-
食管癌是一种消化道恶性肿瘤,在全球发病率很高且患病人数在逐年上升。2020年全球新发确诊食管癌病例60.4万,因食管癌死亡人数约54.4万,发病率和病死率分别位居全球恶性肿瘤第7位和第6位[1]。我国是全球食管癌发病人数和死亡人数最多的国家,食管癌俨然已成为严重威胁我国国民健康的主要恶性肿瘤之一。在过去的几十年里,食管癌的治疗模式主要是手术切除结合放化疗,但该疗法效果并不理想,预后差,病死率高。近些年,分子靶向治疗和免疫治疗应运而生,打破了食管癌的治疗瓶颈,在食管癌的治疗中发挥重要作用,正逐步成为食管癌的一线治疗方案。本研究综述目前主要的食管癌治疗靶点及其相关靶向药物的研究进展。
Research progress on targeted therapy and immunotherapy for esophageal cancer
-
摘要: 食管癌是一种在全球发病率和致死率都极高的恶性肿瘤,其致病因素复杂、多样,早期无明显症状,大部分患者在初诊时已是中晚期,预后差。常规手术切除结合放化疗的治疗模式已经不能满足当前疾病的治疗需求,亟需寻找新的治疗策略。分子靶向治疗和免疫治疗是近些年兴起的新型治疗方法,打破了食管癌的治疗瓶颈,已经成为食管癌治疗的重要组成部分。该研究就目前食管癌分子靶向治疗和免疫治疗的主要靶点及其相关靶向药物的研究进展进行综述,为精准医学在食管癌领域的应用提供借鉴。Abstract: Esophageal cancer is a malignant tumor with high incidence and mortality rate in the world and its pathogenic factors are complex and diverse. There are no obvious symptoms in the early stage, and most patients are in the middle to late stage at the initial diagnosis. The prognosis of esophageal cancer is poor. The treatment mode of conventional surgical resection combined with chemoradiotherapy can no longer meet the current treatment needs of disease, and new treatment strategies are urgently needed. Molecular targeted therapy and immunotherapy are new treatment methods that have emerged in recent years, which have broken the therapeutic bottleneck and have been proven to play important roles in the treatment of esophageal cancer. The current research progress of the main targets and their related targeted drugs in molecular targeted therapy and immunotherapy for esophageal cancer were reviewed in this article, which provided reference for the application of precision medicine in the field of esophageal cancer.
-
Key words:
- esophageal cancer /
- molecular targeted therapy /
- immunotherapy /
- molecular mechanisms
-
[1] SUNG H, FERLAY J, SIEGEL R L, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries[J]. CA Cancer J Clin, 2021, 71(3):209-249. doi: 10.3322/caac.21660 [2] YAMAOKA T, OHBA M, OHMORI T. Molecular-targeted therapies for epidermal growth factor receptor and its resistance mechanisms[J]. Int J Mol Sci, 2017, 18(11):2420. doi: 10.3390/ijms18112420 [3] HANAWA M, SUZUKI S, DOBASHI Y, et al. EGFR protein overexpression and gene amplification in squamous cell carcinomas of the esophagus[J]. Int J Cancer, 2006, 118(5):1173-1180. doi: 10.1002/ijc.21454 [4] RUHSTALLER T, THUSS-PATIENCE P, HAYOZ S, et al. Neoadjuvant chemotherapy followed by chemoradiation and surgery with and without cetuximab in patients with resectable esophageal cancer: a randomized, open-label, phase III trial (SAKK 75/08)[J]. Ann Oncol, 2018, 29(6):1386-1393. doi: 10.1093/annonc/mdy105 [5] HUANG Z H, MA X W, ZHANG J, et al. Cetuximab for esophageal cancer: an updated meta-analysis of randomized controlled trials[J]. BMC Cancer, 2018, 18(1):1-13. doi: 10.1186/s12885-017-3892-2 [6] SUNTHARALINGAM M, WINTER K, ILSON D, et al. Effect of the addition of cetuximab to paclitaxel, cisplatin, and radiation therapy for patients with esophageal cancer[J]. JAMA Oncol, 2017, 3(11):1520-1528. doi: 10.1001/jamaoncol.2017.1598 [7] LU M, WANG X C, SHEN L, et al. Nimotuzumab plus paclitaxel and cisplatin as the first line treatment for advanced esophageal squamous cell cancer: a single centre prospective phase Ⅱ trial[J]. Cancer Sci, 2016, 107(4):486-490. doi: 10.1111/cas.12894 [8] DUTTON S J, FERRY D R, BLAZEBY J M, et al. Gefitinib for oesophageal cancer progressing after chemotherapy (COG): a phase 3, multicentre, double-blind, placebo-controlled randomised trial[J]. Lancet Oncol, 2014, 15(8):894-904. doi: 10.1016/S1470-2045(14)70024-5 [9] XIE C, JING Z, LUO H, et al. Chemoradiotherapy with extended nodal irradiation and/or erlotinib in locally advanced oesophageal squamous cell cancer: long-term update of a randomised phase 3 trial[J]. Br J Cancer, 2020, 123(11):1616-1624. doi: 10.1038/s41416-020-01054-6 [10] XU X L, LING Z Q, CHEN W, et al. The overexpression of VEGF in esophageal cancer is associated with a more advanced TMN stage: a meta-analysis[J]. Cancer Biomark, 2013, 13(2):105-113. doi: 10.3233/CBM-130343 [11] FUCHS C S, TOMASEK J, YONG C J, et al. Ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (REGARD): an international, randomised, multicentre, placebo-controlled, phase 3 trial[J]. Lancet, 2014, 383(9911):31-39. doi: 10.1016/S0140-6736(13)61719-5 [12] WILKE H, MURO K, VAN CUTSEM E, et al. Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (RAINBOW): a double-blind, randomised phase 3 trial[J]. Lancet Oncol, 2014, 15(11):1224-1235. doi: 10.1016/S1470-2045(14)70420-6 [13] XU R H, ZHANG Y Q, PAN H M, et al. Efficacy and safety of weekly paclitaxel with or without ramucirumab as second-line therapy for the treatment of advanced gastric or gastroesophageal junction adenocarcinoma (RAINBOW-Asia): a randomised, multicentre, double-blind, phase 3 trial[J]. Lancet Gastroenterol Hepatol, 2021, 6(12):1015-1024. doi: 10.1016/S2468-1253(21)00313-7 [14] LI Y W, FENG H, REN P, et al. Safety and efficacy of apatinib monotherapy for unresectable, metastatic esophageal cancer: a single-arm, open-label, phase Ⅱ study[J]. Oncologist, 2020, 25(10):e1464-e1472. doi: 10.1634/theoncologist.2020-0310 [15] CHU L, LIANG F, ZHANG J, et al. A phase II study of apatinib in patients with recurrent/metastatic esophageal squamous cell carcinoma (ESCC)[J]. Ann Oncol, 2019, 30:ix177. [16] ALMHANNA K, ROSA M, HENDERSON-JACKSON E, et al. Her-2 expression in gastroesophageal intestinal Metaplasia, dysplasia, and adenocarcinoma[J]. Appl Immunohistochem Mol Morphol, 2016, 24(9):633-638. doi: 10.1097/PAI.0000000000000243 [17] 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 gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial[J]. Lancet, 2010, 376(9742):687-697. doi: 10.1016/S0140-6736(10)61121-X [18] DOI T, SHITARA K, NAITO Y, et al. Safety, pharmacokinetics, and antitumour activity of trastuzumab deruxtecan (DS-8201), a HER2-targeting antibody-drug conjugate, in patients with advanced breast and gastric or gastro-oesophageal tumours: a phase 1 dose-escalation study[J]. Lancet Oncol, 2017, 18(11):1512-1522. doi: 10.1016/S1470-2045(17)30604-6 [19] MAKIYAMA A, SUKAWA Y, KASHIWADA T, et al. Randomized, phase Ⅱ study of trastuzumab beyond progression in patients with HER2-positive advanced gastric or gastroesophageal junction cancer: WJOG7112G (T-ACT study)[J]. J Clin Oncol, 2020, 38(17):1919-1927. doi: 10.1200/JCO.19.03077 [20] GUO X F, LI S S, ZHU X F, et al. Lapatinib in combination with paclitaxel plays synergistic antitumor effects on esophageal squamous cancer[J]. Cancer Chemother Pharmacol, 2018, 82(3):383-394. doi: 10.1007/s00280-018-3627-3 [21] HECHT J R, BANG Y J, QIN S K, et al. Lapatinib in combination with capecitabine plus oxaliplatin in human epidermal growth factor receptor 2-positive advanced or metastatic gastric, esophageal, or gastroesophageal adenocarcinoma: trio-013/LOGiC: a randomized phase Ⅲ trial[J]. J Clin Oncol, 2016, 34(5):443-451. doi: 10.1200/JCO.2015.62.6598 [22] LYONS T G, KU G Y. Systemic therapy for esophagogastric cancer: targeted therapies[J]. Chin Clin Oncol, 2017, 6(5):48. doi: 10.21037/cco.2017.07.02 [23] SAHIN U, SCHULER M, RICHLY H, et al. A phase Ⅰ dose-escalation study of IMAB362 (Zolbetuximab) in patients with advanced gastric and gastro-oesophageal junction cancer[J]. Eur J Cancer, 2018, 100:17-26. doi: 10.1016/j.ejca.2018.05.007 [24] TÜRECI O, SAHIN U, SCHULZE-BERGKAMEN H, et al. A multicentre, phase Ⅱa study of zolbetuximab as a single agent in patients with recurrent or refractory advanced adenocarcinoma of the stomach or lower oesophagus: the MONO study[J]. Ann Oncol, 2019, 30(9):1487-1495. doi: 10.1093/annonc/mdz199 [25] SAHIN U, TÜRECI Ö, MANIKHAS G, et al. FAST: a randomised phase Ⅱ study of zolbetuximab (IMAB362) plus EOX versus EOX alone for first-line treatment of advanced CLDN18.2-positive gastric and gastro-oesophageal adenocarcinoma[J]. Ann Oncol, 2021, 32(5):609-619. doi: 10.1016/j.annonc.2021.02.005 [26] LORDICK F, AL-BATRAN S E, GANGULI A, et al. Patient-reported outcomes from the phase Ⅱ FAST trial of zolbetuximab plus EOX compared to EOX alone as first-line treatment of patients with metastatic CLDN18.2+ gastroesophageal adenocarcinoma[J]. Gastric Cancer, 2021, 24(3):721-730. doi: 10.1007/s10120-020-01153-6 [27] CREEMERS A, EBBING E A, PELGRIM T C, et al. A systematic review and meta-analysis of prognostic biomarkers in resectable esophageal adenocarcinomas[J]. Sci Rep, 2018, 8(1):13281. doi: 10.1038/s41598-018-31548-6 [28] IVESON T, DONEHOWER R C, DAVIDENKO I, et al. Rilotumumab in combination with epirubicin, cisplatin, and capecitabine as first-line treatment for gastric or oesophagogastric junction adenocarcinoma: an open-label, dose de-escalation phase 1b study and a double-blind, randomised phase 2 study[J]. Lancet Oncol, 2014, 15(9):1007-1018. doi: 10.1016/S1470-2045(14)70023-3 [29] CATENACCI D V T, TEBBUTT N C, DAVIDENKO I, et al. Rilotumumab plus epirubicin, cisplatin, and capecitabine as first-line therapy in advanced MET-positive gastric or gastro-oesophageal junction cancer (RILOMET-1): a randomised, double-blind, placebo-controlled, phase 3 trial[J]. Lancet Oncol, 2017, 18(11):1467-1482. doi: 10.1016/S1470-2045(17)30566-1 [30] CHUANG W Y, CHANG Y S, CHAO Y K, et al. Phosphorylated mTOR expression correlates with podoplanin expression and high tumor grade in esophageal squamous cell carcinoma[J]. Int J Clin Exp Pathol, 2015, 8(10):12757-12765. [31] HIRASHIMA K, BABA Y, WATANABE M, et al. Phosphorylated mTOR expression is associated with poor prognosis for patients with esophageal squamous cell carcinoma[J]. Ann Surg Oncol, 2010, 17(9):2486-2493. doi: 10.1245/s10434-010-1040-1 [32] LI S J, WANG Z Q, HUANG J, et al. Clinicopathological and prognostic significance of mTOR and phosphorylated mTOR expression in patients with esophageal squamous cell carcinoma: a systematic review and meta-analysis[J]. BMC Cancer, 2016, 16(1):1-17. doi: 10.1186/s12885-015-2026-y [33] ECKELMAN B P, SALVESEN G S, SCOTT F L. Human inhibitor of apoptosis proteins: why XIAP is the black sheep of the family[J]. EMBO Rep, 2006, 7(10):988-994. doi: 10.1038/sj.embor.7400795 [34] TU H L, COSTA M. XIAP’s profile in human cancer[J]. Biomolecules, 2020, 10(11):1493. doi: 10.3390/biom10111493 [35] PENG H, WANG L G, WANG X Z, et al. The correlation between XIAP gene polymorphisms and esophageal squamous cell carcinoma susceptibility and prognosis in a Chinese population[J]. Pathol Res Pract, 2017, 213(12):1482-1488. doi: 10.1016/j.prp.2017.10.008 [36] DIZDAR L, JÏ¿½NEMANN L, WERNER T, et al. Clinicopathological and functional implications of the inhibitor of apoptosis proteins survivin and XIAP in esophageal cancer[J]. Oncol Lett, 2018, 15(3):3779-3789. [37] JIN Y X, LU X Y, WANG M D, et al. X-linked inhibitor of apoptosis protein accelerates migration by inducing epithelial-mesenchymal transition through TGF-β signaling pathway in esophageal cancer cells[J]. Cell Biosci, 2019, 9(1):76. doi: 10.1186/s13578-019-0338-3 [38] IWAI Y, ISHIDA M, TANAKA Y, et al. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade[J]. Proc Natl Acad Sci USA, 2002, 99(19):12293-12297. doi: 10.1073/pnas.192461099 [39] DOI T, PIHA-PAUL S A, JALAL S I, et al. Safety and antitumor activity of the anti-programmed death-1 antibody pembrolizumab in patients with advanced esophageal carcinoma[J]. J Clin Oncol, 2018, 36(1):61-67. doi: 10.1200/JCO.2017.74.9846 [40] SHAH M A, KOJIMA T, HOCHHAUSER D, et al. Efficacy and safety of pembrolizumab for heavily pretreated patients with advanced, metastatic adenocarcinoma or squamous cell carcinoma of the esophagus[J]. JAMA Oncol, 2019, 5(4):546-550. doi: 10.1001/jamaoncol.2018.5441 [41] JANJIGIAN Y Y, MARON S B, CHATILA W K, et al. First-line pembrolizumab and trastuzumab in HER2-positive oesophageal, gastric, or gastro-oesophageal junction cancer: an open-label, single-arm, phase 2 trial[J]. Lancet Oncol, 2020, 21(6):821-831. doi: 10.1016/S1470-2045(20)30169-8 [42] KELLY R J, AJANI J A, KUZDZAL J, et al. Adjuvant nivolumab in resected esophageal or gastroesophageal junction cancer[J]. N Engl J Med, 2021, 384(13):1191-1203. doi: 10.1056/NEJMoa2032125 [43] DOKI Y, AJANI J A, KATO K, et al. Nivolumab combination therapy in advanced esophageal squamous-cell carcinoma[J]. N Engl J Med, 2022, 386(5):449-462. doi: 10.1056/NEJMoa2111380 [44] YANG W X, XING X B, YEUNG S C J, et al. Neoadjuvant programmed cell death 1 blockade combined with chemotherapy for resectable esophageal squamous cell carcinoma[J]. J Immunother Cancer, 2022, 10(1):e003497. doi: 10.1136/jitc-2021-003497 [45] WAGENER-RYCZEK S, SCHOEMMEL M, KRAEMER M, et al. Immune profile and immunosurveillance in treatment-naive and neoadjuvantly treated esophageal adenocarcinoma[J]. Cancer Immunol Immunother, 2020, 69(4):523-533. doi: 10.1007/s00262-019-02475-w [46] BANG Y J, CHO J Y, KIM Y H, et al. Efficacy of sequential ipilimumab monotherapy versus best supportive care for unresectable locally advanced/metastatic gastric or gastroesophageal junction cancer[J]. Clin Cancer Res, 2017, 23(19):5671-5678. doi: 10.1158/1078-0432.CCR-17-0025 [47] RALPH C, ELKORD E, BURT D J, et al. Modulation of lymphocyte regulation for cancer therapy: a phase Ⅱ trial of tremelimumab in advanced gastric and esophageal adenocarcinoma[J]. Clin Cancer Res, 2010, 16(5):1662-1672. doi: 10.1158/1078-0432.CCR-09-2870
计量
- 文章访问数: 6273
- HTML全文浏览量: 2328
- PDF下载量: 73
- 被引次数: 0