影响胆囊癌的炎症指标及治疗
Inflammatory Indexes Affecting Gallbladder Carcinoma and Its Treatment
DOI: 10.12677/ACM.2023.1351036, PDF, HTML, XML, 下载: 214  浏览: 335 
作者: 高肖肖:青海大学研究生院,青海 西宁;王宏宾*:青海大学附属医院普通外科学一科,青海 西宁
关键词: 胆囊癌炎症指数临床分期靶向治疗Gallbladder Carcinoma Inflammation Index Clinical Staging Targeted Therapy
摘要: 胆囊癌(GBC)是一种高度恶性的肿瘤,占胆道恶性肿瘤的80%~95%。胆囊癌在所有胆囊切除术中的发生率为0.2%~3%,在所有腹腔镜胆囊切除术中的发生率为0.09%~2%。手术切除是胆囊癌唯一的根治办法。手术应根据TNM分期决定手术切术范围,充分的术前评估对于提高根治率改善病人预后极为重要。胆囊癌的预后较差,总的5年生存率不到5%。在疾病早期,如果进行分期调整治疗,5年生存率可达75%。其发病率较低,症状不清,导致大多数患者在诊断时表现为晚期,导致其预后差。胆囊癌的全身化疗方案与胆管癌相似,包括吉西他滨和顺铂一线化疗,FOLFOX第二线化疗方案。目前正在进行的第三阶段临床试验可能会改变这种癌症的系统治疗模式。
Abstract: Gallbladder carcinoma (GBC) is a highly malignant tumor, accounting for 80%~95% of biliary ma-lignancies. The incidence of gallbladder cancer is 0.2%~3% in all cholecystectomies and 0.09%~2% in all laparoscopic cholecystectomies. Surgical excision is the only cure for gallbladder cancer. The scope of surgical resection should be determined according to TNM staging. Adequate preoperative evaluation is very important to improve the radical rate and prognosis of patients. Gallbladder can-cer has a poor prognosis, with an overall 5-year survival rate of less than 5%. In the early stages of the disease, with stage-adjusted treatment, the 5-year survival rate is 75%. Its low incidence and unclear symptoms result in the late stage of diagnosis in most patients, resulting in a poor progno-sis. The systemic chemotherapy regimen for gallbladder cancer is similar to that for cholangiocar-cinoma, including first-line chemotherapy with gemcitabine and cisplatin and second-line chemo-therapy with FOLFOX. A Phase 3 clinical trial currently underway could change the systematic treatment paradigm for this cancer.
文章引用:高肖肖, 王宏宾. 影响胆囊癌的炎症指标及治疗[J]. 临床医学进展, 2023, 13(5): 7429-7435. https://doi.org/10.12677/ACM.2023.1351036

1. 背景

青海地处我国青藏高原,平均海拔3000米以上,由于其特殊的地理位置及地理环境,导致该地区处于长期缺氧状态,长期生活于此会对人体各器官功能造成一定程度的伤害。机体处于长期缺氧、缺血的状态会损伤细胞,严重时可导致细胞的坏死,加之该地区总体经济条件、医疗条件、卫生条件差,这些都是该地区恶性肿瘤发病率较高的原因,研究表明,高海拔地区血小板计数值较低于低海拔地区 [1] ,长期生活在高海拔地区,人们的身体对低氧环境产生一些生理反应,例如红细胞增多(血细胞比容 > 65%)、血红蛋白的升高(女性Hb ≥ 19 g/dL,男性Hb ≥ 21 g/dL),主要表现为缺氧(氧分压 < 85%),血小板凝聚 [2] [3] ,高原地区的血小板水平相比于其他地区血小板数目水平,数值较低的原因主要为:① 随着海拔的不断升高,其大气压及氧分压逐渐降低,使得机体严重缺氧,导致骨髓产生血小板的能力降低,使血小板数量下降。② 长期生活于高原地区的人们的红细胞数目相对偏高,血小板数量的减少与继发性红细胞增多有一定程度的联系,红细胞数目的增多,使血液处于高度凝结状态,血小板易于黏附聚集,使血液中血小板的消耗增多,导致血小板计数进一步减少 [4] 。③ 大量衰老的红细胞需要经过脾脏的滤过作用,脾脏的破坏、清除功能的超负荷运作导致脾脏增大,扣留—部分血小板,导致血液中的血小板数目减少 [5] 。

2. 发病机制

胆囊结石症被认为是公认的胆囊癌的首要发病因素,胆囊结石和慢性胆囊炎是胆囊恶性肿瘤发生的重要危险因素 [6] 。胆囊结石长期刺激胆囊壁,导致胆囊壁内部急性或慢性炎症,最终使得胆囊壁瓷化及胆囊组织细胞变性,组织内存在异常细胞,被视为肿瘤的癌前病变 [6] ,晚期胆囊癌可扩散至肝脏,十二指肠、胰腺、结肠和胃等器官,慢性细菌感染、原发性硬化性胆管炎、胰胆管异常连接和几种类型的胆囊息肉等因素与胆囊癌的高风险相关。越来越多的证据表明,炎症途径与肿瘤的发生发展密切相关 [7] [8] [9] ,炎症反应在肿瘤的发生、发展和转移过程中发挥着重要的作用,同时也会影响机体的免疫监视以及对治疗的反应 [10] 。近年来应用炎症指标来预测肿瘤患者预后成为研究热点,炎症指标,如血小板相关指数,主要包括血小板计数(platelet count, PLT)、平均血小板体积(mean platelet volume, MPV)、血小板分布宽度(platelet distribution width, PDW)、血小板压积(plateletcrit, PCT)。中性粒细胞与淋巴细胞比值(NLR)、血小板与淋巴细胞比值(PLR)和格拉斯哥预后评分(GPS),已被证明对GBC有预后价值 [11] [12] [13] 。但这些指标较为单一,预测患者预后的作用有限,系统免疫炎症指数(Systemic immune inflammatory index, SII)作为由中性粒细胞、血小板、淋巴细胞组合而成的新型炎症指标已被证实与多种实体瘤预后相关 [14] [15] [16] [17] 。

3. 炎症指数

Zhang等 [18] 研究发现,与健康对照组相比胆囊癌患者MPV低、PDW高,同时检测到PDW与淋巴结转移显著相关,而未发现MPV与肿瘤大小、分化及转移有关联,其机制尚不清楚。因此,MPV和PDW是早期胆囊癌检测中有效的血液学参数。在预后方面,目前仅有一项研究 [19] 根据ROC曲线分析得出PLT的最佳临界点为178 × 109 L−1,PLT ≤ 178 × 109 L−1的患者总生存率明显高于PLT > 178 × 109 L−1的患者,由此得出PLT升高可高度提示胆囊癌预后不良。

4. 分期治疗

TNM分期由美国癌症联合委员会提出,目前常用的是2010年出版的第七版分期,TNM分期主要依据T (肿瘤浸润组织层次),N (淋巴结的转移方向),M (有无远处转移)来对患者进行具体分期(见表1),这种分期方式详细分类了淋巴结的分组,对于不同组患者的治疗也有了详细的治疗方案,可以更好地判断患者预后。

Table 1. Clinical staging of gallbladder carcinoma

表1. 胆囊癌的临床分期

TX:原发肿瘤无法判断;T0:无原发肿瘤证据;Tis:原位癌;T1:肿瘤侵犯固有层或肌层;T1a:肿瘤侵犯固有层;T1b:肿瘤侵犯肌层;T2:肿瘤侵犯肌层周围结缔组织,尚未侵及浆膜及肝脏;T3:肿瘤侵透浆膜或直接侵犯肝脏,或侵犯临近一个组织器官;T4:肿瘤直接侵犯肝动脉或门静脉主干或侵犯两个及以上的器官组织;N:淋巴结转移;NX:区域淋巴结转移无法评价;N0:无区域淋巴结转移;N1:胆囊管、胆总管或肝动脉、门静脉淋巴结转移;N2:腹主动脉、下腔静脉、肠系膜上动脉或腹腔干旁淋巴结转移;M:远处转移;MX:远处转移无法评估;M0:无远处转移;M1:有远处转移。

Tis期、T1a期胆囊癌

该分期胆囊癌属于原位癌、肿瘤侵犯黏膜固有层。此期患者通常有良好的5年预后,大多数临床报告显示,选择手术的T1肿瘤患者的生存率为75%或更高 [20] [21] 。

T1b期胆囊癌

该分期肿瘤侵犯到胆囊肌层,此期肿瘤发生淋巴结转移的可能性更大,单纯行胆囊切除无法达到根治的效果,此时需行胆囊癌根治 [22] [23] 。该术式要求切除胆囊及胆囊床周围肝脏(沿胆囊床周围2 cm肝脏的楔形切除)和区域性淋巴结清扫术。

T2期胆囊癌

肿瘤侵及肌周结缔组织,未超出浆膜或进入肝脏。胆囊和肝脏之间的边界是Glisson鞘延续形成的结缔组织,多达33%的T2肿瘤患者在邻近肝实质有微转移 [24] 。此期患者手术范围国际上大多建议行解剖性肝切除(肝IVb段 + V段)和区域淋巴结清扫术。Yoon等 [25] 与Shirobe等 [26] 研究发现使用腹腔镜对无明显肝脏受侵的T2期患者行根治性手术其5年生存率分别为83.3%和90.2%。

T3、T4期胆囊癌

T3期肿瘤穿透浆膜直接侵入肝脏或者侵犯一个邻近器官、结构。此期已属于进展期胆囊癌,侵袭性及转移性极强,为达到根治的目的,肝切除的程度应根据原发性胆囊肿瘤的解剖位置而定。若肿瘤直接侵犯肝脏,专家一致认为,至少应该切除4b/5段胆囊窝周围的肝脏,以获得最佳的边缘间隙 [24] 。

T4期患者肿瘤侵及门静脉或肝动脉主干,或直接侵入两个或更多肝外器官或结构。此期患者已属晚期,全身状况差,多伴有淋巴转移及远处转移。因此,多数行姑息性治疗,配合辅助治疗,预后仍较差。

有学者 [27] 认为,对于无远处转移(T4N0-1M0期)的胆囊癌患者,手术切除仍有希望达到根治的效果,有助于改善患者预后。Y Zhou等 [28] 的回顾性分析认为,联合肝、胰十二指肠切除术对晚期胆囊癌患者仍能提高其生存时间。T4期及T3期胆囊癌淋巴结转移率较高,达75%以上 [29] 。其远处转移率也较多,患者预后较差。由于组织受侵较多,中晚期胆囊癌手术常需进行联合脏器切除,包括右半肝切除、联合尾状叶的扩大切除,甚至肝动脉、门静脉重建,胰十二指肠切除等。其手术难度较大,R0切除较难实现,即使腹壁实现了微创,脏器也无法实现微创。因此普遍观点现阶段不推荐中晚期胆囊癌作为腹腔镜手术的适应证 [30] 。

5. 根治性切除

胆囊癌根治术的历史始与20世纪初的胆囊切除术联合胆囊窝楔形切除术(无区域淋巴结清扫)。然而,即使肿瘤局限于胆囊壁,该手术后的结局也令人失望,这可能是由于缺乏区域淋巴结切除术 [31] [32] 。1954年,Glenn等 [33] 首次提出了一种根治性切除术,即针对局限性胆囊癌的“根治性胆囊切除术”(Glenn手术),拟行局部淋巴结清扫术(门静脉淋巴结清扫术)。Pack等 [34] 于1955年和Fahim等 [35] 于1962年提倡根治性切除术,包括肝切除术和门静脉淋巴结清扫术,该策略仍然是美国国家综合癌症网络(NCCN)推荐的病理性T1b (pT1b)或更晚期胆囊癌的策略 [36] 。扩大根治性胆囊切除术适用于pT2肿瘤和一些pT3肿瘤伴局限性肝侵犯,前提是区域淋巴结病变限于中度(最多2个阳性淋巴结)。广泛的pT3病变、pT4病变或明显的淋巴结病变似乎超出了这种根治性手术的范围。

6. 全身放化疗

对于晚期或无法行手术治疗的胆囊癌患者,全身化疗的一线方案是联合吉西他滨和顺铂。然而,这种化疗方案的疗效有限,总体生存期中位数 < 1年 [26] 。在该III期试验中,纳入400例患者,分别采用吉西他滨联合顺铂与单药吉西他滨进行治疗。该试验报告中位无进展生存期为8个月,总生存期为11个月。在分析中有149名患者患有胆囊癌,与单用吉西他滨相比,双线化疗延长了这些患者的总生存期(HR0.61,范围,0.41~0.89)。这项研究确立了吉西他滨联合顺铂作为晚期不可切除胆囊癌的化疗标准。在胆囊癌中还研究了其他联合用药方案,包括吉西他滨加奥沙利铂(GEMOX),吉西他滨 + 卡培他滨和吉西他滨 + 替吉奥S1 [37] 。这些组合方案在一线治疗中的疗效可能与吉西他滨加顺铂相似。最近,报道了吉西他滨,顺铂和纳布–紫杉醇针对胆道癌患者(包括胆囊癌)的II期试验 [38] 。主要研究指标为无进展生存期,次要研究指标为总体生存期,总体缓解率和安全性。中位随访时间为12.2个月,中位无进展生存期为11.8个月(95%CI: 6.0~15.6)。部分反应和疾病控制率分别为43%和84%。中位总生存期为19.2个月(95%CI:13.2至无法估计)。研究显示肿瘤类型对治疗效果没有显着影响。中性粒细胞减少是化疗最常见的毒性反应(32%)。该方案目前正在晚期胆道癌的III期研究中进行研究。在欧盟,正在研究5-氟尿嘧啶,伊立替康和奥沙利铂(FOLFIRINOX)方案与吉西他滨和顺铂对晚期胆囊癌的治疗方案。这些研究的结果可能会完善当前胆囊癌一线治疗的选择 [39] 。直到最近,还没有建立胆囊癌标准的二线化疗方案。在一次基于20项研究针对胆囊癌二线治疗的系统评价中,加权总缓解率约为5.1%,中位无进展生存期为4个月 [40] 。在另一项研究中,从二线治疗开始以来,胆囊癌患者的平均总生存期为11个月(95%CI: 8.8~13.1),尤其是24.8%的患者平均总生存期为9.4个月(95%CI: 7.2~12.3) [41] 。最近,在ABC-06试验中,有162例患者(每组81例)被随机分为FOLFOX和支持治疗。21%患有胆囊癌 [42] 。在150次总体生存事件后,调整后的HR为0.69 (P = 0.031),FOLFOX方案患者受益更多。FOLFOX组的中位总体生存期为6.2个月,而仅支持治疗组的中位生存期为5.3个月。因此,FOLFOX被报告为胆囊癌第二线推荐化疗方案,与仅支持治疗相比,此方案提高了生存率。一旦完整的报告发布,更多细节可能有助于进一步指导此类方案的适用性。

7. 靶向治疗与免疫治疗

尽管针对胆囊癌的靶向治疗药物的临床试验落后于较常见的胃肠道癌,但EGFR,MEK,VEGFR和PI3-Kinase抑制剂的临床试验已经完成。II/III期随机试验未能揭示任何靶向药物联合化疗对胆道癌的优越性。目前胆囊癌靶向治疗研究的最主要的两个领域是Her2/neu和DNA修复基因改变 [43] [44] 。在胆囊癌人群中,这些遗传畸变的频率为10%至15%,使其成为特定抑制剂的潜在靶标。临床病理报告已经表明确实存在一部分胆囊癌患者在靶向治疗中获益。目前正在胆囊癌中探索采用检查点抑制剂的免疫疗法,初步数据表明单药检查点抑制剂的疗效中等。迄今为止,最大的研究是与Pembrolizumab进行的KEYNOTE-158研究 [45] 。该研究招募了104名BTC患者,其中58%为PD-L1+。据报道,此方案患者病情缓解率为6%,疾病稳定率为16%,缓解时间为6至16个月以上。免疫治疗组合可能比单药检查点抑制剂更有可能治疗胆囊癌。

8. 结论

在过去的十年中,胆囊癌的治疗方法已经有了很大的进步,目前,将胆囊癌和其他胆管癌纳入全身化疗试验是合理的。然而,在靶向和免疫治疗的情况下,必须考虑胆囊癌与其他胆管癌之间的遗传异质性。下一代测序研究表明Her2/neu,DNA修复和PI3-激酶基因改变可能是胆囊癌研究的潜在领域。正在进行的一线系统化疗、靶向治疗和免疫治疗试验可能会导致这种疾病的范式转变。

参考文献

[1] Wang, Z., Liu, H., Dou, M., Du, X., Hu, J., Su, N., Wang, Y., Zhang, R. and Li, C. (2017) The Quality Changes in Fresh Frozen Plasma of the Blood Donors at High Altitude. PLOS ONE, 12, e0176390.
https://doi.org/10.1371/journal.pone.0176390
[2] Frisancho, A.R. (2013) Developmental Functional Adaptation to High Altitude: Review. American Journal of Human Biology, 25, 151-168.
https://doi.org/10.1002/ajhb.22367
[3] Weitz, C.A. and Garruto, R.M. (2004) Growth of Han Migrants at High Altitude in Central Asia. American Journal of Human Biology, 16, 405-419.
https://doi.org/10.1002/ajhb.20042
[4] 张彦博, 汪源, 刘学良, 等. 人与高原[M]. 西宁: 青海人民出版社, 1996: 347.
[5] 张永海, 郁慕仪, 向兴利, 毛辉青. 不同海拔高度正常成人脾脏大小CT观测[J]. 高原医学杂志, 1999, 9(1): 23-27.
[6] Donohue, J.H., Na-gorney, D.M., Grant, C.S., Tsushima, K., Ilstrup, D.M. and Adson, M.A. (1990) Carcinoma of the Gallbladder: Does Radical Resection Improve Outcome. Archives of Surgery, 125, 237-241.
https://doi.org/10.1001/archsurg.1990.01410140115019
[7] Gukovsky, I., Li, N., Todoric, J., Gukovskaya, A. and Karin, M. (2013) Inflammation, Autophagy, and Obesity: Common Features in the Pathogenesis of Pancreatitis and Pancreatic Cancer. Gastroenterology, 144, 1199-1209.
https://doi.org/10.1053/j.gastro.2013.02.007
[8] Yang, L. and Karin, M. (2014) Roles of Tumor Suppressors in Regulating Tumor-Associated Inflammation. Cell Death & Differentiation, 21, 1677-1686.
https://doi.org/10.1038/cdd.2014.131
[9] Roxburgh, C.S. and McMillan, D.C. (2010) Role of Systemic Inflam-matory Response in Predicting Survival in Patients with Primary Operable Cancer. Future Oncology, 6, 149-163.
https://doi.org/10.2217/fon.09.136
[10] Grivennikov, S.I., Greten, F.R and Karin, M. (2010) Immunity, Inflamma-tion, and Cancer. Cell, 140, 883-899.
https://doi.org/10.1016/j.cell.2010.01.025
[11] Mady, M., Prasai, K., Tella, S.H., Yadav, S., Hallemeier, C.L., Rakshit, S., et al. (2020) Neutrophil to Lymphocyte Ratio as a Prognostic Marker in Metastatic Gallbladder Cancer. HPB, 22, 1490-1495.
https://doi.org/10.1016/j.hpb.2020.02.002
[12] Jansson, H., Cornillet, M., Björkström, N.K., Sturesson, C. and Sparrelid, E. (2020) Prognostic Value of Preoperative Inflflammatory Markers in Resectable Biliary Tract Can-cer—Validation and Comparison of the Glasgow Prognostic Score and Modifified Glasgow Prognostic Score in a West-ern Cohort. European Journal of Surgical Oncology, 46, 804-810.
https://doi.org/10.1016/j.ejso.2019.12.008
[13] Saqib, R., Pathak, S., Smart, N., Nunes, Q., Rees, J., Finch Jones, M. and Poston, G. (2018) Prognostic Signifificance of Pre-Operative Inflflammatory Markers in Resected Gallbladder Cancer: A Systematic Review. ANZ Journal of Surgery, 88, 554-559.
https://doi.org/10.1111/ans.14300
[14] Biswas, T., Kang, K.H., Gawdi, R., et al. (2020) Using the Systemic Im-mune-Inflammation Index (SII) as a Mid-Treatment Marker for Survival among Patients with Stage-III Locally Advanced Non-Small Cell Lung Cancer (NSCLC). International Journal of Environmental Research and Public Health, 17, 7995-8008.
https://doi.org/10.3390/ijerph17217995
[15] Jomrich, G., Gruber, E.S., Winkler, D., et al. (2020) Systemic Im-mune-Inflammation Index (SII) Predicts Poor Survival in Pancreatic Cancer Patients Undergoing Resection. Journal of Gastrointestinal Surgery, 24, 610-618.
https://doi.org/10.1007/s11605-019-04187-z
[16] Yatabe, S., Eto, K., Haruki, K., et al. (2020) Signification of Systemic Immune-Inflammation Index for Prediction of Prognosis after Resecting in Patients with Colorectal Cancer. In-ternational Journal of Colorectal Disease, 35, 1549-1555.
https://doi.org/10.1007/s00384-020-03615-w
[17] Ozbek, E., Besiroglu, H., Ozer, K., Horsanali, M.O. and Gorgel, S.N. (2020) Systemic Immune Inflammation Index is a Promising Non-Invasive Marker for the Prognosis of the Patients with Localized Renal Cell Carcinoma. International Urology and Nephrology, 52, 1455-1463.
https://doi.org/10.1007/s11255-020-02440-y
[18] Zhang, X., Niu, Y., Wang, X., et al. (2018) Mean Platelet Vol-ume and Platelet Distribution Width Are Associated with Gallbladder Cance. Asian Pacific Journal of Cancer Prevention, 19, 351-355.
[19] Wang, R.-T., Zhang, L.-Q., Mu, Y.-P., et al. (2015) Prognostic Significance of Preoperative Platelet Count in Patients with Gallbladder Cancer. World Journal of Gastroenterology, 21, 5303-5310.
https://doi.org/10.3748/wjg.v21.i17.5303
[20] Javle, M., Churi, C., Kang, H.C., et al. (2015) HER2/Neu-Directed Therapy for Biliary Tract Cancer. Journal of Hematology & Oncology, 8, Article No. 58.
https://doi.org/10.1186/s13045-015-0155-z
[21] Lowery, M.A., Ptashkin, R., Jordan, E., et al. (2018) Comprehen-sive Molecular Profiling of Intrahepatic and Extrahepatic Cholangiocarcinomas: Potential Targets for Intervention. Clini-cal Cancer Research, 24, 4154-4161.
https://doi.org/10.1158/1078-0432.CCR-18-0078
[22] 董家鸿, 王剑明, 曾建平. 胆囊癌诊断和治疗指南(2015版) [J]. 临床肝胆病杂志, 2016, 32(3): 411-419.
[23] Sachs, T.E., Akintorin, O. and Tseng, J. (2018) How Should Gallbladder Cancer Be Managed? Advances in Surgery, 52, 89-100.
https://doi.org/10.1016/j.yasu.2018.04.003
[24] Shirobe, T. and Maruyama, S. (2015) Laparoscopic Radical Chol-ecystectomy with Lymph Node Dissection for Gallbladder Carcinoma. Surgical Endoscopy, 29, 2244-2250.
https://doi.org/10.1007/s00464-014-3932-9
[25] Yoon, Y.-S., Han, H.-S., Cho, J.-Y., et al. (2015) Is Laparoscopy Contraindicated for Gallbladder Cancer? A 10-Year Prospective Cohort Study. Journal of the American College of Sur-geons, 221, 847-853.
https://doi.org/10.1016/j.jamcollsurg.2015.07.010
[26] Valle, J., Wasan, H., Palmer, D.H., et al. (2010) Cisplatin plus Gemcitabine versus Gemcitabine for Biliary Tract Cancer. New England Journal of Medicine, 362, 1273-1281.
https://doi.org/10.1056/NEJMoa0908721
[27] Chen, C., Geng, Z., Shen, H., et al. (2016) Long-Term Outcomes and Prognostic Factors in Advanced Gallbladder Cancer: Focus on the Advanced T Stage. PLOS ONE, 11, e0166361.
https://doi.org/10.1371/journal.pone.0166361
[28] Zhou, Y., Zhang, Z., Wu, L. and Li, B. (2016) A Systematic Review of Safety and Efficacy of Hepatopancreatoduodenectomy for Biliary and Gallbladder Cancers. HPB, 18, 1-6.
https://doi.org/10.1016/j.hpb.2015.07.008
[29] Goetze, T.O. (2015) Gallbladder Carcinoma: Prognostic Factors and Therapeutic Options. World Journal of Gastroenterology, 21, 12211-12217.
https://doi.org/10.3748/wjg.v21.i43.12211
[30] 刘杰, 张成武. 腹腔镜技术在胆道恶性肿瘤外科治疗中的应用[J]. 肝胆胰外科杂志, 2018, 30(2): 89-93.
[31] Sheinfeld, W. (1947) Cholecystectomy and Partial Hepatectomy for Carcinoma of the Gall Bladder with Local Liver Extension. Surgery, 22, 48-58.
[32] Glenn, F. and Hays, D.M. (1953) Carcinoma of the Extrahepatic Biliary Tract. Surgical Clinics of North America, 33, 479-492.
https://doi.org/10.1016/S0039-6109(16)33892-0
[33] Glenn, F. and Hays, D.M. (1954) The Scope of Radical Surgery in the Treatment of Malignant Tumors of the Extrahepatic Biliary Tract. Surgery, Gynecology and Obstetrics, 99, 529-541.
[34] Pack, G.T., Miller, T.R. and Brasfield, R.D. (1955) Total Right Hepatic Labectomy for Cancer of the Gallbladder: Report of Three Cases. Annals of Surgery, 142, 6-16.
https://doi.org/10.1097/00000658-195507000-00002
[35] Fahim, R.B., McDonald, J.R., Richards, J.C. and Ferris, D.O. (1962) Carcinoma of the Gallbladder: A Study of Its Modes of Spread. Annals of Surgery, 156, 114-124.
https://doi.org/10.1097/00000658-196207000-00021
[36] Benson 3rd, A.B., Abrams, T.A., Ben-Josef, E., Bloom-ston, P.M., Botha, J.F., Clary, B.M., Covey, A., Curley, S.A., D’Angelica, M.I., Davila, R., Ensminger, W.D., Gibbs, J.F., Laheru, D., Malafa, M.P., Marrero, J., Meranze, S.G., Mulvihill, S.J., Park, J.O., Posey, J.A., Sachdev, J., Salem, R., Sigurdson, E.R., Sofocleous, C., Vauthey, J.N., Venook, A.P., Goff, L.W., Yen, Y. and Zhu, A.X. (2009) NCCN Clinical Practice Guidelines in Oncology: Hepatobiliary Cancers. Journal of the National Comprehensive Cancer Net-work, 7, 350-391.
https://doi.org/10.6004/jnccn.2009.0027
[37] Javle, M., Rashid, A., Churi, C., et al. (2014) Molecular Characteri-zation of Gallbladder Cancer Using Somatic Mutation Profiling. Human Pathology, 45, 701-708.
https://doi.org/10.1016/j.humpath.2013.11.001
[38] Shroff, R.T., Javle, M.M., Xiao, L., et al. (2019) Gemcitabine, Cisplatin, and nab-Paclitaxel for the Treatment of Advanced Biliary Tract Cancers: A Phase 2 Clinical Trial. JAMA On-cology, 5, 824-830.
https://doi.org/10.1001/jamaoncol.2019.0270
[39] Phelip, J.-M., Edeline, J., Blanc, J.F., et al. (2019) Modified FOLFIRINOX versus CisGem First-Line Chemotherapy for Locally Advanced Non Resectable or Metastatic Biliary Tract Cancer (AMEBICA)-PRODIGE 38: Study Protocol for a Randomized Controlled Multicenter Phase II/III Study. Digestive and Liver Disease, 51, 318-320.
https://doi.org/10.1016/j.dld.2018.11.018
[40] Lamarca, A., Hubner, R.A., David Ryder, W. and Valle, J.W. (2014) Second-Line Chemotherapy in Advanced Biliary Cancer: A Systematic Review. Annals of Oncology, 25, 2328-2338.
https://doi.org/10.1093/annonc/mdu162
[41] Goff, L.W., Lowery, M.A., Jordan, E., et al. (2016) Second-Line Chemotherapy (CTx) Outcomes in Advanced Biliary Cancers (ABC): A Retrospective Multicenter Analysis. Journal of Clinical Oncology, 34, e15636.
https://doi.org/10.1200/JCO.2016.34.15_suppl.e15636
[42] Lamarca, A., Palmer, D.H., Wasan, H.S., et al. (2019) A Randomised Phase III, Multi-Centre, Open-Label Study of Active Symptom Control (ASC) Alone or ASC with Ox-aliplatin / 5-FU Chemotherapy (ASC+mFOLFOX) for Patients (pts) with Locally Advanced/Metastatic Biliary Tract Cancers (ABC) Previously-Treated with Cisplatin/Gemcitabine (CisGem) Chemotherapy. Journal of Clinical Oncology, 37, e4003.
https://doi.org/10.1200/JCO.2019.37.15_suppl.4003
[43] Lamarca, A., Barriuso, J., McNamara, M.G. and Valle, J.W. (2018) Biliary Tract Cancer: State of the Art and Potential Role of DNA Damage Repair. Cancer Treatment Reviews, 70, 168-177.
https://doi.org/10.1016/j.ctrv.2018.09.002
[44] Jain, A., Kwong, L.N. and Javle, M. (2016) Genomic Profiling of Biliary Tract Cancers and Implications for Clinical Practice. Current Treatment Options in Oncology, 17, Ar-ticle No. 58.
https://doi.org/10.1007/s11864-016-0432-2
[45] Ueno, M., Chung, H.C., Nagrial, A., et al. (2018) Pembrolizumab for Advanced Biliary Adenocarcinoma: Results from the Multicohort, Phase II KEYNOTE-158 Study. Annals of Oncology, 29, VIII210.
https://doi.org/10.1093/annonc/mdy282.009