中国临床肿瘤学会(CSCO)黑色素瘤诊疗指南 2017.V1(txt+pdf+epub+mobi电子书下载)


发布时间:2020-06-04 02:50:08

点击下载

作者:中国临床肿瘤学会指南工作委员会

出版社:人民卫生出版社

格式: AZW3, DOCX, EPUB, MOBI, PDF, TXT

中国临床肿瘤学会(CSCO)黑色素瘤诊疗指南 2017.V1

中国临床肿瘤学会(CSCO)黑色素瘤诊疗指南 2017.V1试读:

前言

基于循证医学证据和精准医学基本原则制定中国常见癌症的诊断和治疗指南,是中国临床肿瘤学会(CSCO)的基本任务之一。近年来,国际上指南的制定出现了一个新的趋向,即基于资源可及性的指南,这尤其适合发展中国家和地区差异性显著的国家。中国是一个幅员辽阔但地区发展不平衡的发展中国家,CSCO指南必须兼顾到地区发展不平衡、药物和治疗措施的可及性以及肿瘤治疗的价值三个方面。因此,CSCO指南形成了这样的特点,每一个临床问题的诊治指南,分为基本策略和可选策略两部分。基本策略属于可及性好的普适性诊治措施,肿瘤治疗价值相对稳定;可选策略多属于在国际或国内已有高级别证据,但可及性差或效价比超出国人承受能力的药物或治疗措施,如机器人手术。对于一些欧美已批准上市但我国尚不可及的药物,指南专门列出作为临床医生参考。 CSCO指南工作委员会相信,基于资源可及性的指南,是目前最适合我国国情的指南,我们期待大家的反馈并将持续改进,保持CSCO指南的时效性。中国临床肿瘤学会指南工作委员会一 影像和分期诊断影像和分期诊断上述证据级别全部为2A类证据注释

对于临床初步判断无远处转移的黑色素瘤患者,活检一般建议完整切除,不建议穿刺活检或局部切除,部分切取活检不利于组织学诊断和厚度测量,增加了误诊和错误分期风险。如病灶面积过大或已有远处转移需要确诊的,可行局部切取活检。

前哨淋巴结活检是病理分期评估区域淋巴结是否转移的手段。肿瘤厚度>1mm推荐行前哨淋巴结活检。通常不推荐对原发肿瘤厚度≤0.75mm的患者行前哨淋巴结活检,传统的危险因素例如溃疡、高有丝分裂率及淋巴血管侵犯在这些患者前哨淋巴结活检中的指导意义有限。这些危险因素一旦出现,是否行前哨淋巴结活检需考虑患者的个人意愿。病灶厚度为0.76~1mm的可结合临床考虑行前哨淋巴结活检[3-6]。

如临床怀疑区域淋巴结转移,建议首选淋巴结超声,淋巴结转移[7]的超声表现特征:淋巴结呈类圆形,髓质消失,边缘型血流。参考文献

1.Xing Y, Bronstein Y, Ross MI, et al.Contemporary diagnostic imaging modalities for the staging and surveillance of melanoma patients: a meta-analysis.J Natl Cancer Inst, 2011, 103(2): 129-142.

2.Clark PB, Soo V, Kraas J, et al.Futility of fluorodeoxy F-18 positron emission tomography in initial evaluation of patients with T2 to T4 melanoma.Arch Surg, 2006, 141(3): 284-288.

3.Testori A, Mozziuo N.Surgical techniques of melanoma and sentinel node biopsy.Semin Oncol, 2002,29(4):325-328.

4.Mocellin S, Hoon DS, Pilati P, et al.Sentinel lymph node molecular ultrastaging in patients with melanoma: asystematicreview and meta-analysisofprognosis.JClin Oncol, 2007, 25 (12):1588-1595.

5.Donald L.M, Thompson JF, Alistair JC, et al.Sentinel-node biopsy or nodal observation in melanoma.New Engl Journal Med, 2006, 355: 1307-1317.

6.Morton DL, Thompson JF, Cochran AJ, et al.Final trial report of sentinel-node biopsy versus nodal observation in melanoma.N Engl J Med, 2014, 370: 599-609.

7.Voit C, Van Akkooi AC, Schafer-Heserberg G, et al.Ultrasound morphology criteria predict metastatic disease of the sentinel nodes in patients with melanoma.J Clin Oncol, 2010, 28(5): 847-852.二 病理学诊断和分子分型病理学诊断和分子分型上述证据级别全部为2A类证据注释

1.送检标本处理:标本需完整送检,手术外科医生做好标记切缘,10%甲醛溶液固定标本达6~48h。

2.专家组建议病理报告中必须包括的内容为肿瘤厚度、是否溃疡和有丝分裂率,这三个指标与T分期直接相关,也是判断预后的3个[1-4]最重要的特征。

3.有丝分裂率(mitotic rate,MR)是肿瘤增殖的指标,记为每平方毫米的有丝分裂细胞数。最新的AJCC分期指南建议运用“热点”[4,5]技术推算有丝分裂率。Barnhill等比较了MR与溃疡作为局限期黑色素瘤预后的重要性,对MR和溃疡、肿瘤厚度进行多因素分析,发现MR(<1、1~6、>6)是最重要的独立预后因素。另外还有很多研[6-9]究也证实了MR是皮肤黑色素瘤的重要预后因子。MR≥1的患者疾病特异生存期(DSS较差)是预后的独立不良因素,在浸润厚度≤1mm的患者中尤为显著。在该组患者中,MR可以代替Clark分级,以[10,11]区分ⅠA期和ⅠB期 。

4.对切缘阳性的,需描述范围(如是原位还是浸润性);切缘阴性的,美国病理学家协会(CAP)指南要求以毫米为单位报告显微镜下测量的肿瘤与切缘的横向或纵向距离。

5.微卫星灶指直径大于0.05mm,距离原发灶至少0.3mm的真皮网状层、脂膜或脉管中的瘤巢,与区域淋巴结转移相关性高。初次活检或扩大切除标本中出现局部微卫星灶分期归为N2c(ⅢB期);出现微卫星灶的患者需要做前哨淋巴结活检,若前哨淋巴结阳性,则分期[12,13]为N3(ⅢC期) 。

6.建议所有患者治疗前都做基因检测,目前成熟的靶点是BRAF、CKIT和NRAS,与预后、分子分型和晚期治疗有关。黑色素瘤依基因变异可分为4种基本类型:①肢端型;②黏膜型;③慢性日光损伤型(CSD);④非慢性日光损伤型(non-CSD,包括原发病灶不明型)。其中日光损伤型主要包括头颈部和四肢黑色素瘤,日光暴露较多,高倍镜下可观察到慢性日光晒伤小体。肢端型和黏膜型发生KIT基因变异较多,其次为BRAF突变;非慢性日光损伤型,如躯干黑色素瘤,大部分发生BRAF基因V600E突变(60%)或NRAS突变[14-17](20%)。我国502例原发黑色素瘤标本KIT基因检测结果显示总体突变率为10.8%,基因扩增率为7.4%;其中肢端型、黏膜型、慢性日光损伤型、非慢性日光损伤型和原发灶不明型分别为11.9%和7.3%,9.6%和10.2%,20.7%和3.4%,8.1%和3.2%及7.8%和5.9%。我国468例原发黑色素瘤标本BRAF突变率为25.9%,肢端和黏膜黑色素瘤的突变率分别为17.9%和12.5%,其中15号外显子的V600E是最常见的突变位点(87.3%)。多因素分析显示KIT基因和BRAF基因突变均是黑色素瘤的独立预后因素,危险系数分别为1.989(95%CI 1.263~[18,3.131)和1.536(95%CI 1.110~2.124), P分别为0.003和0.0119]。参考文献

1.Balch CM, Gershenwald JE, Soong SJ, et al.Final version of 2009 AJCC melanoma staging and classification.J Clin Oncol, 2009, 27(36): 6199-6206.

2.Thompson JF, Soong SJ, Balch CM, et al.Prognostic significance of mitotic rate in localized primary cutaneous melanoma:an analysis of patients in the multi-institutional American Joint Committee on Cancer melanoma staging database.J Clin Oncol, 2011, 29(16): 2199-2205.

3.Balch CM, Gershenwald JE, Soong SJ, et al.Multivariate analysis of prognostic factors among 2313 patients with stageⅢmelanoma: comparison of nodal micrometastases versus macrometastases.J Clin Oncol, 2010, 28(14): 2452-2459.

4.Edge SB, Carducci M, Byrd DR.AJCC Cancer Staging Manual.7th ed.New York: Springer-Verlag New York, LLC, 2009.

5.Piris A, Mihm MC, Duncan LM.AJCC melanoma staging update: impact on dermatopathology practice and patient management.J Cutan Pathol, 2011, 38(5): 394-400.

6.Azzola MF, Shaw HM, Thompson JF, et al.Tumor mitotic rate is a more powerful prognostic indicator than ulceration in patients with primary cutaneous melanoma:an analysis of 3661 patients from a single center.Cancer, 2003, 97(6): 1488-1498.

7.Francken AB, Shaw HM, Thompson JF, et al.The prognostic importance of tumor mitotic rate confirmed in 1317 patients with primary cutaneous melanoma and long follow-up.Ann Surg Oncol, 2004,11(4):426-433.

8.Gimotty PA, Elder DE, Fraker DL, et al.Identification of high-risk patients among those diagnosed with thin cutaneous melanomas.J Clin Oncol, 2007, 25(9): 1129-1134.

9.Thompson JF, Soong SJ, Balch CM, et al.Prognostic significance of mitotic rate in localized primary cutaneous melanoma:an analysis of patients in the multi-institutional American Joint Committee on Cancer melanoma staging database.J Clin Oncol, 2011, 29(16): 2199-2205.

10.Paek SC, Griffith KA,Johnson TM, et al.The impact of factors beyond Breslow depth on predicting sentinel lymph node positivity in melanoma.Cancer, 2007, 109(1): 100-108.

11.Sondak VK, Taylor JM, Sabel MS, et al.Mitotic rate and younger age are predictors of sentinel lymph node positivity: lessons learned from the generation of a probabilistic model.Ann Surg Oncol, 2004,11(3):247-258.

12.College of American Pathologists.Protocol for the examination of specimens from patients with melanoma of the skin.2013.

13.Harrist TJ, Rigel DS, Day CL Jr, et al. “Microscopic satellites” are more highly associated with regional lymph node metastases than is primary melanoma thickness.Cancer, 1984, 53(10):2183-2187.

14.Cancer Genome Atlas Network.Genomic Classification of Cutaneous Melanoma.Cancer Genome Atlas Network.Cell, 2015, 161(7): 1681-1696.

15.High WA, Robinson WA.Genetic mutations involved in melanoma: a summary of our current understanding.Adv Dermatol, 2007, 23: 61-79.

16.Curtin JA, Busam K, Pinkel D, et al.Somatic activation of KIT in distinct subtypes of melanoma.J Clin Oncol, 2006, 24(26): 4340-4346.

17.Curtin JA, Fridlyand J, Kageshita T, et al.Distinct sets of genetic alterations in melanoma.N Engl J Med, 2005, 353(20): 2135-2147.

18.Kong Y, Si L, Zhu Y, et al.Large-scale analysis of KIT aberrations in Chinese patients with Melanoma.Clin Cancer Res, 2011, 17(7): 1684-1691.

19.Si L, Kong Y, Xu X, et al.Prevalence of BRAF V600E mutation in Chinese melanoma patients:large scale analysis of BRAF and NRAS mutations in a 432-case cohort.Eur J Cancer, 2012, 48(1):94-100.三 基于原发部位、分期和分子分型的综合治疗(皮肤来源)(一)0期、ⅠA、ⅠB期恶性黑色素瘤的治疗注释

1.外科切缘是指外科医生进行手术时测量到的临床切缘,而不是病理医生测量的大体或病理切缘。可根据患者具体的原发病灶解剖结[1-6]构和功能对切缘进行调整。参看附表6手术切缘。通常需要根据活检病理报告的厚度来决定进一步扩大切除的切缘。对于活检病理未能报告明确深度或病灶巨大的患者,可考虑直接扩切2cm。

2.原位癌:对于面积较大的原位癌,如雀斑痣样黑色素瘤,可能需要大于0.5cm的切缘才能保证完整切除。对于部分切缘阳性无法手术的患者,可行咪喹莫特外敷或局部放疗(2类证据)。

3.外科手术标准:皮肤恶性黑色素瘤的切除要求完整切除皮肤以及深达肌筋膜的皮下组织。通常无须切除筋膜,但对浸润较深的原发[7]灶(>4mm)可考虑切除筋膜。[8-9]

4.危险因素:包括溃疡、高有丝分裂率及淋巴血管侵犯等。

5.厚度>1mm的患者可考虑进行前哨淋巴结活检,可于完整切除的同时进行亦可分次进行。鉴于我国皮肤黑色素瘤的溃疡比例发生率[10]高达60%以上,且伴有溃疡发生的皮肤黑色素瘤预后较差,故当活检技术或病理检测技术受限从而无法获得可靠的浸润深度时,合并溃疡的患者均推荐前哨淋巴结活检。前哨淋巴结活检有助于准确获得[11]N分期,提高患者的无复发生存率,但对总生存期无影响。如果发现前哨淋巴结阳性,一般应及时进行淋巴结清扫。前哨淋巴结内低肿瘤负荷(前哨淋巴结的转移灶直径<0.1mm)的患者无须接受扩大淋[12]巴结清扫。参考文献

1.Cascinelli N.Margin of resection in the management of primary melanoma.Semin Surg Oncol, 1998,14(4):272-275.

2.Cohn-Cedermark G, Rutqvist LE, Andersson R, et al.Long term results of a randomized study by the Swedish Melanoma Study Group on 2-cm versus 5-cm resection margins for patients with cutaneous melanoma with a tumor thickness of 0.8-2.0mm.Cancer, 2000, 89(7): 1495-1501.

3.Khayat D, Rixe O, Martin G, et al.Surgical margins in cutaneous melanoma(2 cm versus 5 cm for lesions measuring less than 2.1-mm thick).Cancer, 2003, 97(8): 1941-1946.

4.Balch CM, Soong SJ, Smith T, et al.Long-term results of a prospective surgical trial comparing 2 cm vs.4 cm excision margins for 740 patients with 1-4 mm melanomas.Ann Surg Oncol, 2001, 8(2):101-108.

5.Thomas JM,Newton-Bishop J,A’ Hern R, et al.Excision margins in high-risk malignant melanoma.N Engl J Med, 2004, 350(8): 757-766.

6.Gillgren P, Drzewiecki KT,Niin M,et al.2-cm versus 4-cm surgical excision margins for primary cutaneous melanoma thicker than 2 mm: a randomised, multicentre trial.Lancet, 2011, 378(9803):1635-1642.

7.Kimbrough CW,McMasters KM,Davis EG.Principles of surgical treatment of malignant melanoma.Surg Clin North Am, 2014, 94(5): 973-988, vii.

8.Mitteldorf C, Bertsch HP, Jung K, et al.Sentinel Node Biopsy Improves Prognostic Stratification in Patients with Thin(pT1)Melanomas and an Additional Risk Factor.Ann Surg Oncol, 2014, 21(7):2252-2258.

9.Wong SL, Brady MS, Busam KJ, et al.Results of sentinel lymph node biopsy in patients with thin melanoma.Ann Surg Oncol, 2006, 13(3): 302-309.

10.Chi Z, Li S, Sheng X, et al.Clinical presentation, histology, and prognoses of malignant melanoma in ethnic Chinese: A study of 522 consecutive cases.BMC Cancer, 2011, 11: 85.

11.Morton DL, Thompson JF, Cochran AJ, et al.Final trial report of sentinel-node biopsy versus nodal observation in melanoma.N Engl J Med, 2014, 370(7): 599-609.

12.Van der Ploeg AP, Van Akkooi AC, Rutkowski P, et al.Prognosis in patients with sentinel node-positive melanoma is accurately defined by the combined Rotterdam tumor load and Dewar topography criteria.J Clin Oncol, 2011, 29(16): 2206-2214.(二)ⅡA、ⅡB期恶性黑色素瘤的治疗注释

对于ⅡB~Ⅲ期的高危黑色素瘤,推荐大剂量干扰素辅助治疗。多项临床研究证实大剂量干扰素α-2b能延长患者的无复发生存期,但[1-3][4]对总生存的影响尚不明确。meta分析亦证实上述观点。关于干[5-11]扰素的最优剂量和给药时间一直在探讨中。针对我国患者,推荐2采用改良的干扰素剂量1500万IU/m,d1-5×4w+900万IU,tiw×48w治[10]2疗1年,亦可遵循NCCN指南推荐的标准剂量(2000万IU/m,d1-52×4w,1000万IU/m,tiw×48w。对于高龄、有合并症或无法耐受的患[5]者,亦可选用1个月大剂量干扰素代替1年干扰素。参考文献

1.Kirkwood JM, Strawderman MH, Ernstoff MS, Smith TJ, Borden EC, Blum RH.Interferon alfa-2b adjuvant therapy of high-risk resected cutaneous melanoma:the Eastern Cooperative Oncology Group Trial EST 1684.J Clin Oncol, 1996, 14(1): 7-17.

2.Kirkwood JM,Ibrahim JG, Sondak VK, et al.High-and low-dose interferon alfa-2b in high-risk melanoma: first analysis of intergroup trial E1690/S9111/C9190.J Clin Oncol, 2000, 18 (12):2444-2458.

3.Kimbrough CW,McMasters KM,Davis EG.High-dose interferon alfa-2b significantly prolongs relapsefree and overall survival compared with the GM2-KLH/QS-21 vaccine in patients with resected stageⅡB-Ⅲ melanoma: results of intergroup trial E1694/S9512/C509801.Surg Clin North Am, 2014, 94(5):973-988,vii.

4.Mocellin S, Pasquali S, Rossi CR, et al.Interferon alpha adjuvant therapy in patients with high-risk melanoma:a systematic review and meta-analysis.J Natl Cancer Inst, 2010, 102(7): 493-501.

5.Pectasides D,Dafni U,Bafaloukos D,et al.Randomized phaseⅢ study of 1 month versus 1 year of adjuvant high-dose interferon alfa-2b in patients with resected high-risk melanoma.J Clin Oncol, 2009,20; 27(6): 939-944.

6.Cascinelli N, Bufalino R, Morabito A, et al.Results of adjuvant interferon study in WHO melanoma programme.Lancet, 1994, 343: 913-914.

7.Hauschild A,Weichenthal M,rass K,et al.efficacy of lowdose interferon{alpha}2a 18 versus 60 months of treatment inpatients with primary melanoma of≥1.5mm tumor thickness: results of a randomized phase Ⅲ DecoG trial.J Clin Oncol, 2010, 28: 841-846.

8.Eggermont AM, Suciu S, MacKier, et al.Post-surgery adjuvant therapy with intermediate doses of interferon alfa 2b versusobservation in patients with stageⅡb/Ⅲ melanoma(eortc18952): randomised controlled trial.Lancet, 2005, 366: 1189-1196.

9.Eggermont AM, Suciu S,Santinami M, et al.Adjuvant therapy with pegylated interferon alfa-2b versus observation alone inresected stageⅢmelanoma: final results of eortc 18991,a randomised phaseⅢtrial.Lancet, 2008, 372: 117-126.

10.Mao L,Si L,Chi Z,et al.A randomised phaseⅡ trial of 1 month versus 1 year of adjuvant high-dose interferon α-2b in high risk acral melanoma patients.Eur J Cancer, 2011, 47(10): 1498-1503.

11.Agarwala SS,Lee SJ, Yip W, et al.PhaseⅢ Randomized Study of 4 Weeks of High-Dose Interferonα-2b in Stage T2bNO, T3a-bNO, T4a-bNO, and T1-4N1a-2a(microscopic)Melanoma: A Trial of the Eastern Cooperative Oncology Group-American College of Radiology Imaging Network Cancer Research Group(E1697).J Clin Oncol, 2017, 35(8): 885-892.(三)ⅢA、ⅢB、ⅢC期恶性黑色素瘤的治疗ⅢA、ⅢB、ⅢC期恶性黑色素瘤的治疗(续表)注释[1]

1.淋巴结清扫原则

1)区域淋巴结须充分清扫。

2)受累淋巴结基部须完全切除。

3)通常来说,切除并检出的淋巴结个数如下:腹股沟≥10个,腋窝≥15个,颈部≥15个。

4)在腹股沟区,如临床发现股浅淋巴结或转移淋巴结数≥3个,选择性行髂窝和闭孔区淋巴结清扫。

5)如果盆腔影像学检查提示有盆腔淋巴结转移,或术中Cloquet(股管)淋巴结活检病理阳性,需行髂窝和闭孔区淋巴结清扫。

6)头颈部皮肤原发的患者,如超声怀疑有区域淋巴结转移,推荐引流区颈部淋巴结清扫。如受客观条件所限仅行转移淋巴结切除,需采用淋巴结超声或CT、MRI严密监测淋巴结复发情况。[2-4]

2.淋巴结辅助放疗原则(2B类证据)

1)LDH<1.5倍正常值上限和

2)淋巴结转移情况

a)淋巴结结外侵犯。

b)腮腺受累淋巴结≥1个,无论大小。

c)颈部受累淋巴结≥2个,和(或)受累淋巴结大小≥3cm。

d)腋窝受累淋巴结≥2个,和(或)受累淋巴结大小≥4cm。

e)腹股沟受累淋巴结≥3个,和(或)受累淋巴结大小≥4cm。

需同时满足第1)条和第2)条中的任意一条。辅助放疗可提高局部控制力,但未能改善无复发生存时间或总生存时间,可能增加不良反应(水肿、皮肤、皮下组织纤维化、疼痛等)。仅推荐用于以控制局部复发为首要目的的患者,或在无法进行全身性辅助治疗的患者中作为备选。目前缺乏中国循证医学证据。

3.2015年美国FDA批准ipilimumab用于Ⅲ期黑色素瘤术后的辅助[5]治疗,国内尚未上市,且缺乏与干扰素的直接对照。

4.移行转移(in-transit metastasis)指原发病灶(周围直径2cm以外)与区域淋巴结之间,通过淋巴管转移的皮肤、皮下或软组织转移结节。

5.卫星灶(satellite)指在原发病灶周围直径2cm内发生的转移结节。

6.隔离热灌注化疗(ILP)和隔离热输注化疗(ILI)(2A类证据)主要用于肢体移行转移的治疗。ILI是一种无氧合、低流量输注化疗药物的局部治疗手段,通过介入动静脉插管来建立化疗通路输注美法仑(马法兰)。有研究称,Ⅲ期MM有效率约80%,CR率达31%[6-8]~63%。

7.瘤体内药物注射:其作用机制为局部消融肿瘤和诱导全身抗肿瘤免疫,目前可选用的药物包括干扰素、白介素、BCG(2B类证据)。T-VEC为HSV-1衍生的溶瘤免疫治疗药物,已被美国FDA批准[9]用于治疗黑色素瘤,并可诱导远处部位肿瘤细胞死亡。另一种瘤体[10]内注射的药物PV-10,尚在临床试验中。

8.全身治疗参见Ⅳ期。参考文献

1.Kimbrough CW,McMasters KM,Davis EG.Principles of surgical treatment of malignant melanoma.Surg Clin North Am,2014,94(5): 973-988, vii.

2.Burmeister BH, Henderson MA, Ainslie J, et al.Adjuvant radiotherapy versus observation alone for patients at risk of lymph-node field relapse after therapeutic lymphadenectomy for melanoma:a randomised trial.Lancet Oncol, 2012, 13(6): 589-597.

3.Henderson MA, Burmeister BH, Ainslie J, et al.Adjuvant lymph-node field radiotherapy versus observation only in patients with melanoma at high risk of further lymph-node field relapse after lymphadenectomy(ANZMTG 01.02/TROG 02.01):6-year follow-up of a phase

4.Randomised controlled trial.Lancet Oncol, 2015, 16(9): 1049-1060.

5.Eggermont AM, Chiarion-Sileni V, Grob JJ, et al.Adjuvant ipilimumab versus placebo after complete resection of high-risk stageⅢ melanoma(EORTC 18071): a randomised, double-blind, phase 3 trial.Lancet Oncol, 2015, 16(5): 522-530.

6.Thompson JF, Hunt JA, Shannon KF, et al.Frequency and duration of remission after isolated limb perfusion for melanoma.Arch Surg, 1998, 132(8): 903-907.

7.Beasley GM, Caudle A, Petersen RP, et al.A multi-institutional experience of solated limb infusion:defining response and toxicity in the US.J Am Coll Surg, 2009, 208(5): 706-715; discussion 715-717.

8.Boesch CE, Meyer T, Waschke L, et al.Long-term outcome of hyperthermic isolated limb perfusion(HILP)in the treatment of locoregionally metastasized malignant melanoma of the extremities.Int J Hyperthermia, 2010, 26(1): 16-20.

9.Andtbacka RH, Kaufman HL, Collichio F, et al.Talimogene Laherparepvec Improves durable response rate in patients with advanced melanoma.J Clin Oocol, 2015, 33(25): 2780-2788.

10.Thompson JF, Agarwala SS, Smithers BM, et al.Phase 2 study of intralesional PV-10 in refractory metastatic melanoma.Ann Surg Oncol, 2015, 22(7): 2135-2142.(四)可完全切除的Ⅳ期恶性黑色素瘤的治疗注释[1-2]

1.转移灶切除应符合R0切除的原则。如有残余病灶,则应按不可切除的Ⅳ期对待。

2.缺乏有效的辅助治疗证据。参考文献

1.Kimbrough CW,McMasters KM,Davis EG.Principles of surgical treatment of malignant melanoma.Surg Clin North Am,2014,94(5): 973-988, vii.

2.Wei IH,Healy MA,Wong SL.Surgical treatment options for stageⅣ melanoma.Surg Clin North Am,2014, 94(5): 1075-1089, ix.(五)不可手术切除的Ⅳ期黑色素瘤的治疗皮肤播散性(不可手术切除)Ⅳ期黑色素瘤患者的治疗原则(1)存在脑转移患者的治疗a见黑色素瘤放疗原则

注释

1.脑转移灶的治疗:

对于存在脑转移的患者,应优先处理中枢神经系统(CNS)的病灶,以延迟或防止出现瘤内出血、癫痫或神经相关功能障碍。黑色素瘤脑转移的治疗应基于症状、脑转移灶的数目和部位来综合考虑。立[1-5][6-8]体定向放疗(SRS)和(或)全脑放疗(WBRT)均可作为一线治疗或术后辅助治疗应用于临床。与WBRT相比,SRS可能具有更好的长期安全性,能更早地使CNS病灶达到稳定,因此能使患者更早地接受全身系统性抗肿瘤治疗或参加临床研究。对于携带BRAF突变、同时存在颅外和颅内转移的患者,初始治疗有时采用BRAF或BRAF+MEK抑制剂,必要时联合放疗(作为巩固治疗)。在针对颅内病灶的治疗结束后,针对颅外病灶的处理同不伴有颅内转移的患者。ipilimumab可能能够长期地控制颅外转移灶。

若患者同时存在颅内和颅外病灶,可在对CNS病灶进行处理期间或之后予除大剂量IL-2(在既往未经治疗的脑转移中有效率低,并可能加重病灶周围的水肿)以外的全身系统性抗肿瘤治疗。由于联合或序贯应用放疗和系统性抗肿瘤治疗(尤其是BRAF靶向治疗)可能增加治疗相关毒性,因此务必谨慎考虑。[9-15]

2.晚期黑色素瘤的放疗原则:

对于脑转移灶而言,立体定向放疗可作为一线治疗或辅助治疗。全脑放疗可作为一线治疗,也可考虑作为辅助治疗(3类推荐),但作为辅助治疗时疗效不确切,需结合患者个体情况综合选择。

对于其他有症状或即将出现症状的软组织转移灶和(或)骨转移灶而言,可选择放疗,具体剂量和分次没有统一规定。

参考文献

1.Liew DN,Kano H,Kondziolka D,et al.Outcome predictors of Gamma knife surgery for melanoma brain metastases.Clinical article.J Neurosurg, 2011, 114: 769-779.

2.Frakes JM,Figura ND,Ahmed KA,et al.Potential role for LINAC-based stereotactic radiosurgery for the treatment of 5 or more radioresistant melanoma brain metastases.J Neurosurg, 2015, 123(5):1261-1267.

3.Selek U, Chang EL, Hassenbusch SJ, et al.Stereotactic radiosurgical treatment in 103 patients for 153 cerebral melanoma metastases.Int J Radiat Oncol Biol Phys, 2014, 59: 1097-1106.

4.Bernard ME, Wegner RE, Reineman K, et al.Linear accelerator based stereotactic radiosurgery for melanoma brain metastases.J Cancer Res Ther, 2012, 8: 215-221.

5.Rades D, Sehmisch L, Huttenlocher S, et al.Radiosurgery

试读结束[说明:试读内容隐藏了图片]

下载完整电子书


相关推荐

最新文章


© 2020 txtepub下载