髓外多发性骨髓瘤的治疗进展
Advances in the Treatment of Extramedullary Multiple Myeloma
DOI: 10.12677/jcpm.2024.32078, PDF, HTML, XML, 下载: 20  浏览: 34 
作者: 任鹏慧*:青海大学临床医学院,青海 西宁;马晓静:青海大学附属医院血液科,青海 西宁
关键词: 髓外多发性骨髓瘤蛋白酶体抑制剂免疫调节剂治疗自体干细胞移植Extramedullary Multiple Myeloma Proteasome Inhibitor Immunomodulator Therapy Autologous Stem Cell Transplantation
摘要: 多发性骨髓瘤(Multiple myeloma, MM)是一种浆细胞异常克隆性增殖导致的恶性肿瘤,髓外多发性骨髓瘤(Extramedullary multiple myeloma, EMM)为MM细胞恶性增殖后产生侵蚀骨表面或骨外部组织的实体瘤。现有的治疗手段虽使MM的疗效得到巨大改善,但EMM的预后仍较差。本文就EMM的治疗现状主要从蛋白酶体抑制剂(Proteasome inhibitors, PI)联合免疫调节剂治疗(Immunomodulatory drugs, IMiD)、嵌合抗原受体T细胞(Chimeric antigen receptor T-cell, CAR-T)免疫治疗、自体干细胞移植(Autologous stem cell transplantation, ASCT)及单克隆抗体等方面进行综述。
Abstract: Multiple myeloma (MM) is a malignant tumor caused by the abnormal clonal proliferation of plasma cells, and extramedullary multiple myeloma (EMM) is a solid tumor arising from the malignant proliferation of MM cells that erodes the bone surface or tissues outside the bone. Although existing treatments have greatly improved the efficacy of MM, the prognosis of EMM is still poor. In this paper, we focus on the current status of EMM treatment from the perspective of proteasome inhibitors (PI) combined with immunomodulatory drugs (IMiD), chimeric antigen receptor T-cells (CAR-T) immunotherapy, autologous stem cell transplantation (ASCT) and monoclonal antibodies are reviewed.
文章引用:任鹏慧, 马晓静. 髓外多发性骨髓瘤的治疗进展[J]. 临床个性化医学, 2024, 3(2): 530-537. https://doi.org/10.12677/jcpm.2024.32078

1. 引言

多发性骨髓瘤(MM)是一种血液系统肿瘤,其特征是骨髓中恶性浆细胞的克隆增殖。当MM细胞逃离骨髓微环境时,会导致髓外多发性骨髓瘤(Extramedullary multiple myeloma, EMM) [1]。EMM的可能机制包括黏附分子表达减少、趋化因子受体表达改变[2] [3]、四次跨膜蛋白(Traspanins)表达下调[4]。EMM按发生部位分为骨相关髓外骨髓瘤(Extramedullary bone-related multiple myeloma,EMB。MM细胞破坏骨皮质并直接生长到邻近组织)、软组织相关髓外骨髓瘤(Extramedullary soft tissue-related multiple myeloma,EMS。MM细胞通过血行播散侵入软组织形成肿块,与骨骼无关)。后者往往预后较差[5] [6]。目前,EMM的治疗方案主要是以蛋白酶体抑制剂(Proteasome inhibitors, PI)及免疫调节剂(Immunomodulatory drugs, IMiD)为基础方案,自体干细胞移植(Autologous stem cell transplantation, ASCT)在EMM治疗中仍占重要地位。同时,新型药物,如单克隆抗体、XPO-1抑制剂以及嵌合抗原受体T细胞(Chimeric antigen receptor T-cell, CAR-T)免疫治疗等在临床中也有应用。

2. 蛋白酶体抑制剂(PI)

2.1. 硼替佐米

根据Mayo Clinic在2017年[7]的推荐,硼替佐米具有广泛的组织渗透性[8],目前常用于髓外疾病(Extramedullary disease, EMD)治疗。Laura等[9]报道,4例复发或难治性多发性骨髓瘤(RRMM)患者中有3例在硼替佐米治疗后EMD消失。Landau等[10]的研究显示,14例EMM患者依次接受硼替佐米、脂质体多柔比星和地塞米松(BDD)联合/不联合硼替佐米(BTD或TD)治疗,总生存率为86% (12/14)。Lakshman等[11]研究了141例接受VDT-PACE (地塞米松/沙利度胺/顺铂/多柔比星/环磷酰胺/依托泊苷/硼替佐米)治疗的RRMM患者。在VDT-PACE治疗开始时,EMM患者与非EMM患者的总体缓解率(ORR)没有差异(57.1% vs. 52.9%; P = 0.631)。这表明含硼替佐米的方案可能减轻RRMM中EMD的不良结局。

2.2. 伊沙佐米

基于ixazomib的方案在EMM患者中的研究有限。根据3期TOURMALINE-MM1研究的结果,伊沙佐米联合来那度胺和地塞米松(IRD)可显著改善RRMM患者的无进展生存期(PFS) [12]。然而,Minarik等人发现EMM患者并未从IRD治疗中获益。IRD组患者的无进展生存期(PFS)与RD (来那度胺、地塞米松)组相似(6.5个月vs 10.9个月,HR 1.24 [0.54~2.86]) [13]。ixazomib在EMD中的疗效需要在未来的研究中确认。

2.3. 卡非佐米

在复发性MM患者中,在来那度胺和地塞米松的基础上加用卡非佐米可显著改善PFS [14]。在周等人的研究中,45例RRMM EMM患者接受了基于卡非佐米的治疗。45例患者中有33例具有可用于确定EMM最佳反应的影像学随访数据。他们观察到血清学和髓外ORR分别为59%和27%,其中9例患者部分缓解(PR)、9例疾病稳定(SD)、9例进展性疾病(PD)和6例混合缓解EMD病变。中位PFS和OS分别为5个月(95% CI, 3.5~6.5)和10个月(95% CI, 7.5~12.5) [15]。Muchtar等人报道,与非EMD患者相比,接受含卡非佐米方案的EMD患者的反应持续时间(DOR)更短(3.9个月vs. 9.3个月,P = 0.06)。两种患者的ORR差异无统计学意义(40% vs. 49%, P = 0.39),但EMM患者的临床获益缓解率(CBR)显著低于无EMM患者,表明存在EMM的患者中真正无反应者的发生率更高[16]。因此,卡非佐米对EMM的作用有限。

3. 免疫调节药物(IMiDs)

3.1. 沙利度胺

沙利度胺是治疗MM的第一代免疫调节药物,Laura报道,11例EMD患者中均未对沙利度胺单药治疗有反应[17]。同时,在Bladé等[18]的研究中,5例EMD患者均未对沙利度胺有反应。Avigdor等[19]和Anagnostopoulos等[20]报道,尽管骨髓中反应良好,但接受沙利度胺治疗的MM患者仍发生EMD。也有报道称,沙利度胺对EMD缺乏疗效[21]因此,沙利度胺已被证明对治疗EMD无效。

3.2. 来那度胺

Calvo-Villas等[22]分析了18例来那度胺联合地塞米松治疗的EMD患者。在中位治疗期为7年后,61.1% (11例患者)在EMM中表现出反应,包括44.4% (8例患者)完全消失,16.6% (3例患者)的体积缩小。该研究的缓解率与先前报道的硼替佐米[23] [24] [25] [26]和来那度胺[27]相似。中位OS和PFS分别为14.6个月和9.8个月。在他们的研究系列研究中,他们还证实来那度胺治疗后EMD的缓解率高于沙利度胺[17] [18]

3.3. 泊马度胺

泊马度胺被批准用于治疗RRMM,特别是来那度胺难治性患者[28]。Short等[29]报道了13例EMD患者,所有患者均接受了新型药物治疗,其中100%的患者暴露于沙利度胺或来那度胺,78%的患者暴露于硼替佐米。泊马度胺加低剂量地塞米松治疗后,2例患者达到CR (EMD完全消失),2例患者出现PR (≥EMD降低50%),2例SD。与无EMM的患者相比,所有EMM患者的OS均显著缩短。在Jiménez-Segura等[30]的研究中,21例骨相关的患者中只有2例对泊马度胺–地塞米松治疗有髓外反应。从泊马度胺–地塞米松治疗开始,中位PFS为1.7个月,中位OS为4.5个月。这项研究表明,泊马度胺–地塞米松治疗对EMD患者的疗效有限,尤其是软组织相关。根据我们的经验[31],我们对6例EMM患者进行了一项观察性研究,这些患者既往采用的多线治疗为4种(范围2~5)。髓外复发后,接受基于泊马度胺的治疗(硼替佐米/地塞米松/泊马度胺、泊马度胺/阿霉素/地塞米松、达雷妥尤单抗/泊马度胺/地塞米松、地塞米松/泊马度胺/顺铂/阿霉素/环磷酰胺/依托泊苷、泊马度胺/伊沙佐米/地塞米松),83%的患者出现髓外反应,CR为50%,PR为33%,1例患者出现髓外进展。血清学反应方面,1例患者为CR,4例为PR,1例为PD。诊断为EMD后的中位PFS为5个月,中位OS为8个月。因此,泊马度胺对EMD的疗效需要进一步研究,可以考虑与其他新型药物联合使用。

4. 自体干细胞移植(ASCT)

关于ASCT是否能克服不良预后,目前尚无统一结论。软组织相关组的ASCT比骨相关组更有效[32]。Lee等[33]报道了275例MM患者,其中54例在诊断时患有EMM。在154例符合移植条件的患者中,27例EMM患者和127例非EMM患者的OS和PFS没有差异(两年OS:82.9% vs 84.7%,P = 0.487;两年PFS:54.2% vs 58.7%,P = 0.339)。同时,在121例不符合移植条件的患者中,27例存在EMM的患者和94例不存在EMM的患者的结局存在差异(两年OS:44.2% vs 80.8%,P = 0.007;两年PFS:39.6% vs 48.2%,P = 0.054)。这项研究表明,ASCT可以克服EMM引起的不良预后影响,而EMM是不适合移植的患者的不良预后因素。然而,在Kumar等人的研究中,44例患者中有16例在ASCT后达到CR,而没有EMM的患者中有142例达到CR (36.4%对89.9%,P < 0.002)。ASCT后的CR率是OS和PFS最重要的预测因子,EMM患者的中位OS和中位PFS比无EMD患者短(中位OS:32个月vs 100个月,P < 0.001;中位PFS:18个月vs 44个月,P < 0.001)。EMD患者ASCT缓解率较低,PFS和OS较差,原因可能是大多数患者为ISS III期和Durie Salmon IIIB期[34]。移植方案的选择仍存在争议。EMBT的一项回顾性研究得出结论,串联ASCT和单次ASCT的PFS和OS相同[35]。Gagelmann等人发现,串联自体移植可显著提高高危细胞遗传学EMM患者的生存率[36]

5. 靶向CD38的单克隆抗体

5.1. 达雷妥尤单抗

达雷妥尤单抗在EMD中的疗效有限,这种现象可以通过髓外恶性克隆浆细胞上CD38表达降低来解释[37]。一项真实世界的回顾性研究显示,单药达雷妥尤单抗在41例RRMM患者中疗效适中,其中包括32%的EMM患者。ORR为24.4%,CBR为39%。中位OS和中位PFS分别为1.9个月和6.5个月[38]。在SIRIUS试验中,106例患者接受了16 mg/kg的达雷妥尤单抗治疗,其中14例为EMM,只有3例患者有反应[39]。Pick等人报道,在9例接受达雷妥尤单抗治疗的EMM患者中,只有2例达到PR,但均在50日和85日内发生PD [40]

5.2. 伊妥昔单抗

Isatuximab是一种单克隆抗体,可与人细胞CD38受体上的特异性表位结合[41]。ICARIA-MM试验证明了伊萨妥昔单抗联合泊马度胺和地塞米松治疗RRMM的益处。在该试验中,isatuximab-泊马度胺–地塞米松组的中位PFS为11.5个月(95% CI, 8.9~13.9),而泊马度胺–地塞米松组的中位PFS为6.5个月(95% CI, 4.5~8.3) [42]。在本次试验中,24名患者出现EMM,其中Isa-Pd组14名患者,Pd组10名患者。Isa-Pd组(4.57个月)的PFS延长Pd组(1.56个月)。ORR也有所改善:Isa-Pd组和Pd组分别为50% (7/14)和10% (1/10)。2例患者髓外病变在第3周期显示CR,在第4周期显著减少。因此,Isa-Pd在RRMM伴有EMD的患者中显示出良好的疗效。

5.3. 埃洛妥珠单抗

Elotuzumab是针对SLAMF7 (也称为CS1)单克隆抗体的首创人源化免疫球蛋白G1,通过CD16通路直接介导抗体依赖性细胞介导的细胞毒性(ADCC)并激活NK细胞,从而发挥杀死MM细胞的双重作用[43] [44]。ELOQUENT-2试验显示,依洛妥珠单抗联合来那度胺或泊马度胺联合低剂量地塞米松治疗RRMM患者,结果令人鼓舞[45] [46]。一项病例报告显示,1例RRMM患者在接受elotuzumab/来那度胺/地塞米松8个周期治疗后EMD消失[47]。Danhof等[48]报道了15例EMM患者(8例EM-B,3例EM-S,4例EM-B和EM-S)接受了基于elotuzumab的治疗。1名患者VGPR和5名PR的ORR为40%,相比之下,分别低于ELOQUENT-2和ELOQUENT-3临床试验的79%和53%。对于髓外病变,4例患者出现消退或SD,6例患者出现PD。他们还报道,在胸膜EMD患者中,与BMPC相比,髓外部位浆细胞的SLAMF7表达较低,这可能解释了接受elotuzumab治疗的EMD患者结局相对较差的原因。目前,elotuzumab在EMD中的疗效正在研究中,SLAMF7靶向的免疫疗法,如抗体–药物偶联物和CAR-T细胞,有望进一步发展。

6. 塞利尼索

塞利尼索是一种XPO1介导的核输出抑制剂,已于2019年7月在美国获批,用于治疗既往接受过至少4种治疗且至少2种PIs/IMiD和抗CD38单克隆抗体难治的RRMM患者。在STROM研究中,Yee等人分析了27例接受塞利尼索联合低剂量地塞米松(Sel-dex)治疗的EMD患者,其中16例可用于评估EMD。他们发现5名患者获得了客观反应,2名患者获得了最小缓解(MR),4名患者获得了SD,5名患者获得了客观PD。这些观察结果支持了Sel-dex在EMD患者中活跃的发现[49]

7. 抗BCMA嵌合抗原受体(CAR) T细胞疗法

抗BCMA CAR-T细胞疗法已显示出对RRMM的疗效[50]。Xu等[51]报道了17例MM患者接受了抗BCMA CAR-T细胞治疗,结果显示88%的患者有反应。经过11个月的随访,47.1%的患者仍处于持续缓解状态,这表明抗BCMA CAR-T细胞疗法在治疗MM方面显示出非常好的疗效。值得注意的是,本研究中的五名EMM患者中有四名预后不良,均在一年内复发,并且所有复发的髓外病变都出现在髓质进展的新部位。在邓等人的报道[52]中,20名RRMM患者被纳入研究,其中7名患者患有EMD。尽管EMD患者的细胞因子释放综合征(CRS)和免疫效应细胞相关神经毒性综合征(ICANS)的分级要高得多,但EMM患者和无EMM患者的ORR没有差异(71.43% vs 80%, P = 0.876)。两组在180天时的PFS率和OS率没有差异(PFS率:42.9% vs 84.6%,P = 0.068;OS率:71.5% vs 92.3%,P = 0.220)。然而,在360天时,EMM患者的PFS率和OS率均低于无EMM患者(PFS率:28.6% vs 72.5%,P = 0.037;OS率:28.6% vs 81.0%,P = 0.030)。结果表明,抗BCMA CAR-T细胞疗法可以为有反应的EMM患者提供短暂的缓解。通过桥接造血干细胞移植和放疗是否可以增强其疗效,需要进一步探索。

综上所述,EMM的治疗仍是临床难点。但是随着卡非佐米、塞利尼索及单克隆抗体等新药的上市以及CAR-T细胞治疗的出现,EMM患者的疗效得到明显改善,生存期明显延长。但是,伴EMM患者的生存期仍明显低于其他没有EMM的患者,预后仍不佳。因此,目前仍需要对EMM的发病机制进行深入研究,进一步研发具有新的作用机制的药物,探索更多的药物联合方案。

NOTES

*通讯作者。

参考文献

[1] Wang, X., Luo, K., Xu, Q., Chi, L., Guo, Y., Jia, C., et al. (2024) Prognostic Marker CD27 and Its Micro-Environmental in Multiple Myeloma. BMC Cancer, 24, Article No. 352.
https://doi.org/10.1186/s12885-024-11945-z
[2] Sakae, Y., Takada, H., Ichinose, S., Nakajima, M., Sakai, A. and Ogawa, R. (2023) Treatment with YIGSR Peptide Ameliorates Mouse Tail Lymphedema by 67 kDa Laminin Receptor (67LR)-Dependent Cell-Cell Adhesion. Biochemistry and Biophysics Reports, 35, Article 101514.
https://doi.org/10.1016/j.bbrep.2023.101514
[3] Yang, Y., Li, J., Lei, W., Wang, H., Ni, Y., Liu, Y., et al. (2023) CXCL12-CXCR4/CXCR7 Axis in Cancer: From Mechanisms to Clinical Applications. International Journal of Biological Sciences, 19, 3341-3359.
https://doi.org/10.7150/ijbs.82317
[4] Quagliano, A., Gopalakrishnapillai, A. and Barwe, S.P. (2023) Tetraspanins Set the Stage for Bone Marrow Microenvironment-Induced Chemoprotection in Hematologic Malignancies. Blood Advances, 7, 4403-4413.
https://doi.org/10.1182/bloodadvances.2023010476
[5] Wang, J., Shen, N., Shen, X., Zhang, R., Jin, Y., Li, J., et al. (2023) Survival Trends and Prognostic Factors of Patients with Newly Diagnosed Multiple Myeloma Accompanied with Extramedullary Disease. Annals of Medicine, 55, Article 2281657.
https://doi.org/10.1080/07853890.2023.2281657
[6] Sammartano, V., Cerase, A., Venanzi, V., Mazzei, M.A., Vangone, B.E., Gentili, F., et al. (2022) Central Nervous System Myeloma and Unusual Extramedullary Localizations: Real Life Practical Guidance. Frontiers in Oncology, 12, Article 934240.
https://doi.org/10.3389/fonc.2022.934240
[7] Dingli, D., Ailawadhi, S., Bergsagel, P.L., Buadi, F.K., Dispenzieri, A., Fonseca, R., et al. (2017) Therapy for Relapsed Multiple Myeloma. Mayo Clinic Proceedings, 92, 578-598.
https://doi.org/10.1016/j.mayocp.2017.01.003
[8] Gagelmann, N., Eikema, D., Koster, L., Netelenbos, T., McDonald, A., Stoppa, A., et al. (2023) Impact of Newly Diagnosed Extramedullary Myeloma on Outcome after First Autograft Followed by Maintenance: A CMWP-EBMT Study. European Journal of Haematology, 111, 181-190.
https://doi.org/10.1111/ejh.13981
[9] Li, Y., Sun, Z. and Qu, X. (2022) Advances in the Treatment of Extramedullary Disease in Multiple Myeloma. Translational Oncology, 22, Article 101465.
https://doi.org/10.1016/j.tranon.2022.101465
[10] Tabchi, S., Nair, R., Kunacheewa, C., Patel, K.K., Lee, H.C., Thomas, S.K., et al. (2019) Retrospective Review of the Use of High-Dose Cyclophosphamide, Bortezomib, Doxorubicin, and Dexamethasone for the Treatment of Multiple Myeloma and Plasma Cell Leukemia. Clinical Lymphoma Myeloma and Leukemia, 19, 560-569.
https://doi.org/10.1016/j.clml.2019.05.001
[11] Lakshman, A., Singh, P.P., Rajkumar, S.V., Dispenzieri, A., Lacy, M.Q., Gertz, M.A., et al. (2017) Efficacy of VDT Pace-Like Regimens in Treatment of Relapsed/Refractory Multiple Myeloma. American Journal of Hematology, 93, 179-186.
https://doi.org/10.1002/ajh.24954
[12] Moreau, P., Masszi, T., Grzasko, N., Bahlis, N.J., Hansson, M., Pour, L., et al. (2016) Oral Ixazomib, Lenalidomide, and Dexamethasone for Multiple Myeloma. New England Journal of Medicine, 374, 1621-1634.
https://doi.org/10.1056/nejmoa1516282
[13] Minarik, J., Pika, T., Radocha, J., Jungova, A., Straub, J., Jelinek, T., et al. (2021) Survival Benefit of Ixazomib, Lenalidomide and Dexamethasone (IRD) over Lenalidomide and Dexamethasone (Rd) in Relapsed and Refractory Multiple Myeloma Patients in Routine Clinical Practice. BMC Cancer, 21, Article No. 73.
https://doi.org/10.1186/s12885-020-07732-1
[14] Stewart, A.K., Rajkumar, S.V., Dimopoulos, M.A., Masszi, T., Špička, I., Oriol, A., et al. (2015) Carfilzomib, Lenalidomide, and Dexamethasone for Relapsed Multiple Myeloma. New England Journal of Medicine, 372, 142-152.
https://doi.org/10.1056/nejmoa1411321
[15] Zhou, X., Flüchter, P., Nickel, K., Meckel, K., Messerschmidt, J., Böckle, D., et al. (2020) Carfilzomib Based Treatment Strategies in the Management of Relapsed/Refractory Multiple Myeloma with Extramedullary Disease. Cancers, 12, Article 1035.
https://doi.org/10.3390/cancers12041035
[16] Muchtar, E., Gatt, M.E., Rouvio, O., Ganzel, C., Chubar, E., Suriu, C., et al. (2015) Efficacy and Safety of Salvage Therapy Using Carfilzomib for Relapsed or Refractory Multiple Myeloma Patients: A Multicentre Retrospective Observational Study. British Journal of Haematology, 172, 89-96.
https://doi.org/10.1111/bjh.13799
[17] Rosiñol, L., Cibeira, M.T., Bladé, J., et al. (2004) Extramedullary Multiple Myeloma Escapes the Effect of Thalidomide. Haematologica, 89, 832-836.
[18] Bladé, J., Perales, M., Rosiñol, L., Tuset, M., Montoto, S., Esteve, J., et al. (2001) Thalidomide in Multiple Myeloma: Lack of Response of Soft-Tissue Plasmacytomas. British Journal of Haematology, 113, 422-424.
https://doi.org/10.1046/j.1365-2141.2001.02765.x
[19] Avigdor, A., Raanani, P., Levi, I., Hardan, I. and Ben-Bassat, I. (2001) Extramedullary Progression Despite a Good Response in the Bone Marrow in Patients Treated with Thalidomide for Multiple Myeloma. Leukemia & Lymphoma, 42, 683-687.
https://doi.org/10.3109/10428190109099330
[20] Anagnostopoulos, A., Hamilos, G., Zorzou, M.P., Grigoraki, V., Anagnostou, D. and Athanasios Dimopoulos, M. (2004) Discordant Response or Progression in Patients with Myeloma Treated with Thalidomide-Based Regimens. Leukemia & Lymphoma, 45, 113-116.
https://doi.org/10.1080/1042819031000151860
[21] Myers, B., Grimley, C., Crouch, D. and Dolan, G. (2001) Lack of Response to Thalidomide in Plasmacytomas. British Journal of Haematology, 115, 234-234.
https://doi.org/10.1046/j.1365-2141.2001.0115_1cr-6.x
[22] Calvo-Villas, J.M., Alegre, A., Calle, C., Hernández, M.T., García-Sánchez, R. and Ramírez, G. (2011) Lenalidomide Is Effective for Extramedullary Disease in Relapsed or Refractory Multiple Myeloma. European Journal of Haematology, 87, 281-284.
https://doi.org/10.1111/j.1600-0609.2011.01644.x
[23] Patriarca, F., Prosdocimo, S., Tomadini, V., et al. (2005) Efficacy of Bortezomib Therapy for Extramedullary Relapse of Myeloma after Autologous and Non-Myeloablative Allogeneic Transplantation. Haematologica, 90, 278-279.
[24] Laura, R., Cibeira, M.T., Uriburu, C., Yantorno, S., Salamero, O., Bladé, J., et al. (2006) Bortezomib: An Effective Agent in Extramedullary Disease in Multiple Myeloma. European Journal of Haematology, 76, 405-408.
https://doi.org/10.1111/j.0902-4441.2005.t01-1-ejh2462.x
[25] Paubelle, E., Coppo, P., Garderet, L., Azizi, L., Bories, D., Gorin, N., et al. (2005) Complete Remission with Bortezomib on Plasmocytomas in an End-Stage Patient with Refractory Multiple Myeloma Who Failed All Other Therapies Including Hematopoietic Stem Cell Transplantation: Possible Enhancement of Graft-vs-Tumor Effect. Leukemia, 19, 1702-1704.
https://doi.org/10.1038/sj.leu.2403855
[26] Krauth, M., Bankier, A., Valent, P., Kalhs, P. and Drach, J. (2005) Sustained Remission Including Marked Regression of a Paravertebral Plasmacytoma in a Patient with Heavily Pretreated, Relapsed Multiple Myeloma after Treatment with Bortezomib. Leukemia Research, 29, 1473-1477.
https://doi.org/10.1016/j.leukres.2005.05.003
[27] Alegre, A., Aguado, B., Giraldo, P., Ríos, E., Cánovas, A., Ibáñez, Á., et al. (2011) Lenalidomide Is Effective as Salvage Therapy in Refractory or Relapsed Multiple Myeloma: Analysis of the Spanish Compassionate Use Registry in Advanced Patients. International Journal of Hematology, 93, 351-360.
https://doi.org/10.1007/s12185-011-0785-z
[28] Shah, N., Chari, A., Scott, E., Mezzi, K. and Usmani, S.Z. (2020) B-Cell Maturation Antigen (BCMA) in Multiple Myeloma: Rationale for Targeting and Current Therapeutic Approaches. Leukemia, 34, 985-1005.
https://doi.org/10.1038/s41375-020-0734-z
[29] Short, K.D., Rajkumar, S.V., Larson, D., Buadi, F., Hayman, S., Dispenzieri, A., et al. (2011) Incidence of Extramedullary Disease in Patients with Multiple Myeloma in the Era of Novel Therapy, and the Activity of Pomalidomide on Extramedullary Myeloma. Leukemia, 25, 906-908.
https://doi.org/10.1038/leu.2011.29
[30] Jiménez-Segura, R., Granell, M., Gironella, M., Abella, E., García-Guiñón, A., Oriol, A., et al. (2019) Pomalidomide-Dexamethasone for Treatment of Soft-Tissue Plasmacytomas in Patients with Relapsed/Refractory Multiple Myeloma. European Journal of Haematology, 102, 389-394.
https://doi.org/10.1111/ejh.13217
[31] Li, Y., Ji, J., Lu, H., Li, J. and Qu, X. (2022) Pomalidomide-Based Therapy for Extramedullary Multiple Myeloma. Hematology, 27, 88-94.
https://doi.org/10.1080/16078454.2021.2019364
[32] Li, J., Shen, K., Huang, W., Li, L., Chen, H., Chen, W., et al. (2014) Autologous Stem Cell Transplant Can Overcome Poor Prognosis in Patients with Multiple Myeloma with Extramedullary Plasmacytoma. Leukemia & Lymphoma, 55, 1687-1690.
https://doi.org/10.3109/10428194.2013.853296
[33] Lee, S., Kim, J., Jeon, Y., Yoon, J., Shin, S., Eom, K., et al. (2014) Impact of Extramedullary Plasmacytomas on Outcomes According to Treatment Approach in Newly Diagnosed Symptomatic Multiple Myeloma. Annals of Hematology, 94, 445-452.
https://doi.org/10.1007/s00277-014-2216-8
[34] Kumar, L., Gogi, R., Patel, A.K., Mookerjee, A., Sahoo, R.K., Malik, P.S., et al. (2017) Multiple Myeloma with Extramedullary Disease: Impact of Autologous Stem Cell Transplantation on Outcome. Bone Marrow Transplantation, 52, 1473-1475.
https://doi.org/10.1038/bmt.2017.165
[35] Gagelmann, N., Eikema, D.J., Iacobelli, S., et al. (2018) Impact of Extramedullary Disease in Patients with Newly Diag-Nosed Multiple Myeloma Undergoing Autologous Stem Cell Transplantation: A Study from the Chronic Malignancies Working Party of the EBMT. Haematologica, 103, 890-897.
[36] Gagelmann, N., Eikema, D., Koster, L., Caillot, D., Pioltelli, P., Lleonart, J.B., et al. (2019) Tandem Autologous Stem Cell Transplantation Improves Outcomes in Newly Diagnosed Multiple Myeloma with Extramedullary Disease and High-Risk Cytogenetics: A Study from the Chronic Malignancies Working Party of the European Society for Blood and Marrow Transplantation. Biology of Blood and Marrow Transplantation, 25, 2134-2142.
https://doi.org/10.1016/j.bbmt.2019.07.004
[37] Jelinek, T., Sevcikova, T., Zihala, D., Popkova, T., Kapustova, V., Broskevicova, L., et al. (2021) Limited Efficacy of Daratumumab in Multiple Myeloma with Extramedullary Disease. Leukemia, 36, 288-291.
https://doi.org/10.1038/s41375-021-01343-w
[38] Jullien, M., Trudel, S., Tessoulin, B., Mahé, B., Dubruille, V., Blin, N., et al. (2019) Single-Agent Daratumumab in Very Advanced Relapsed and Refractory Multiple Myeloma Patients: A Real-Life Single-Center Retrospective Study. Annals of Hematology, 98, 1435-1440.
https://doi.org/10.1007/s00277-019-03655-5
[39] Lonial, S., Weiss, B.M., Usmani, S.Z., Singhal, S., Chari, A., Bahlis, N.J., et al. (2016) Daratumumab Monotherapy in Patients with Treatment-Refractory Multiple Myeloma (SIRIUS): An Open-Label, Randomised, Phase 2 Trial. The Lancet, 387, 1551-1560.
https://doi.org/10.1016/s0140-6736(15)01120-4
[40] Pick, M., Vainstein, V., Goldschmidt, N., Lavie, D., Libster, D., Gural, A., et al. (2018) Daratumumab Resistance Is Frequent in Advanced-Stage Multiple Myeloma Patients Irrespective of CD38 Expression and Is Related to Dismal Prognosis. European Journal of Haematology, 100, 494-501.
https://doi.org/10.1111/ejh.13046
[41] Dimopoulos, M., Bringhen, S., Anttila, P., Capra, M., Cavo, M., Cole, C., et al. (2021) Isatuximab as Monotherapy and Combined with Dexamethasone in Patients with Relapsed/Refractory Multiple Myeloma. Blood, 137, 1154-1165.
https://doi.org/10.1182/blood.2020008209
[42] Attal, M., Richardson, P.G., Rajkumar, S.V., San-Miguel, J., Beksac, M., Spicka, I., et al. (2019) Isatuximab Plus Pomalidomide and Low-Dose Dexamethasone versus Pomalidomide and Low-Dose Dexamethasone in Patients with Relapsed and Refractory Multiple Myeloma (ICARIA-MM): A Randomised, Multicentre, Open-Label, Phase 3 Study. The Lancet, 394, 2096-2107.
https://doi.org/10.1016/s0140-6736(19)32556-5
[43] Lonial, S., Dimopoulos, M., Palumbo, A., White, D., Grosicki, S., Spicka, I., et al. (2015) Elotuzumab Therapy for Relapsed or Refractory Multiple Myeloma. New England Journal of Medicine, 373, 621-631.
https://doi.org/10.1056/nejmoa1505654
[44] Cho, S., Xing, L., Anderson, K.C. and Tai, Y. (2021) Promising Antigens for the New Frontier of Targeted Immunotherapy in Multiple Myeloma. Cancers, 13, Article 6136.
https://doi.org/10.3390/cancers13236136
[45] Dimopoulos, M.A., Lonial, S., White, D., Moreau, P., Weisel, K., San-Miguel, J., et al. (2020) Elotuzumab, Lenalidomide, and Dexamethasone in RRMM: Final Overall Survival Results from the Phase 3 Randomized ELOQUENT-2 Study. Blood Cancer Journal, 10, Article No. 91.
https://doi.org/10.1038/s41408-020-00357-4
[46] Dimopoulos, M.A., Dytfeld, D., Grosicki, S., Moreau, P., Takezako, N., Hori, M., et al. (2018) Elotuzumab Plus Pomalidomide and Dexamethasone for Multiple Myeloma. New England Journal of Medicine, 379, 1811-1822.
https://doi.org/10.1056/nejmoa1805762
[47] Kashima, E., Fujieda, A., Nato, Y., et al. (2020) Successful Treatment with a Combination of Elotuzumab, Lenalidomide and Dexamethasone of Extramedullary Disease in a Patient with Refractory Multiple Myeloma. Rinsho Ketsueki, 61, 223-227.
https://doi.org/10.11406/rinketsu.61.223
[48] Danhof, S., Rasche, L., Mottok, A., Steinmüller, T., Zhou, X., Schreder, M., et al. (2021) Elotuzumab for the Treatment of Extramedullary Myeloma: A Retrospective Analysis of Clinical Efficacy and SLAMF7 Expression Patterns. Annals of Hematology, 100, 1537-1546.
https://doi.org/10.1007/s00277-021-04447-6
[49] Chim, C.S., Kumar, S.K., Orlowski, R.Z., Cook, G., Richardson, P.G., Gertz, M.A., et al. (2017) Management of Relapsed and Refractory Multiple Myeloma: Novel Agents, Antibodies, Immunotherapies and Beyond. Leukemia, 32, 252-262.
https://doi.org/10.1038/leu.2017.329
[50] Ali, S.A., Shi, V., Maric, I., Wang, M., Stroncek, D.F., Rose, J.J., et al. (2016) T Cells Expressing an Anti-B-Cell Maturation Antigen Chimeric Antigen Receptor Cause Remissions of Multiple Myeloma. Blood, 128, 1688-1700.
https://doi.org/10.1182/blood-2016-04-711903
[51] Xu, J., Chen, L., Yang, S., Sun, Y., Wu, W., Liu, Y., et al. (2019) Exploratory Trial of a Biepitopic CAR T-Targeting B Cell Maturation Antigen in Relapsed/Refractory Multiple Myeloma. Proceedings of the National Academy of Sciences, 116, 9543-9551.
https://doi.org/10.1073/pnas.1819745116
[52] Deng, H., Liu, M., Yuan, T., Zhang, H., Cui, R., Li, J., et al. (2021) Efficacy of Humanized Anti-BCMA CAR T Cell Therapy in Relapsed/Refractory Multiple Myeloma Patients with and without Extramedullary Disease. Frontiers in Immunology, 12, Article 720571.
https://doi.org/10.3389/fimmu.2021.720571