弥漫性大B细胞淋巴瘤治疗进展
Advances in the Treatment of Diffuse Large B-Cell Lymphoma
DOI: 10.12677/ACM.2024.142542, PDF, HTML, XML, 下载: 75  浏览: 155 
作者: 伊再提·亚森江, 郭 莉*:新疆医科大学第三临床医学院,新疆 乌鲁木齐
关键词: 弥漫大B细胞淋巴瘤非霍奇金淋巴瘤预后治疗Diffuse Large B-Cell Lymphoma Non-Hodgkin’s Lymphoma Prognosis Treatment
摘要: 弥漫性大B细胞淋巴瘤(diffuse large B cell lymphoma, DLBCL)是非霍奇金淋巴瘤(non-Hodgkin’s lymphoma, NHL)最常见的亚型,包括一组形态学、遗传学和临床上不同的异质性疾病。据估计,全球每年有150,000例新发病例的大B细胞淋巴瘤,患者通常表现为进行性淋巴结肿大、结外病变或两者兼而有之,需要治疗。在过去的二十年里,人们对弥漫性大B细胞淋巴瘤的流行病学、组织学、免疫表型、遗传学特征、临床表现、化疗疗效及预后和生物异质性有了更深入的了解,从而完善了疾病分类并开发了新的治疗方法。在此,我们回顾了DLBCL治疗手段的最新进展,并展望与讨论了DLBCL患者未来的治疗前景。
Abstract: Diffuse large B cell lymphoma (DLBCL) is the most common subtype of non-Hodgkin’s lymphoma (NHL) and comprises a heterogeneous group of morphologically, genetically, and clinically distinct diseases. An estimated 150,000 new cases of large B-cell lymphoma occur globally each year, and patients usually present with progressive lymph node enlargement, extranodal lesions, or both, requiring treatment. Over the past two decades, a better understanding of the epidemiology, his-tology, immunophenotype, genetic features, clinical presentation, chemotherapy efficacy and prog-nosis, and biological heterogeneity of diffuse large B-cell lymphomas has led to refinement of dis-ease classifications and development of new therapeutic approaches. Here, we review the recent advances in the treatment of DLBCL and look forward to and discuss the future prospects for the treatment of patients with DLBCL.
文章引用:伊再提·亚森江, 郭莉. 弥漫性大B细胞淋巴瘤治疗进展[J]. 临床医学进展, 2024, 14(2): 3899-3904. https://doi.org/10.12677/ACM.2024.142542

1. 引言

弥漫性大B细胞淋巴瘤(diffuse large B-cell lymphoma, DLBCL)是淋巴瘤最常见的亚型,约占非霍奇金淋巴瘤(non-Hodgkin lymphoma, NHL)的40% [1] ,其包括一组不同的疾病实体,每个疾病实体都具有独特的分子和临床病理特征。DLBCL可能自发产生,也可能是低级别B细胞淋巴瘤转化的结果,最常见的形式是B细胞慢性淋巴细胞白血病(如Richter转化)。DLBCL是一种高级别淋巴瘤,可能发生在结节或结节外部位,包括胃肠道、睾丸、甲状腺、皮肤、乳腺、骨骼或大脑 [2] 。

目前,约三分之二的DLBCL患者在接受以利妥昔单抗为基础的化疗免疫疗法前线治疗后,可获得持久缓解并治愈 [3] 。然而,对其余患者来说,自体干细胞移植(ASCT) [4] 仅能为四分之一的病例提供治愈机会,而早期复发或难治性DLBCL的生存结果仍然令人沮丧 [5] 。值得注意的是,在治疗模式方面,最近有几种较新的方案获得批准,目前可用于一线治疗和复发治疗,结束了过去十年中负面临床试验匮乏的局面 [6] 。

2. 病理特征和分子分类

DLBCL的诊断依赖于对肿瘤组织的详细检查,最好由血液病理专家对切除活检标本进行评估 [7] 。除形态学特征外,淋巴瘤的准确分类还需要专门的检测,包括免疫组化、流式细胞术、荧光原位杂交(FISH)和分子检测。通过细针穿刺获得的活检标本不足以进行病理评估。虽然核心活检标本经常被使用,但往往不足以进行全面评估,只有在无法进行切除活检时才应进行核心活检。DLBCL可划分为两种不同的分子亚型,即生殖中心B细胞样(GCB)亚型和活化B细胞样(ABC)亚型;10%~15%的病例无法分类 [8] 。这些亚型被认为来自不同的淋巴分化阶段(原发细胞),依赖于不同的致癌机制,其中ABC亚型的预后较差(3年无进展生存率约为40%至50%,而GCB亚型为75%) [9] 。DLBCL的ABC亚型以慢性B细胞受体信号转导和核因子κB激活为特征,而GCB亚型则表达生殖中心B细胞中常见的基因,包括BCL6和EZH2。这种表型上的区别是有意义的,因为靶向药物可能在某一亚型中具有优先活性。另外,基于免疫组化的算法,如Hans算法,也可用于将病例分为GCB和非GCB (后者包括ABC亚型和大多数未分类病例),不过这些算法仅提供了基因表达谱分析的近似值,存在分类错误的风险 [10] 。

3. DLBCL遗传病变的机制

体细胞超突变(SHM)和转换重组(CSR)对于执行有效的免疫反应至关重要 [11] 。然而,由于它们能够引入DNA断裂,因此使GC B细胞的基因组面临持续的风险。此外,DNA损伤检查点因BCL6的转抑活性而沉默。因此,许多与DLBCL发病有关的结构改变(即染色体易位和异常体细胞高突变(ASHM))都源于这两种反应之一发生的错误 [12] 。在淋巴瘤易感小鼠模型中消融AID (SHM和CSR都需要的酶)能够防止MYC-IgH重排的形成和DLBCL的发生,从而正式证明了这一模型 [13] 。ASHM产生的病变通常分布在启动子近端序列中,根据靶基因的基因组结构,可能会影响非翻译区和编码区。因此,ASHM能够改变基因的转录调控或改变关键的结构/功能特性 [14] 。虽然目前还没有对ASHM可能造成的广泛遗传损伤进行全面鉴定,但这一机制很可能通过改变不同病例的多种细胞通路而导致DLBCL的异质性。

4. 预后因素

国际预后指数(IPI)仍是预测预后和在临床试验中对患者进行分层的主要临床工具 [15] 。IPI在现代得到了验证和完善,美国国家综合癌症网络IPI (NCCN-IPI)可对高危患者进行更大程度的区分 [16] 。然而,这些临床指标并不能用于识别极高风险患者或鉴别生物异质性。许多生物因素都与预后相关,这些因素尚未整合到一个有效的预后指数中。随着科学技术的不断进展,综合患者个体化生物学特征后,将会提出更多更适用于临床的评分标准。

5. 免疫疗法

5.1. 嵌合抗原受体T细胞和自然杀伤细胞(NK)

嵌合抗原受体(CAR)T细胞正迅速成为治疗复发/难治性(r/r) DLBCL的一种前景广阔的细胞免疫疗法。在CD19引导的CAR T细胞疗法中,axicabtagene ciloleucel (axi-cel,市场名为Yescarta) [17] 、lisocabtagene maraleucel (liso-cel) [18] 和tisagenlecleucel (市场名为Kymriah) [19] 的强大疗效已得到证实。在ZUMA-1研究中,101名既往接受过中位三线治疗的难治性侵袭性B细胞NHL患者接受了至少1.0 × 106个CAR阳性T细胞/公斤的治疗,研究者评估的ORR为83%,CR率为54% [17] 。ZUMA-1的2年随访数据表明,axi-cel可使r/r DLBCL患者获得持久应答并显著改善OS,且长期安全性可控 [20] 。另有93名不符合条件或ASCT后疾病进展的患者接受了tisagenlecleucel治疗,最佳ORR为52%,CR率为40%。初次应答后1年,估计无复发生存率为65%,表明tisagenlecleucel有持久的应答 [19] 。与历史数据相比,这些CAR T细胞产品在重度预处理的DLBCL患者中产生了意想不到的持久反应,这促进了Yescarta和Kymriah获得FDA批准。随着获得这些CAR T细胞产品的机会增加,r/r DLBCL患者可在二线方案中接受CAR T细胞治疗,甚至作为一种新的治疗手段。

5.2. 双CAR T细胞或与免疫检查点抑制剂联用

然而,尽管临床反应显著,但反应的持久性不高、治疗相关毒性以及耗时的生产是限制自体CAR T细胞疗法临床应用的主要障碍。CD19 CAR T细胞疗法后复发的部分原因是CD19丢失或程序性死亡配体1 (PD-L1)上调 [21] 。在这方面,可考虑使用能分泌人类抗PD-L1抗体的CAR T细胞、双CAR T细胞以及免疫检查点抑制剂。在治疗B细胞淋巴瘤方面,靶向CD19和CD20或CD22的双CAR T细胞很有吸引力。在一项1期试验中,一种靶向CD19和CD22的双特异性CAR T产品在5例r/r DLBCL患者(1例CR,2例PR)中取得了60%的ORR,且毒性可耐受 [22] 。此外,联合使用抗CD19和抗CD20 CAR T细胞治疗21例r/r DLBCL患者,总反应率(ORR)达81.0%,CR率达52.4% [20] 。有报道称,武装的CAR T细胞能分泌抗PD-L1抗体,可抵御T细胞衰竭,提高对小鼠肾细胞癌的疗效 [23] 。用pembrolizumab阻断程序性细胞死亡蛋白1 (PD1)对一些CD19 CAR T细胞治疗后进展的DLBCL患者安全有效。

5.3. 免疫调节药物

来那度胺作为一种免疫调节剂,被证实对免疫系统有多种影响,还能通过影响参与维持肿瘤生长和存活的细胞因子的产生和活性来改变肿瘤微环境。同时,来那度胺可通过与脑龙结合抑制下游NF-κB信号传导,直接产生肿瘤毒性 [24] 。来那度胺与R-CHOP21 (R2-CHOP)联合用药似乎在多项2期研究中获益,尤其是对非GCB和高风险亚组 [25] 。在REMARC研究中,对于对一线R-CHOP有反应的老年患者,来那度胺维持治疗24个月比安慰剂延长了PFS,但未发现OS获益 [26] 。然而,针对未经治疗的ABC-DLBCL的3期ROBUST研究并未达到PFS这一主要终点,不过在国际预后指数(IPI)评分较高和疾病分期较晚的患者中观察到了有利于R2-CHOP21的PFS阳性趋势 [27] 。同样,来那度胺和R-miniCHOP的3期研究数据显示,80岁以上患者的预后没有改善 [28] 。与此同时,ECOG-ACRINI412研究达到了主要终点,显示使用R-CHOP21联合来那度胺的PFS明显更好 [29] 。造成试验结果不同的可能原因包括不同的剂量(ROBUST中来那度胺15 mg d1-14和ECOG-ACRIN 1412中25 mg d1-10)、资格标准(ROBUST中仅ABC亚型,ECOG-ACRIN 1412中既有ABC也有GCB)和治疗时间(31天内)、这表明来那度胺的使用不应仅限于ABC-DLBCL,及时治疗可能会使这种侵袭性淋巴瘤患者进一步获益。尽管如此,来那度胺作为单一疗法 [30] 或联合挽救性化疗(如R-ICE [31] 和R-ESHAP [32] )已被证明对r/r DLBCL有效。

6. 复发或难治性DLBCL

约有30%~40%的DLBCL患者在一线化疗免疫治疗后仍难治或复发 [33] 。在二线治疗中,尽管约三分之二的患者会继续复发,但大剂量化疗后进行ASCT仍是治愈性治疗方案之一 [34] 。尤其是一线治疗后1年内难治或复发的患者,其治疗效果令人沮丧。三种CD19导向CAR-T疗法——axicabtagene ciloleucel [17] [20] 、tisagenlecleucel [19] [35] 和lisocabtagene maraleucel [36] ——已被批准用于复发或难治性大B细胞淋巴瘤(包括DLBCL)且接受过2线或2线以上系统治疗的成人患者。最近,两项随机III期试验也显示,对于一线治疗后1年内复发或难治的患者,CAR-T细胞产品优于ASCT [37] 。特别是关键的ZUMA-7试验报告,与标准化疗免疫疗法相比,axicabtagene ciloleucel的总生存率更高,随后是大剂量化疗和自体干细胞移植。axicabtagene ciloleucel的四年总生存率为54.6%,标准疗法为46% [5] 。美国FDA刚刚批准 glofitamab和epcoritamab用于治疗经过两线或更多线系统治疗后复发/难治的DLBCL患者 [38] 。一个有待回答的挑战性问题将是使用这些新型治疗方法的最佳顺序以及最佳组合方法。

7. 结论

随着时间的推移,DLBCL的诊断算法也在不断演变,从组织结构和细胞形态学的解剖病理学评估,到免疫表型的纳入,以及基因重排的评估。在分子医学和精准肿瘤学的新时代,DLBC的诊断已经并将继续根据临床相关性和应用进行改进。在DLBCL的治疗中加入免疫疗法已确定了新的治疗标准,在患者免疫健康状况较好的早期整合这些药物,并使用改进的策略来发现次优治疗反应,可能会对前期治疗方法产生重大影响。这些新发现可能暗示了DLBCL的薄弱环节,可用于治疗复发/难治性病例和未来的结节外播散。目前研究的重点是收集大样本的病例,利用不断发展的各种技术,如组织芯片、基因芯片、免疫组化等,以便更全面地认识DLBCL,这将有利于DLBCL从经验治疗向靶向治疗、为未来的DLBCL治疗策略提供新的见解。

NOTES

*通讯作者。

参考文献

[1] Susanibar-Adaniya, S. and Barta, S.K. (2021) 2021 Update on Diffuse Large B Cell Lymphoma: A Review of Current Data and Potential Applications on Risk Stratification and Management. American Journal of Hematology, 96, 617-629.
https://doi.org/10.1002/ajh.26151
[2] Lenz, G., et al. (2008) Stromal Gene Signatures in Large-B-Cell Lym-phomas. The New England Journal of Medicine, 359, 2313-2323.
https://doi.org/10.1056/NEJMoa0802885
[3] Poletto, S., et al. (2022) Treatment Strategies for Patients with Diffuse Large B-Cell Lymphoma. Cancer Treatment Reviews, 110, Article ID: 102443.
https://doi.org/10.1016/j.ctrv.2022.102443
[4] Gisselbrecht, C., et al. (2010) Salvage Regimens with Autologous Transplantation for Relapsed Large B-Cell Lymphoma in the Rituximab Era. Journal of Clinical Oncology, 28, 4184-4190.
https://doi.org/10.1200/JCO.2010.28.1618
[5] Westin, J.R., et al. (2023) Survival with Axicabtagene Ciloleucel in Large B-Cell Lymphoma. The New England Journal of Medicine, 389, 148-157.
https://doi.org/10.1056/NEJMoa2301665
[6] Nastoupil, L.J. and Bartlett, N.L. (2023) Navigating the Evolving Treatment Landscape of Diffuse Large B-Cell Lymphoma. Journal of Clinical Oncology, 41, 903-913.
https://doi.org/10.1200/JCO.22.01848
[7] Cheson, B.D., et al. (2014) Recommendations for Initial Evaluation, Staging, and Response Assessment of Hodgkin and Non-Hodgkin Lymphoma: The Lugano Classification. Journal of Clinical Oncology, 32, 3059-3068.
https://doi.org/10.1200/JCO.2013.54.8800
[8] Rosenwald, A., et al. (2002) The Use of Molecular Profiling to Predict Survival after Chemotherapy for Diffuse Large-B-Cell Lymphoma. The New England Journal of Medicine, 346, 1937-1947.
https://doi.org/10.1056/NEJMoa012914
[9] Scott, D.W., et al. (2015) Prognostic Significance of Diffuse Large B-Cell Lymphoma Cell of Origin Determined by Digital Gene Expression in Formalin-Fixed Paraffin-Embedded Tissue Biopsies. Journal of Clinical Oncology, 33, 2848-2856.
https://doi.org/10.1200/JCO.2014.60.2383
[10] Meyer, P.N., et al. (2011) Immunohistochemical Methods for Predicting Cell of Origin and Survival in Patients with Diffuse Large B-Cell Lymphoma Treated with Rituximab. Journal of Clinical Oncology, 29, 200-207.
https://doi.org/10.1200/JCO.2010.30.0368
[11] Muramatsu, M., et al. (2000) Class Switch Recombination and Hypermutation Require Activation-Induced Cytidine Deaminase (AID), A Potential RNA Editing Enzyme. Cell, 102, 553-563.
https://doi.org/10.1016/S0092-8674(00)00078-7
[12] Kuppers, R. and Dalla-Favera, R. (2001) Mechanisms of Chromosomal Translocations in B Cell Lymphomas. Oncogene, 20, 5580-5594.
https://doi.org/10.1038/sj.onc.1204640
[13] Ramiro, A.R., et al. (2004) AID Is Required for C-Myc/IgH Chro-mosome Translocations in Vivo. Cell, 118, 431-438.
https://doi.org/10.1016/j.cell.2004.08.006
[14] Pasqualucci, L., et al. (2001) Hypermutation of Multiple Pro-to-Oncogenes in B-Cell Diffuse Large-Cell Lymphomas. Nature, 412, 341-346.
https://doi.org/10.1038/35085588
[15] International Non-Hodgkin’s Lymphoma Prognostic Factors Project (1993) A Predictive Model for Aggressive Non- Hodgkin’s Lymphoma. The New England Journal of Medicine, 329, 987-994.
https://doi.org/10.1056/NEJM199309303291402
[16] Ruppert, A.S., et al. (2020) International Prognostic Indices in Diffuse Large B-Cell Lymphoma: A Comparison of IPI, R-IPI, and NCCN-IPI. Blood, 135, 2041-2048.
https://doi.org/10.1182/blood.2019002729
[17] Neelapu, S.S., et al. (2017) Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. The New England Journal of Medicine, 377, 2531-2544.
https://doi.org/10.1056/NEJMoa1707447
[18] Schuster, S.J., et al. (2017) Chimeric Antigen Receptor T Cells in Refractory B-Cell Lymphomas. The New England Journal of Medicine, 377, 2545-2554.
https://doi.org/10.1056/NEJMoa1708566
[19] Schuster, S.J., et al. (2019) Tisagenlecleucel in Adult Relapsed or Refractory Diffuse Large B-Cell Lymphoma. The New England Journal of Medicine, 380, 45-56.
https://doi.org/10.1056/NEJMoa1804980
[20] Locke, F.L., et al. (2019) Long-Term Safety and Activity of Axicabtagene Ciloleucel in Refractory Large B-Cell Lymphoma (ZUMA-1): A Single-Arm, Multicentre, Phase 1-2 Trial. The Lancet Oncology, 20, 31-42.
https://doi.org/10.1016/S1470-2045(18)30864-7
[21] Bukhari, A., et al. (2019) Rapid Relapse of Large B-Cell Lymphoma after CD19 Directed CAR-T-Cell Therapy due to CD-19 Antigen Loss. American Journal of Hematology, 94, E273-E275.
https://doi.org/10.1002/ajh.25591
[22] Spiegel, J.Y., et al. (2021) CAR T Cells with Dual Targeting of CD19 and CD22 in Adult Patients with Recurrent or Refractory B Cell Malignancies: A Phase 1 Trial. Nature Medicine, 27, 1419-1431.
https://doi.org/10.1038/s41591-021-01436-0
[23] Suarez, E.R., et al. (2016) Chimeric Antigen Receptor T Cells Secreting Anti-PD-L1 Antibodies More Effectively Regress Renal Cell Carcinoma in a Humanized Mouse Model. Oncotarget, 7, 34341-34355.
https://doi.org/10.18632/oncotarget.9114
[24] Garciaz, S., et al. (2016) Lenalidomide for the Treatment of B-Cell Lymphoma. Expert Opinion on Investigational Drugs, 25, 1103-1116.
https://doi.org/10.1080/13543784.2016.1208170
[25] Castellino, A., et al. (2018) Lenalidomide Plus R-CHOP21 in Newly Diagnosed Diffuse Large B-Cell Lymphoma (DLBCL): Long-Term Follow-Up Results from a Combined Analysis from Two Phase 2 Trials. Blood Cancer Journal, 8, Article No. 108.
https://doi.org/10.1038/s41408-018-0145-9
[26] Thieblemont, C., et al. (2017) Lenalidomide Maintenance Com-pared with Placebo in Responding Elderly Patients with Diffuse Large B-Cell Lymphoma Treated with First-Line Rituximab Plus Cyclophosphamide, Doxorubicin, Vincristine, and Prednisone. Journal of Clinical Oncology, 35, 2473-2481.
https://doi.org/10.1200/JCO.2017.72.6984
[27] Nowakowski, G.S., et al. (2021) ROBUST: A Phase III Study of Lenalidomide Plus R-CHOP Versus Placebo Plus R-CHOP in Previously Untreated Patients with ABC-Type Diffuse Large B-Cell Lymphoma. Journal of Clinical Oncology, 39, 1317-1328.
https://doi.org/10.1200/JCO.20.01366
[28] Oberic, L., et al. (2021) Subcutaneous Rituximab-MiniCHOP Com-pared with Subcutaneous Rituximab-MiniCHOP Plus Lenalidomide in Diffuse Large B-Cell Lymphoma for Patients Age 80 Years or Older. Journal of Clinical Oncology, 39, 1203-1213.
https://doi.org/10.1200/JCO.20.02666
[29] Nowakowski, G.S., et al. (2021) Addition of Lenalidomide to R-CHOP Improves Outcomes in Newly Diagnosed Diffuse Large B-Cell Lymphoma in A Randomized Phase II US In-tergroup Study ECOG-ACRIN E1412. Journal of Clinical Oncology, 39, 1329-1338.
https://doi.org/10.1200/JCO.20.01375
[30] Hernandez-Ilizaliturri, F.J., et al. (2011) Higher Response to Lenalidomide in Relapsed/Refractory Diffuse Large B-Cell Lymphoma in Nongerminal Center B-Cell-Like than in Germinal Center B-Cell-Like Phenotype. Cancer, 117, 5058-5066.
https://doi.org/10.1002/cncr.26135
[31] Feldman, T., et al. (2014) Addition of Lenalidomide to Rituximab, Ifosfamide, Carboplatin, Etoposide (RICER) in First-Relapse/Primary Refractory Diffuse Large B-Cell Lymphoma. British Journal of Haematology, 166, 77-83.
https://doi.org/10.1111/bjh.12846
[32] Martin, A., et al. (2016) Lenalidomide in Combination with R-ESHAP in Patients with Relapsed or Refractory Diffuse Large B-Cell Lymphoma: A Phase 1b Study from GELTAMO Group. British Journal of Haematology, 173, 245-252.
https://doi.org/10.1111/bjh.13945
[33] Nuvvula, S., Dahiya, S. and Patel, S.A. (2022) The Novel Therapeutic Landscape for Relapsed/Refractory Diffuse Large B Cell Lymphoma. Clinical Lymphoma, Myeloma and Leukemia, 22, 362-372.
https://doi.org/10.1016/j.clml.2021.11.010
[34] Crump, M., et al. (2014) Randomized Comparison of Gemcitabine, Dexamethasone, and Cisplatin versus Dexamethasone, Cytarabine, and Cisplatin Chemotherapy before Autologous Stem-Cell Transplantation for Relapsed and Refractory Aggressive Lymphomas: NCIC-CTG LY.12. Journal of Clinical Oncology, 32, 3490-3496.
https://doi.org/10.1200/JCO.2013.53.9593
[35] Chong, E.A., et al. (2021) Five-Year Outcomes for Refractory B-Cell Lymphomas with CAR T-Cell Therapy. The New England Journal of Medicine, 384, 673-674.
https://doi.org/10.1056/NEJMc2030164
[36] Schuster, S.J., et al. (2021) Long-Term Clinical Outcomes of Tisagenlecleucel in Patients with Relapsed or Refractory Aggressive B-Cell Lymphomas (JULIET): A Multicentre, Open-Label, Single-Arm, Phase 2 Study. The Lancet Oncology, 22, 1403-1415.
https://doi.org/10.1016/S1470-2045(21)00375-2
[37] Locke, F.L., et al. (2022) Axicabtagene Ciloleucel as Se-cond-Line Therapy for Large B-Cell Lymphoma. The New England Journal of Medicine, 386, 640-654.
https://doi.org/10.1056/NEJMoa2116133
[38] Reddy, A., et al. (2017) Genetic and Functional Drivers of Diffuse Large B Cell Lymphoma. Cell, 171, 481-494.e15.