomized phase III study comparing irinotecan combined with 5-fluorouracil and folinic acid to cisplatin combined with 5-fluorouracil in chemotherapy naive patients with advanced adenocarcinoma of the stomach or esophagogastric junction. Annals of Oncology 2008;(19):1450–1457. doi: doi:10.1093/annonc/mdn166.; Guimbaud R., Louvet C., Ries P., Ychou M., Maillard E., André T., et al. Prospective, randomized, multicenter, phase III study of fluorouracil, leucovorin, and irinotecan versus epirubicin, cisplatin, and capecitabine in advanced gastric adenocarcinoma: a French intergroup (Fédération Francophone de Cancérologie Digestive, Fédération Nationale des Centres de Lutte Contre le Cancer, and Groupe; Coopérateur Multidisciplinaire en Oncologie) study. J Clin Oncol. 2014;(32):3520–3526. doi:10.1200/ JCO.2013.54.1011.; Van Cutsem E., Moiseyenko V.M., Tjulandin S. et al. Phase III study of docetaxel and cisplatin plus fluorouracil compared with cisplatin and fluorouracil as first-line therapy for advanced gastric cancer: a report of the V325 Study Group. J Clin Oncol. 2006;(24):4991–4997. doi:10.1200/JCO.2006.06.842.; Al-Batran S.-E., Hartmann J.T., Hofheinz R., et al. Biweekly fluorouracil, leucovorin, oxaliplatin, and docetaxel (FLOT) for patients with metastatic adenocarcinoma of the stomach or esophagogastric junction: a phase II trial of the Arbeitsgemeinschaft Internistische Onkologie. Annals of Oncology. 2008;(19):1882–1887. doi:10.1093/annonc/mdn403.; Bang Y.-J., Van Cutsem E., Feyereislova A., et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastrooesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet. 2010;(376):687–697. doi:10.1016/S0140-6736(10)61121-X.; Thuss-Patience P.C., Kretzschmar A., Bichev D., Deist T., Hinke A., Breithaupt K., Dogan Y., Gebauer B., Schumacher G., Reichardt P. Survival advantage for irinotecan versus best supportive care as second-line chemotherapy in gastric cancer–a randomised phase III study of the Arbeitsgemeinschaft Internistische Onkologie (AIO). Eur J Cancer. 2011;(47):2306-2314. doi:10.1016/j.ejca.2011.06.002.; Ford H., Marshall A., Wadsley J., Coxon F.Y., et al. Cougar-02: A randomized phase III study of docetaxel versus active symptom control in advanced esophagogastric adenocarcinoma. Lancet Oncol. 2014;(15):78–86. doi:10.1016/S1470-2045(13)70549-7.; Kang J.H., Lee S.I., Lim do H., Park K.W., Oh S.Y., Kwon H.C., et al. Salvage chemotherapy for pretreated gastric cancer: a randomized phase III trial comparing chemotherapy plus best supportive care with best supportive care alone. J Clin Oncol. 2012;(30):1513-1518. doi:10.1200/JCO.2011.39.4585.; Hironaka S., Ueda S., Yasui H., Nishina T., Tsuda M., Tsumura T., et al. Randomized, open-label phase III study comparing irinotecan with paclitaxel in patients with advanced gastric cancer without severe peritoneal metastasis after failure of prior combination chemotherapy using fluoropyrimidine plus platinum:WJOG 4007 Trial. J Clin Oncol. 2013;31(35):4438–4444. doi:10.1200/JCO.2012.48.5805.; Fuchs C.S., Tomasek J., Yong C.J., Dumitru F., Passalacqua R., Goswami C., et al. Ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (REGARD): an international, randomised, multicentre, placebo-controlled, phase 3 trial. Lancet. 2014;(383):31-39. doi:10.1016/S0140-6736(13)61719-5.; Wilke H., Muro K., Van Cutsem E., Oh S.C., Bodoky G., Shimada Y., et al.; RAINBOW Study Group. Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastrooesophageal junction adenocarcinoma (RAINBOW): a double-blind, randomised phase 3 trial. Lancet Oncol. 2014;(15):1224-1235. doi:10.1016/S1470-2045(14)70420-6.; Galdy S., Cella C.A., Spada F., Murgioni S., Frezza A.M., Ravenda S.P., Zampino M.G., Fazio N., et al. Systemic therapy beyond first-line in advanced gastric cancer: An overview of the main randomized clinical trials. Crit Rev Oncol Hematol. 2016;(99):1-12. doi:10.1016/j.critrevonc.2015.09.004.; Tian S., Quan H., Xie C., Guo H., Lü F., Xu Y. et al. YN968D1 is a novel and selective inhibitor of vascular endothelial growth factor receptor-2 tyrosine kinase with potent activity in vitro and in vivo. Cancer Sci. 2011;102(7):1374-80. doi:10.1111/j.1349-7006.2011.01939.x.; Spratlin J.L., Cohen R.B., Eadens M., Gore L., Camidge D.R., Diab S. et al. Phase I pharmacologic and biologic study of ramucirumab (IMC-1121B), a fully human immunoglobulin G1 monoclonal antibody targeting the vascular endothelial growth factor receptor-2. J Clin Oncol. 2010;28(5):780-787. doi:10.1200/JCO.2009.23.7537.; Lu D., Shen J., Vil M.D., Zhang H., Jimenez X., Bohlen P., Witte L., Zhu Z. Tailoring in vitro selection for a picomolar affinity human antibody directed against vascular endothelial growth factor receptor 2 for enhanced neutralizing activity. J Biol Chem. 2003;278(44):43496-43507. doi:10.1074/jbc.M307742200.; Di Bartolomeo M., Niger M., Tirino G., et al. Ramucirumab as Second-Line Therapy in Metastatic Gastric Cancer: Real-World Data from the RAMoss Study. Targeted Oncology. 2018;13(Issue 2):227–234. doi:10.1007/s11523-018-0562-5.; Klempner S.J., Maron S.B., Chase K., et al. Initial Report of Second-Line FOLFIRI in Combination with Ramucirumab in Advanced Gastroesophageal Adenocarcinomas: A MultiInstitutional Retrospective Analysis. The Oncologist. 2019;(24):475–482. doi:10.1634/theoncologist.2018-0602.; Therasse P., Arbuck S.G., Eisenhauer E.A., et al. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst. 2000;(92):205–216. doi:10.1093/jnci/92.3.205.; Fuchs C.S., Muro K., Tomasek J., Van Cutsem E., Cho J.Y., Oh S-C., et al. Prognostic Factor Analysis of Overall Survival in Gastric Cancer from Two Phase III Studies of Second-line Ramucirumab (REGARD and RAINBOW) Using Pooled Patient Data. J Gastric Cancer. 2017;17(2):132-144. doi:10.5230/jgc.2017.17.; Tabernero J., Yoshino T., Cohn A.L., Obermannova R., Bodoky G., Garcia-Carbonero R., Nasroulah F. Ramucirumab versus placebo in combination with second-line FOLFIRI in patients with metastatic colorectal carcinoma that progressed during or after first-line therapy with bevacizumab, oxaliplatin, and a fluoropyrimidine (RAISE): a randomised, doubleblind, multicentre, phase 3 study. The Lancet Oncology. 2015;16(5):499–508. doi:10.1016/s1470-2045(15)70127-0.; https://www.med-sovet.pro/jour/article/view/5238
-
3Academic Journal
المؤلفون: R. Orlova V., A. Ivanova K., G. Raskin A., S. Kutukova I., A. Androsova V., Р. Орлова В., А. Иванова К., Г. Раскин А., С. Кутукова И., А. Андросова В.
المصدر: Malignant tumours; Том 9, № 1 (2019); 16-21 ; Злокачественные опухоли; Том 9, № 1 (2019); 16-21 ; 2587-6813 ; 2224-5057
مصطلحات موضوعية: colon cancer, chemokine receptors, prognostic factor, adjuvant therapy, рак ободочной кишки, хемокиновые рецепторы, прогностический фактор, адъювантное лечение
وصف الملف: application/pdf
Relation: https://www.malignanttumors.org/jour/article/view/611/419; Каприн А. Д., Старинский В. В., Петрова Г. В. Состояние онкологической помощи населению России в 2016 году, Москва 2017 (стр.157).; Мерабишвили В. М. Злокачественные новообразования в северо западном федеральном округе России (заболеваемость, смертность, контингенты, выживаемость больных), выпуск третий под ред. Беляева А. М, СПб,2017, стр.22 32.; Journal of Clinical Oncology,27,19, 01.07.2009.; D. Sargent ASCO 2014,#3507; Практические рекомендации по лечению злокачественных опухолей Российского общества клинической онкологии, версия 2017 (стр.261 264).; Expression pattern of CXC chemokine receptor 4 is correlated with lymph node metastasis in human invasive ductal carcinoma. Masahiro Kato, Joji Kitayama, Shinsuke Kazama, Hirokazu Nagawa; Breast Cancer Research, October,2003.; Чердынцева Н. В., Гервас П. А., Литвяков Н. В. с соавт. Хемокины и прогрессия злокачественных новообразований. Медицинская Иммунология.–2006. – Т. 8, – № 2 3. – С.355 356.; А. С. Симбирцев Цитокины: классификация и биологические функции // Цитокины и воспаление. 2004. Том 3 № 2. – С.16 22.; Strieter R. M., Polverini P. J., Kunkel S. L. et al. The functional role of the ELR motif in CXC chemokine mediated angiogenesis. J Biol Chem. 1995. 270 (45): 27348 57.; Rivera CG, Bader JS, Popel AS. Angiogenesis associated crosstalk between collagens, CXC chemokines, and thrombospondin domain containing proteins. Ann Biomed Eng. 2011; 39 (8):2213 22.; Бережная Н. М., Чехун В. Ф. Иммунология злокачественного роста – 2005; Muller A, Homey B, Soto H, et al. Involvement of chemokine receptors in breast cancer metastasis. Nature. 2001; 410:50 56.; Zeelenberg IS, Ruuls Van Stalle L, Roos E. The chemokine receptor CXCR4 is required for outgrowth of colon carcinoma micrometastases. Cancer Res. 2003 Jul 1; 63 (13):3833 9.; Sehgal, A., Keener, C., Boynton, AL., Warrick, J., and Murphy, GP. CXCR 4, a chemokine receptor, is overexpressed in and required for proliferation of glioblastoma tumor cells.1998. J. Surg. Oncol. 69, 99 104; Uchida D, Begum NM, Almofti A, Nakashiro K, Kawamata H, Tateishi Y, Hamakawa H, Yoshida H, Sato M. Possible role of stromal cellderived factor 1/CXCR4 signaling on lymph node metastasis of oral squamous cell carcinoma. Exp Cell Res 2003. 290: 289 302.; Yu X et all J Exp Clin Cancer Res. 2019 Jan 24;38 (1):32. doi:10.1186/s13046-018-1014-x. CXCL12/CXCR4 promotes inflammation driven colorectal cancer -progression through activation of RhoA signaling by sponging miR 133a 3p.; Kim J, Mori T, Chen SL, Amersi FF, Martinez SR, Kuo C, Turner RR, Ye X, Bilchik AJ, Morton DL, Hoon DS. Chemokine receptor CXCR4 expression in patients with melanoma and colorectal cancer liver metastases and the associa tion with disease outcome. Ann Surg. 2006 Jul; 244 (1):113 20.; https://www.malignanttumors.org/jour/article/view/611
-
4Academic Journal
المؤلفون: G. Raskin A., A. Ivanova K., R. Orlova V., Г. Раскин А., А. Иванова К., Р. Орлова В.
المصدر: Advances in Molecular Oncology; Том 4, № 1 (2017); 35-39 ; Успехи молекулярной онкологии; Том 4, № 1 (2017); 35-39 ; 2413-3787 ; 2313-805X ; 10.17650/2313-805X-2017-4-1
مصطلحات موضوعية: neuroendocrine tumor, Ki-67 proliferation index, scintigraphy with octreotide, positron emission tomography, нейроэндокринная опухоль, индекс пролиферации Ki-67, сцинтиграфия с октреотидом, позитронно-эмиссионная томография
وصف الملف: application/pdf
Relation: https://umo.abvpress.ru/jour/article/view/86/100; Горбунова В.А. Нейроэндокринные опухоли. Общие принципы диагностики и лечения. М.: Кодекс, 2015. 456 с. [Gorbunova V.A. Neuroendocrine tumors. General principles of diagnostics and treatment. Moscow: Kodeks, 2015. 456 p. (In Russ.)].; Симоненко В.Б., Дудин П.А., Маканин М.А. Нейроэндокринные опухоли. М.: ГЭОТАР- Медиа, 2010. 237 с. [Simonenko V.B., Dudin P.A., Makanin M.A. Neuroendocrine tumors. Moscow: GEOTAR-Media, 2010. 237 p. (In Russ.)].; WHO classification of tumors the digestive system. Ed. by F.T. Bosman, F. Carneiro, R.H. Hruban, N.D. Theise. Lyon: IARC Press, 2010.; WHO classification of tumours of the lung, pleura, thymus and heart. Ed. by W.D. Travis, E. Brambilla, A.P. Burke et al. Lyon: IARC Press, 2015.; Sorbye H., Welin S., Langer S.W. et al. Predictive and prognostic factors for treatment and survival in 305 patients with advanced gastrointestinal neuroendocrine carcinoma (WHO G3): The NORDIC NEC study. Ann Oncol 2013;24(1): 152–60.; Khan S., Luong T.V., Watkins J. et al. A comparison of Ki-67 and mitotic count as prognostic markers for metastatic pancreatic and midgut neuroendocrine neoplasms. Br J Cancer 2013;108(9):1838–45.; Scarpa A., Mantovani W., Capelli P. et al. Pancreatic endocrine tumors: improved TNM staging and histopathological grading permit a clinically efficient prognostic stratification of patients. Mod Pathol 2010;23(6):824–33.; Arco D.D., Pérez D., Medina O. et al. Reliability of Ki-67 determination in FNA samples for grading pancreatic neuroendocrine tumors. Endocr Pathol 2016;27(4):276–83.; Farrell J.M., Pang J.C., Kim G.E., Tabatabai Z.L. Pancreatic neuroendocrine tumors: accurate grading with Ki-67 index on fine-needle aspiration specimens using the WHO 2010/ENETS criteria. Cancer Cytopathol 2014;122(10): 770–8.; Kleist B., Poetsch M. Neuroendocrine differentiation: The mysterious fellow of colorectal cancer. World J Gastroenterol 2015;21(41):11740–7.; Sentani K., Oue N., Noguchi T. et al. Immunostaining of gastric cancer with neuroendocrine differentiation: Reg IV-positive neuroendocrine cells are associated with gastrin, serotonin, pancreatic polypeptide and somatostatin. Pathol Int 2010;60(4):291–7.; Swarts D.R., van Suylen R.J., den Bakker M.A. et al. Interobserver variability for the WHO classification of pulmonary carcinoids. Am J Surg Pathol 2014;38(10):1429–36.; Takeuchi K., Togashi Y., Kamihara Y. et al. Prospective and clinical validation of ALK immunohistochemistry: results from the phase I/II study of alectinib for ALKpositive lung cancer (AF-001JP study). Ann Oncol 2016;27(1):185–92.; WHO classification of tumours of the breast. Ed. by S.R. Lakhani, I.O. Ellis, S.J. Schnitt et al. Lyon: IARC Press, 2012.; Gao L.X., Liu G., Li L. et al. Neuroendocrine carcinoma of breast: a study of tumor morphology and subtyping. Zhonghua Bing Li Xue Za Zhi 2011;40(9):604–9.; WHO classification of tumours. Pathology and genetics of tumours of the breast and female genital organs. Ed. by F.A. Tavassoli, P. Devilee. Lyon: IARC Press, 2003.; Miremadi A., Pinder S.E., Lee A.H. et al. Neuroendocrine differentiation and prognosis in breast adenocarcinoma. Histopathology 2002;40(3):215–22.; Inno A., Bogina G., Turazza M. et al. Neuroendocrine carcinoma of the breast: current evidence and future perspectives. Oncologist 2016;21(1):28–32.; Wei B., Ding T., Xing Y. et al. Invasive neuroendocrine carcinoma of the breast: a distinctive subtype of aggressive mammary carcinoma. Cancer 2010;116(19):4463–73.; Tian Z., Wei B., Tang F. et al. Prognosticsignificance of tumor grading and staging in mammary carcinomas with neuroendocrine differentiation. Hum Pathol 2011;42(8):1169–77.; Kwon S.Y., Bae Y.K., Gu M.J. et al. Neuroendocrine differentiation correlates with hormone receptor expression and decreased survival in patients with invasive breast carcinoma. Histopathology 2014;64(5):647–59.; https://umo.abvpress.ru/jour/article/view/86
-
5Academic Journal
المؤلفون: P. Borisov S., M. Shkol’nik I., R. Orlova V., P. Karlov A., П. Борисов С., М. Школьник И., Р. Орлова В., П. Карлов А.
المصدر: Cancer Urology; Том 12, № 3 (2016); 52-57 ; Онкоурология; Том 12, № 3 (2016); 52-57 ; 1996-1812 ; 1726-9776 ; 10.17650/1726-9776-2016-12-3
مصطلحات موضوعية: metastatic renal cell cancer, targeted medications, progression-free survival, overall survival, метастатический почечно-клеточный рак, таргетные препараты, выживаемость без прогрессирования, общая выживаемость
وصف الملف: application/pdf
Relation: https://oncourology.abvpress.ru/oncur/article/view/608/577; https://oncourology.abvpress.ru/oncur/article/view/608/644; Simard E.P., Ward E.M., Siegel R., Jemal A. Cancers with Increasing Incidence Trends in the United States: 1999 through 2008. CA Cancer J Clin 2012;62(2): 118–28. 1.DOI:10.3322/caac.20141.; Злокачественные новообразования в России в 2013 году (заболеваемость и смертность). Под ред. А.Д. Каприна, В.В. Старинского, Г.В. Петровой. М., 2015. [Malignant tumors in Russia in 2013 (morbidity and fatality). Ed. by: А.D. Kaprin, V.V. Starinskiy, G.V. Petrova. Мoscow, 2015. (In Russ.)].; Keane T., Gilliatt D. Current and future trends in treatment of renal cancer. Eur Urol 2007;(Suppl 6):374–84.; Cohen H.T., McCovern F.J. Renal-Cell Carcinoma. N. Engl J Med 2005;353(23):2477–90.; Motzer R.J., Bacik J., Murphy B.A. Interferon-alfa as a comparative treatment for clinical trials of new therapies against advanced renal cell carcinoma. J Clin Oncol 2002;20(1):289–96.; Coppin C., Porzsolt F., Awa A. et al. Immunotherapy for advanced renal cell cancer. Cochrane Database Syst Rev 2005;(1):CD001425.; Negrier S., Perol D., Ravaud A. et al. Medroxyprogesterone, interferon alfa-2a, interleukin 2, or combination of both cytokines in patients with metastatic renal carcinoma of intermediate prognosis: results of a randomized controlled trial. Cancer 2007;110(11):2468–77.; Motzer R.J., Masumdar M., Murphy B.A. et al. Survival and prognostic stratification of 670 patients with advanced renal cell carcinoma. J Clin Oncol 1999;17(8):2530–40.; Алексеев Б.Я., Калпинский А.С. Эффективность применения таргетной терапии в гетерогенной популяции больных метастатическим раком почки. Онкоурология 2012;8(3): 37–42. [Аlekseev B.Ya., Kalpinskiy А.S. Target therapy efficiency in the geterogenous population of patients with metastatic kidney cancer. Onkourologiya = Оncourology 2012;8(3):37–42. (In Russ.)].; https://oncourology.abvpress.ru/oncur/article/view/608
-
6Academic Journal
المؤلفون: P. Borisov S., R. Orlova V., M. Shkolnik I., P. Karlov A., П. Борисов С., Р. Орлова В., М. Школьник И., П. Карлов А.
المصدر: Cancer Urology; Том 11, № 3 (2015); 55-61 ; Онкоурология; Том 11, № 3 (2015); 55-61 ; 1996-1812 ; 1726-9776 ; 10.17650/1726-9776-2015-11-3
مصطلحات موضوعية: metastatic renal cell carcinoma, incomplete metastasectomy, incomplete cytoreduction, cytoreductive intervention, palliative treatment for metastatic kidney cancer, combination treatment for metastatic kidney cancer, targeted therapy, cytokine therapy, метастатический почечно-клеточный рак, неполная метастазэктомия, неполная циторедукция, циторедуктивное вмешательство, паллиативное лечение метастатического рака почки, комплексное лечение метастатического рака почки, таргетная терапия, терапия цитокина
وصف الملف: application/pdf
Relation: https://oncourology.abvpress.ru/oncur/article/view/489/455; Алексеев Б.Я., Калпинский A.C. Применение таргетных препаратов в лечении метастатического рака почки: последовательное назначение или комбинация. Онкоурология 2010;(4):16–23. [Alekseyev B.Ya., Kalpinskiy A.S. Application of Target Drugs in Treatment of Metastatic Kidney Cancer: Sequential Assignment or a Combination. Oncourology (Onkourologiya) 2010;(4):16–23. (In Russ.)].; Ljungberg B., Bensalah K., Bex A. et al. Guidelines on renal cell carcinoma. European Urology Association Guidelines. Update 2015. P. 30.; Alt A.L., Boorjian S.A., Lohse C.M. et al. Survival after complete surgical resection of multiple metastases from renal cell carcinoma. Cancer 2011;117(13):2873–8.; Kwak C., Park Y.H., Jeong C.W. et al. Metastasectomy without systemic therapy in metastatic renal cell carcinoma: Comparison with conservative treatment. Urol Int 2007;79(2):145–51.; Lee S.E., Kwak C., Byun S.S. et al. Metastatectomy prior to immunochemotherapy for metastatic renal cell Carcinoma. Urol Int 2006;76(3):256–63.; Petralia G., Roscigno M., Zigeuner R. et al. Complete metastasectomy is an independent predictor of cancerspecific survival in patientswith clinically metastatic renal cell carcinoma. Eur Urol Suppl 2010;9:162.; Staehler M., Kruse J., Haseke N. et al. Metastasectomy significantly prolongs survival in patients with metastatic renal cancer. Eur Urol Suppl 2009;8:181.; Eggener S.E., Yossepowitch O., Kundu S. et al. Risk score and metastasectomy ndependently impact prognosisof patients with recurrent renal cell carcinoma. J Urol 2008;180:873–8.; Давыдов М.И., Матвеев В.Б., Полоцкий Б.Е. и др. Хирургическое лечение метастазов рака почки в легких. Российский онкологический журнал 2003;4:15–9. [Davydov M.I., Matveyev V.B., Polotskiy B.E., et al. Surgical Treatment of Kidney Cancer Metastases in the Lungs. Russian Journal of Oncology (Rossiyskiy Onkologicheskiy Zhournal) 2003;4:15–9. (In Russ.)].; Kavolius J.P., Mastorakos D.P., Pavlovich C. et al. Resection of metastatic renal cell carcinoma. J Clin Oncol 1998;16:2261–6.; Fuchs B., Trousdale R.T., Rock M.G. Solitary bony metastasis from renal cell carcinoma: significance of surgical treatment. Clin Orthop Relat Res 2005;(431):187–92.; Кострицкий С.В., Широкорад В.И., Семенов Д.В. и др. Хирургическое лечение метастазов рака почки в длинные трубчатые кости. Онкоурология 2013;(2):18–20. [Kostritskiy S.V., Shirokorad V.I., Semyonov D.V. et al. Surgical Treatment of Kidney Cancer Metastases in Long Bones. Oncourology (Onkourologiya) 2013;(2):18-20. (In Russ.)].; https://oncourology.abvpress.ru/oncur/article/view/489