Open Access

The numerous facets of 1q21+ in multiple myeloma: Pathogenesis, clinicopathological features, prognosis and clinical progress (Review)

  • Authors:
    • Na Liu
    • Zhanzhi Xie
    • Hao Li
    • Luqun Wang
  • View Affiliations

  • Published online on: April 9, 2024     https://doi.org/10.3892/ol.2024.14391
  • Article Number: 258
  • Copyright: © Liu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Multiple myeloma (MM) is a malignant neoplasm characterized by the clonal proliferation of abnormal plasma cells (PCs) in the bone marrow and recurrent cytogenetic abnormalities. The incidence of MM worldwide is on the rise. 1q21+ has been found in ~30‑40% of newly diagnosed MM (NDMM) patients.1q21+ is associated with the pathophysiological mechanisms of disease progression and drug resistance in MM. In the present review, the pathogenesis and clinicopathological features of MM patients with 1q21+ were studied, the key data of 1q21+ on the prognosis of MM patients were summarized, and the clinical treatment significance of MM patients with 1q21+ was clarified, in order to provide reference for clinicians to develop treatment strategies targeting 1q21+.

Introduction

Multiple myeloma (MM) is a neoplasm malignant characterized by the clonal proliferation of abnormal plasma cells (PCs) in the bone marrow and recurrent cytogenetic abnormalities that can lead to bone marrow failure, bone destruction, hypercalcemia, anaemia, infection, kidney dysfunction and neurologic symptoms (1,2). The incidence of MM worldwide is on the rise. In 2020, the age-standardized incidence rate of MM was 1.78 per 100,000 people globally, and the age-standardized mortality rate was 1.14 per 100,000 people globally (35). The occurrence, progression, treatment, resistance and prognosis of MM are all associated with cytogenetic abnormalities (68). Previous studies revealed that cytogenetic abnormalities, such as t(4;14), t(14;16), t(14;20), 1q21+, del(1p), and del(17p), can be used as predictors of poor prognosis (912). Among them, 1q21+ is one of the most common chromosomal abnormalities in MM, including 1q21 gain (three copies) and 1q21 amplification (≥4 copies) (13). 1q21+ has been found in ~30-40% of patients with newly diagnosed MM (NDMM) (14,15).

1q21+ is associated with the pathophysiological mechanisms of disease progression and drug resistance in MM (16,17). Besides, co-existence of certain high-risk cytogenetic abnormalities is common in some patients with MM, and further worsens the prognosis for 1q21+ patients (18,19). Therefore, both the International Myeloma Working Group (IMWG) and Mayo Stratification of Myeloma and Risk-Adapted Therapy (mSMART) 3.0 have taken 1q21+ as an important reference factor for risk stratification (2022). In recent years, the introduction of autologous hematopoietic stem cell transplantation (ASCT), immunomodulatory drugs (IMiDs), proteasome inhibitors (PIs), and an anti-CD38 monoclonal antibody have greatly improved the outcomes of patients with MM (2326). However, the overall survival (OS) of patients with MM and 1q21+ remains to be improved (27,28). In the present review, the pathogenesis and clinicopathological features of MM patients with 1q21+ were reviewed, the key data of 1q21+ on the prognosis of patients with MM were summarized and the clinical treatment significance of MM patients with 1q21+ were clarified, for the purpose of providing reference for clinicians to develop treatment strategies targeting 1q21+.

Pathogenesis and clinicopathological features of 1q21+ in patients with MM

The evolution of MM is a complex and progressive process in which initial carcinogenic events, namely primary immunoglobulin heavy chain (IGH) translocations and hyper-diploid karyotypes, have occurred in the early stages of monoclonal gammopathy of uncertain significance and smouldering MM, followed by disease progression to symptomatic MM (29,30) (Fig. 1). Chromosome arm 1q21+ is a secondary genomic event occurring in patients with MM (16). 1q21+ occurs in subclones of cells with primary translocations, manifesting as chromosome 1q duplications, unbalanced whole-arm translocations of chromosome 1q, or jumping translocations detected by G-banding (3033). Among them, jumping translocations involve unbalanced translocations between non-homologous chromosomes, caused by the exchange of the pericentromeric region of 1q with other chromosomal sites, which involves the de-condensation of pericentromeric chromatin of 1q and the intranuclear replication of 1q, and the subsequent fusion of the acquired region with the centromere or telomere end of another chromosome (34,35). The jumping translocation can increase the copy number of 1q, which may be the potential mechanism of 1q21+ (3036).

The genetic instability of 1q pericentromeric region of 1q12 and its neighbor 1q21, as well as the overexpression of certain genes that may be present during 1q21 amplification, are closely associated with disease progression in MM (14,36). Previous studies have found proliferation and dysregulation of G1/S cell cycle checkpoints in most MM cells with 1q21+ (30). 1q21+ can upregulate the activity of cyclin D by driving the oncogene CDC28 protein kinase regulatory subunit 1B (CKS1B), to further drive the proliferation of downstream cells and promote the circulation of PCs (3739). 1q21+ is a copy number alteration that is most relevant to transcriptome, and corresponds to genome-wide transcriptional regulation, rather than being restricted to this genomic region. It is associated with the upregulation of cyclin D2 (CCND2) and other oncogene-driven proliferation programs, such as PDL1 downregulation and Slam family member 7 (SLAMF7), G protein-coupled receptor class c group 5-member D (GPRC5D), and apoptosis regulator BCL2 family member (MCL1) upregulation predicted by 1q21 gain or amplification (40). Interleukin-6 receptor (IL6R) and ADAR1 (an RNA editing enzyme in 1q21 region near IL6R) are critical genes located within the minimally amplified 1q21 region. The 1q21 state is closely related to the levels of IL6R and ADAR1 (41,42). IL6R and ADAR1 collaborate to induce a hyper-activation of the STAT3 pathway, and high levels of IL6 in MM cells result in sustained activation of the STAT3 pathway (4345). The amplification of 1q21 leads to elevated expression of IL6R and ADAR1. High IL6R confers hypersensitivity to IL6 stimulation, leading to constitutive activation of the STAT3 pathway. However, high expression of ADAR1-P150 results in a hyper-activation of the STAT3 pathway, and ultimately increases the survival rate of MM cells (42). In addition, the expression of some genes (EANP32E, ARNT, BCL9, 2ILF2, MUC1, MCL1, NEK2, PSMD4, PDZK1 and SETDB1) is associated with 1q21+, promoting cell proliferation (31,4655).

In terms of epigenetic regulation, copy number gains of 1q21 originate in part by the hypomethylation of 1q12 pericentromeric heterochromatin (56). A total of 25% of the genes associated with MM-specific methylated regions lie within the 1q21.1 region. The demethylation of Family with sequence similarity 72 (FAM72) gene in the 1q21 region increases the expression of FAM72, while FAM72 promotes MM cell proliferation through the forkhead box M1 (FOXM1) transcription factor signaling pathway (57). Moreover, the transcription factor PBX homeobox (PBX)1 is ectopically expressed by genetic amplification and epigenetic activation of its own preserved regulatory domain. By binding to reprogrammed super-enhancers, PBX3 directly regulates critical oncogenic pathways and FOXM1-dependent transcription programs to activate MM cell proliferation (58).

In addition to gene drive, changes in the bone marrow microenvironment further contribute to the formation of a high-risk disease state with 1q21+ (30). Complement factor C1q, as an activator protein in the classical activation pathway, participates in the recognition stage of classical activation pathway and has the ability to regulate various immune cell responses (59). C1q has two classical cell surface receptors (C1qRs): cC1qR binds to the collagen ‘stalks’ tail, whereas gC1qR binds to the globular ‘heads’ (60). GC1qR promotes MM cell line survival by suppressing the MM-inhibiting role of C1q and contributing to the stabilization of the CKS1BmRNA through insulin-like growth factor 2 mRNA binding protein 3 (IGF2BP3) (61).

In patients with NDMM, the presence of 1q21+ is likely to be associated with other cytogenetic abnormalities (Table I). Previous studies revealed that the presence of 1q21+ was highly related to the co-existence of other high-risk cytogenetic abnormalities, such as t(4;14), t(14;16) and del(17p) (62,63). In addition, 1q21+ is also associated with some markers of disease burdens. Previous studies have shown that 1q21+ was significantly associated with the presence of β2-microglobulin >5.5 mg/l, haemoglobin <10 g/dl, platelet count <100 K/µl, glomerular filtration rate <30 ml/min/1.73 m2, lactate dehydrogenase (LDH) level >300 U/l, and plasma creatinine level (18,19,6468). A retrospective study suggested that patients with 1q21+ were more prone to experience anaemia, hypoalbuminemia, renal insufficiency, high LDH and a high proportion of R-ISS-III (69). Furthermore, they were more likely to be accompanied by del(13q14), t(4;14), and del(1p) (69).

Table I.

Clinicopathological features of patients with 1q21+ multiple myeloma (18,19,62–69).

Table I.

Clinicopathological features of patients with 1q21+ multiple myeloma (18,19,62–69).

CategoryClassificationContent
Cytogenetic abnormalitiesIgH rearrangementt(4;14), t(14;16)
coexisting with 1q21+Variation of chromosome armdel(13q), del(1p), del(17p)
Markers associated with 1q21+Markers of disease burdenβ2-microglobulin >5.5 mg/l, hemoglobin
<10 g/dl, platelet count <100 K/µl, glomerular
filtration rate <30 ml/min/1.73 m2, lactate
dehydrogenase >300 U/l, plasma creatinine
Markers of cell proliferationTK1 (positively correlated)
Markers of bone formationPINP (negative correlation)

[i] TK1, thymidine kinase 1; PINP, amino-terminal propeptide of type I procollagen.

Patients with 1q21+ present with a more aggressive clinical course. Osteopathy is one of the characteristic clinical manifestations of MM. Osteopathy, such as myeloma, is present in >80% of patients with MM at diagnosis, which ranges from osteoporosis to typical lytic lesions, and then to advanced lesions such as structural bone defects or pathological fractures (70). In a retrospective study of 308 patients with MM, the presence of 1q21 gain was associated with a higher level of thymidine kinase (TK) (P=0.026) and a lower level of amino-terminal propeptide of type I procollagen (PINP) (P=0.030) (71). Among them, TK is a cell proliferation marker. Elevated TK1 serum levels may correlate with high activity and high aggressiveness of MM cells, while PINP is closely related to various bone metabolism disorders, tumor bone metastasis and MM progression (72). Similarly, in a prospective study of 64 patients with MM, PINP levels differed dependent on the disease stage, with median concentrations decreasing in advanced stages of patients with MM (73). The standard deviation scores values of PINP were significantly lower in MM patients with advanced bone disease. A significantly lower concentration of PINP was found to be typical for patients with numerous (>3) bone lytic lesions (73). In addition, PINP inversely correlated with LDH. These studies suggested a possible impact of gain 1q21 on advanced bone disease, thus with possible impact on adverse prognosis of the patients with MM.

Impact of 1q21+ on prognosis

With the deepening understanding of MM, the influence or weight of various baseline risk factors in predicting the outcomes of patients with MM may change. Recently, two major prognostic scoring systems have been successively released; namely, the second revision of the International Staging System (R2-ISS) issued by the European Myeloma Network (EMN) and the Mayo Additive Staging System (MASS) developed by the Mayo Clinic based on single-center data (21,22). The R2-ISS stratifies patients with NDMM into the following risk groups: i) low (0), ii) low/intermediate (0.5–1), iii) intermediate/high (1.5–2.5) and iv) high (35) by assigning different weight scores to the six included variables: ISS II, ISS III, del(17p), high LDH, t(4;14) and 1q+ (22). The MASS includes IgH translocation, 1q+, chromosome 17 abnormality, ISS stage III, and elevated LDH as high risk (HR) factors. Patients with 0, 1, or ≥2 HR abnormalities are considered as stage I, II, or III, respectively (21). Both staging systems take 1q+ as a HR factor and use it as a basis for stratification, which has been supported by several recent studies (68,74). A meta-analysis involving 2,596 patients with MM found that 1q gain was associated with inferior survival in NDMM, irrespective of current standard therapies, and should be considered as an independent risk factor (68). Similarly, in a post-hoc analysis of another clinical study, 1q gain at relapse was associated with shorter OS, independent of other risk markers or time of relapse (74). This means that 1q gain can be an independent poor prognostic factor for both NDMM and relapsed MM. A retrospective study of 248 Chinese patients with NDMM detected that 1q21+ was an independent poor prognostic factor in patients with NDMM. According to the mSMART 3.0 stages, double-hit and triple-hit groups had the worst prognosis, with a median progression-free survival (PFS) of 22 months and a median OS of 32 months, respectively (69). Another retrospective study of 505 Chinese NDMM patients (47% with 1q21+) confirmed the prognostic stratification value of R2-ISS staging system in Chinese patients with NDMM and evaluated the impact of 1q21+ on survival. The median PFS and OS of patients carrying the 1q21+ mutation were 33.3 and 62.6 months, respectively, significantly shorter than those without 1q21 abnormality [PFS, 50.8 months; OS, not reached (NR); P<0.001] (75).

1q21 gain has long been considered as an early driver event, and 1q21 amplification a progression-related late event, both of which have negative prognostic implications, although the prognostic implications of increased copy number of 1q21+ are still controversial (Table II) (7685). In a cohort study of 912 symptomatic patients with MM, patients with 1q21+ showed inferior PFS (34 vs. 20 months, P<0.001) and OS (75 vs. 44 months, P<0.001) compared with those without 1q21+, but increased copies of 1q21+ had no effect on prognosis (18). Increasing evidence has identified that patients with MM and three copies of 1q21 had comparable survival with patients with more than three copies. A cohort study demonstrated that in 290 cases of patients with NDMM treated with bortezomib-based therapy and 1q21 copy number ≥3 significantly worse outcomes were observed; and there was no statistically significant difference between median PFS and OS in patients who had three, four, or at least five copies of 1q21 (86). This conclusion was confirmed in another study with an enlarged sample (667 patients) and extended follow-up (87). A real-world study which included 1,068 diagnosed MM Chinese patients reported that the variation in copy number of 1q+ (copy number <3) had no significant impact on the survival of MM patients with 1q abnormalities (88). Similarly, in a meta-analysis, there was no significant difference in the prognosis between 1q21 gain and 1q21 amplification for patients with thalidomide and those with bortezomib, both being associated with adverse outcome (68). There are also differences with the aforementioned views. Several studies have revealed that the prognosis of patients with MM is worse when 1q21 copy number ≥4; for instance, You et al (28) revealed that 1q21 amplification had worse PFS than 1q21 gain (24 months vs. not reached, P=0.0403). Neben et al (77) discovered that a 1q21 copy number of three has a marginal negative effect, and having more than three copies significantly reduces PFS and OS. Similarly, Schmidt et al (82) and Gao et al (89) both revealed that copy number of 1q21 ≥4 led to a poor prognosis. In a recent retrospective study of 794 patients with MM, the median PFS times of normal copy number of 1q, 1q gain and 1q amplification were 49, 50 and 26 months, respectively (P=0.268), and the median OS times were NR, NR and 41 months, respectively (P=0.001), suggesting that 1q21 amplification had a greater negative impact on prognosis than 1q21 gain (90). This might be related to the dose response of genes in the chromosome 1q region.

Table II.

Prognostic effects of 1q21+.

Table II.

Prognostic effects of 1q21+.

FeaturePrognosisNumber of patientsProportion of patients with 1q21+, %Detection threshold for 1q21+ by FISH, %PFS(Refs.)
1q21+ copy number ≥3 No significant912• 3 copies, 16.4203 vs. ≥4 copies: P>0.05(63)
effect • ≥4 copies, 10.9
290Thalidomide group, 48.1203 vs. 4 vs. >4 copies(86)
• 3 copies, 33.6 14.0 (95%CI, 8.07–19.93)
• ≥4 copies, 15.2 vs. 14.0 (95% CI, 5.47–22.53)
Bortezomib group, 47.5 vs. 10.0 (95%CI, 4.12–15.88)
• 3 copies, 27.4 months P>0.05
• ≥4 copies, 22.3
667• 3 copies, 28.2203 vs. ≥4 copies(87)
• ≥4 copies, 15.9 23.2 (18.7–27.7) vs.
22.0 (17.1–26.9)
months P>0.05
1,068• 3 copies, 19.553 vs. ≥4 copies: P>0.05(88)
• ≥4 copies, 12.1
Worse248• 3 copies, 38.7-3 vs. ≥4 copies:(28)
prognosis • ≥4 copies, 13.3 not reached vs.
24 months, P=0.0403
2013 copies, 25.9203 vs. ≥4 copies:(82)
≥4 copies, 12.9 74.5 (55.9–78.8) vs.
34.7 (15.6–61.1) months
1613 copies, 3653 vs. ≥4 copies:(89)
≥4 copies, 23.6 26.0 vs. 15.1 months
194-203 vs. ≥4 copies:(90)
50 vs. 26.0 months
1q21+ co-existence oft(4;14)No significant93453.15.5t(4;14) + 1q21 vs. 1q21:(63)
other high-risk effect P>0.05
cytogenetic 66710020t(4;14) + 1q21 vs. 1q21:(87)
abnormalities P=0.214
Worse prognosis20146.720t(4;14) + 1q21 vs. 1q21:(82)
HR 4.18, P=0.008
2131007.9t(4;14) + 1q21 vs. 1q21:(92)
HR, 3.00; P=0.047,
t(14;16)Worse prognosis93453.15.5t(14;16) + 1q21 vs. 1q21(63)
P<0.0001
5,141--t(14;16) + 1q21 vs. 1q21(91)
20.5 vs. 31.8 months, P=0.003
20146.720t(14;16) + 1q21 vs. 1q21:(82)
HR, 2.80; P=0.036
del(17p)Worse prognosis93453.15.5del(17p) + 1q21 vs. 1q21:(63)
P=0.0221
2131007.9del(17p) + 1q21 vs. 1q21:(92)
HR, 2.33, P=0.023
20146.720del(17p) + 1q21 vs. 1q21:
HR, 2.5; P= 0.01(82)

[i] FISH, fluorescence in situ hybridization; CI, confidence interval; HR, hazard ratio.

The coexistence of 1q21+ with other high-risk cytogenetic abnormalities often affects the prognosis of patients (Table II). A retrospective analysis included 201 patients with NDMM (including 107 patients with 1q21+) who received induction with lenalidomide, bortezomib and dexamethasone (RVD). In subgroup analyses, patients with co-occurring 1q+ and t(4;14), t(14;16) or del(17p) had a median PFS of 25.1 months (95% CI, 12.0–32.6 months), significantly lower than patients with only t(4;14), t(14;16) and/or del(17p) (P=0.02) (82). Another retrospective study of 934 patients with NDMM (53.1% of patients with 1q+), who received IMiDs or PIs, indicated that in 496 cases of patients with 1q+, 24.4% of the patients were accompanied by other high-risk cytogenetic abnormalities, such as del(17p), t(4;14) and t(14;16), and their PFS (P=0.0294) and OS (P=0.0381) were significantly worse than those of patients with 1q+ only (63). Among them, patients with co-occurring 1q+ and t(14;16) had the worst outcome, with significantly shorter PFS (P<0.0001) and OS (P=0.0004) (63). This is consistent with previous studies (82,91). Similar findings were also observed in patients with co-occurring 1q+ and del(17p), with significantly shorter PFS (P=0.0221) and OS (P=0.0046) (63). However, co-occurrence of 1q21+ and t(4;14) remains controversial. Schmidt et al (82) and Pasvolsky et al (92) found that co-occurrence of 1q21+ and t(4;14) was associated with worse PFS. Yang et al (63) identified that t(4;14) had no significant impact on PFS and OS in patients with 1q+. A retrospective study of 667 patients with NDMM revealed that patients with 1q21 gain and t(4; 14) had a worse prognosis, but the incidence and prognostic impact of 1q21 gain does not rely on the highly concurrent t(4;14) (87). 1q21 gain was strongly associated with a shorter median PFS and OS in both t(4;14) negative and positive patients with MM. By contrast, t(4;14) did not retain prognostic value in the 1q21 gain subgroup (87). Furthermore, the detrimental prognostic effect of 1q21+ was more profound in R-ISS-II and R-ISS-III patients. As testified by a cohort study, in R-ISS II patients, OS was significantly lower in those with 1q21+ than in those without 1q21+ (40.1 vs. 70.9 months, P=0.002), while in R-ISS III patients, OS was worse in those with 1q21+ than in those without 1q21+ (16.6 months vs. 43 months, P=0.028) (18).

Treatment of MM patients with 1q21+

ASCT

Currently, different graded treatment strategies are adopted for MM in clinical practice. The treatment strategies for NDMM are determined by ASCT and risk stratification (93). ASCT uses a very large dose of chemoradiotherapy to clear MM cells, destroy the autoimmune system, and transfuse autologous hematopoietic stem cells to rebuild hematopoietic and immune function, so as to achieve the purpose of further eliminating MM cells (9496). ASCT is the upfront choice for newly diagnosed patients with MM. If the patient is younger than 70 years old with favourable performance status, or older than 70 years old with favourable general performance status, ASCT should be the upfront choice after effective therapy (13,97). Previously, a real-world study of 1,068 Chinese patients with NDMM revealed that upfront ASCT could eliminate the adverse prognostic effect of 1q21 gain but not 1q21 amplification (98). Therefore, in clinical practice, ASCT is often combined with other medications as part of a normative overall treatment for NDMM patients (88,98100). A recent retrospective study included patients with NDMM who received ASCT combined with VRD (V, bortezomib; R, lenalidomide; D, dexamethasone) or VRD only for 8 cycles (69). The aforementioned study identified that the proportion of patients with 1q21+ was 54.4% and they showed a poor prognosis with the VRD regimen. However, ASCT could overcome the adverse effects significantly, and it played an important role in the prognosis of patients with 1q21+ (P<0.05) (69). Similarly, in a retrospective study of 1,491 patients with NDMM (100 patients in the 1q+/1p group) who received induction with IMiD combined with PI or bortezomib-cyclophosphamide-dexamethasone, with a pretreatment regimen of Busulfan in combination with melphalan or melphalan only, and who received ASCT, a median OS was not reached in the 1q+/1p-group compared with 81.1 months in the control group (HR, 1.25; CI, 0.3–4.6; P=0.73); the objective response rate (ORR) was 94% in both groups, and ASCT had a positive effect on ORR and OS compared with the historical control (98).

Immunomodulatory agents/proteasome inhibitors

NCCN guidelines state that VRD is recommended as a standard first-line therapy for patients with NDMM, regardless of suitability for transplantation (93,101,102). Thalidomide, lenalidomide and pomalidomide are all IMiDs. IMiDs exert their anti-myeloma activity mainly by binding cereblon (CRBN), the substrate receptor protein of the CRL4 E3 ubiquitin ligase (CRL4CRBN) complex, leading to rapid ubiquitination and degradation of two specific B-cell transcription factors, the Ikaros family of zinc-finger proteins Ikaros (IKZF1) and Aiolos (IKZF3) (103105). Thus, the direct target of IMiDs is CRBN (106,107). IMiDs can directly eliminate MM cells, promote the apoptosis of neovascular cells and enhance the immune reaction of natural killer cells (108110). PIs can covalently bind to the hydroxyl group of the N-terminal threonine of the proteasome and inhibit the normal degradation of intracellular proteins by the proteasome, resulting in myeloma cell cycle arrest and promoting MM cell apoptosis (111,112). The target of PI was the proteasome β5 subunit (113,114). Among them, bortezomib could reversibly inhibit the chymoprotease-like activity of proteasome β5 subunit (115). Carfilzomib inhibits chymotrypsin-like activity by irreversibly binding to the proteasome β5 subunit (116). Ixazomib preferentially binds to the 20S proteasome β5 subunit to inhibit chymotrypsin-like activity (114). A summary of studies on IMiDs and PIs in NDMM patients with 1q21+ over the past 3 years is presented in Table III (81,117121). In a previous study, patients with 1q21 gain receiving bortezomib-based treatment had significantly improved the OS and PFS when compared with non-bortezomib treatment (3-year PFS, 62.8 vs. 8.75%; P=0.0385; 3-year OS, 82.3 vs. 18.8%; P=0.0154) (81). In a cohort study, the combination of PIs and IMiDs produced improved outcomes in patients with 1q21+, but only partially alleviated the effects of 1q21+ (18). However, An et al (86) discovered that 1q21 gain had no significant impact on the prognosis of thalidomide-treated patients and that 1q21 gain had a median PFS (22.4 months vs. 20.0 months; P=0.625) and OS (30.0 vs. 22.0 months, P=0.355) compared with patients without 1q21+. The prognosis was significantly worse in patients treated with bortezomib, compared with patients without 1q21, patients with 1q21 had significant shorter PFS (13.5 months vs. 43.0 months; P<0.001) and OS (24.0 vs. 54.0 months, P<0.001) (86). Similarly, in the ENDURANCE ECOG-ACRIN E1A11 trial, 1q21+ was associated with poorer outcome either with VRD or carfilzomib + lenalidomide + dexamethasone (KRD) (118). This may be due to the high expression of 26S proteasome non-ATPase regulatory subunit 4 (PSMD4) in patients with 1q21+ as PSMD4 may mediate resistance to bortezomib by enhancing proteasome activity, resulting in enhanced protein degradation, decreased protein load and reduced apoptosis (51,122).

Table III.

Summary of studies on immunomodulatory drugs and proteasome inhibitors in patients with newly diagnosed multiple myeloma and 1q21+ over the past 3 years.

Table III.

Summary of studies on immunomodulatory drugs and proteasome inhibitors in patients with newly diagnosed multiple myeloma and 1q21+ over the past 3 years.

DrugTreatment regimenNumber of patientsProportion of patients with 1q21+, %Detection threshold for 1q21+ by FISH, %CRPFSOS(Refs.)
Bortezomib (VRD)The bortezomib group6346.0-Bortezomib group vs.Bortezomib group vs.Bortezomib(81)
is defined as patients non-bortezomibgroup:non-bortezomib groupgroup vs.
receiving 4 cycles of 57.14 vs. 21.43%in 3-year PFS: 62.8 vs.non-bortezomib
BTD chemotherapy. (P=0.01)8.75%; P=0.0385group in 3-year
The non-bortezomib OS: 82.3 vs.
group is defined as 18.8%;
patients receiving a P=0.0154
regimen free of
bortezomib, including
TCD, TD and VADT
Carfilzomib tripleKRD + ASCT: KRD47445.0-Normal 1q group vs.1q gain vs. normal 1q:1q gain vs.(121)
combination therapyX4 + ASCT + KRD 1q gain group vs. 1qHR 1.65, CI 1.14–2.37,normal 1q:
X4, KRD12, KCD + amplification group:P=0.007; 1q amplifi-HR 1.88
ASCT: KCD X4+ 52 vs. 50 vs. 38%cation vs. normal 1q:(CI, 0.98–3.58);
ASCT+ KRD X4a - HR, 3.04 (CI,P=0.056; 1q
1.99–4.65); P<0.001;amplification vs.
1q gain vs. normalnormal 1q:
1q: HR, 1.84 (CI,HR=5.88
1.21–2.81); P=0.004(CI, 3.10–11.17);
P<0.001.
1q gain vs. normal
1q: HR, 3.13
(CI, 1.73–5.68),
P<0.001
Bortezomib andAfter 3–4 cycles of25849.220NAIn patients with +1q,The median OS(117)
dexamethasoneinduction therapy the PFS time was 22.2was47.4 (95%
with bortezomib (95% CI, 15.8–28.5)CI, 34.7–59.5)
and dexamethasone, months and the 3- andmonths in patients
patients were divided 5-year PFS rate waswith +1q, whereas
into the ASCT and 35.1 and 15.3%,OS was not reached
non-ASCT groups respectively.in patients without
based on their age, In patients without+1q (P=0.048)
physical condition and +1q, the PFS was
willingness for ASCT 41.1 (95% CI 28.6–53.6)
months and the 3-and
5-year PFS rate was
51.3 and 26.7%,
respectively
VRD or KRDPatients were rando-1,08730.5-NAPatients with +1q theVRD, 3-year OS,(118)
mized to receive either median PFS was 2974 vs. 86% (HR,
bortezomib or carfilzo- months, while the1.47; P=0.069)
mib, combined with patients without +1qKRD, 3-year OS, 82
lenalidomide and was 42 months (VRD,vs. 88% (HR= 1.34;
dexamethasone, HR= 1.51; P=0.011;P=0.185)
followed by indefinite KRD, HR=1.63;
or 2-year maintenance P=0.002)
therapy with
lenalidomide
BortezomibRetrospective study:25066.820Patients with +1q vs.Patients with +1q vs.Patients with +1q(119)
Triple induction patients without +1q:patients without +1q invs. patients without
therapy with borte- 40.4 vs. 59.6%PFS (median, 35 vs.+1q in OS (median,
zomib, including VTD (P=0.587)55 months, P=0.05)74 vs. 168 months,
or VCD P=0.00025)
LenalidomidePatients were rando-55633-NAPFS of 1q gain patientsOS of 1q gain(120)
mized to receive in the lenalidomidepatients in the
lenalidomide or maintenance grouplenalidomide
lenalidomide + (HR, 1.5; P=0.2)maintenance group
vorinostat 100 days compared with the(HR=1.1; P=0.9)
after induction with observation groupcompared with the
CTD, CRD, or KCRD observation group
and ASCT

a In carfilzomib triple combination therapy, the cut-off level of 1q gain was 10% of nuclei with ≥3 copies of 1q, while 1q amplification was defined as 20% of nuclei with ≥4 copies of 1q. FISH, fluorescence in situ hybridization; CR, complete response; PFS, progression free survival; OS, overall survival; BTD, bortezomib, thalidomide and dexamethasone; TCD, thalidomide, cyclophosphamide and dexamethasone; TD, thalidomide and dexamethasone; VADT, vincristine, doxorubicin, dexamethasone and thalidomide; VCD, bortezomib, cyclophosphamide and dexamethasone; VRD, bortezomib, lenalidomide and dexamethasone; VTD, bortezomib, thalidomide and dexamethasone; ASCT, autologous hematopoietic stem cell transplantation; HR, high risk; CI, confidence interval; CTD, cyclophosphamide, dexamethasone and thalidomide; CRD, cyclophosphamide, lenalidomide, and dexamethasone; KRD, carfilzomib, lenalidomide and dexamethasone; KCRD, carfilzomib combined with CRD.

Subclonal cereblon mutations in patients treated with IMiDs are associated with MM recurrence and prognosis. The NCRI Myeloma XI study enrolled 178 patients with NDMM (19% of patients with 1q+) who received induction therapy with thalidomide, lenalidomide, or carfilzomib and lenalidomide combined with dexamethasone and cyclophosphamide. The aforementioned study found that progressive clonal expansion was a feature of 17.5% 1q gain cases, whereby 1q gain at diagnosis evolved into 1q amplification at relapse. Compared with normal 1q+, 1q+ from presentation and evolution of new gain (1q+) at relapse were both associated with significantly shorter OS (HR, 2.11; P=0.0040; and HR, 2.00; P=0.021, respectively) (71).

Ixazomib combined with lenalidomide-dexamethasone therapy has been shown to be beneficial in patients with relapsed/refractory MM. In a double-blind, randomized and placebo-controlled phase III clinical (TOURMALINE-MM1) study, 722 patients with relapsed/refractory MM were randomized 1:1 to IRD group (ixazomib + lenalidomide + dexamethasone) and RD group (lenalidomide + dexamethasone). The results revealed a 40% prolongation of PFS in the IRD group compared with the RD group (HR, 0.74; 95% CI, 0.59–0.94; P=0.01) (123). Post hoc analysis revealed a significant PFS benefit for the IRD group in the ‘expanded high-risk’ cohort including partial 1q21 amplification patients (HR, 0.66; 95% CI: 0.47–0.93). However, no significant difference was observed between the two groups in patients with 1q21 amplification only (HR, 0.78; 95% CI, 0.49–1.24 for a detection threshold of 3% for 1q21+ by fluorescence in situ hybridization (FISH); HR 0.682; 95% CI, 0.413–1.123 for a detection threshold of 20% for 1q21+ by FISH; HR, 0.683; 95% CI, 0.381–1.224 for a detection threshold of 60% for 1q21+ by FISH (124). This suggests that ixazomib has limited efficacy in patients with 1q21 amplification.

In addition, the copy number of 1q21+ affects the efficacy of IMiDs and PIs. In the FORTE study of induction and consolidation with carfilzomib, 474 transplantation-eligible patients with NDMM were randomized to receive either induction with KRD followed by ASCT and KRD (KRD_ASCT group), 12 KRD cycles (KRD12), or induction with K-cyclophosphamide (C)-d followed by ASCT and KCD (KCD_ASCT group). The proportion of patients with 1q+ in the aforementioned study was 55% (32% with 1q gain and 13% with 1q amplification). As indicated by the results, KRD-ASCT, compared with induction and consolidation with both KRD12 and KCD-ASCT, demonstrated significantly prolonged PFS with an HR of 0.64 (P=0.023) and 0.53 (P<0.001), respectively. 1q amplification presaged lower OS compared with normal 1q (HR, 5.88; 95% CI, 3.10–11.17; P<0.001) and 1q gain (HR, 3.13; 95% CI, 1.73–5.68; P<0.001). However, subgroup analyses based on early treatment suggested that KRD_ASCT completely abrogated the PFS disadvantage conferred by 1q gain (HR, 1.25 vs. normal 1q; 95% CI, 0.58–2.7; P=0.565), but patients with 1q amplification still exhibited a very poor outcome (121,125).

Monoclonal antibodies
CD38

CD38 antibodies have become a critical part of relapsed MM and its first-line therapy. CD38 is highly expressed by MM cells and is a cell surface receptor target for antibody therapy in patients with MM (126). The eliminating effect of CD38 antibody on tumor cells is mainly achieved through the Fc-mediated immune-effector mechanisms, including complement-dependent cytotoxicity, antibody-dependent cellular cytotoxicity, antibody-dependent cellular phagocytosis and secondary crosslinking-induced apoptosis (127). Daratumumab and Isatuximab, both CD38-targeting antibodies, have been approved by FDA. Daratumumab, an IgG1κ humanized monoclonal antibody that binds to CD38, reduces levels of myeloid-derived suppressor cells (CD38+MDSCs), regulatory T cells (CD38+Tregs), and B cells (CD38+Bregs) (128,129). Isatuximab is a chimeric humanized IgG1 monoclonal antibody that binds to a specific epitope on the human cell surface antigen CD38 (130). Isatuximab can induce MM cell death by caspase-dependent apoptosis and lysosome-mediated non-apoptotic cell elimination (131). A summary of studies on CD38 antibodies in patients with 1q21+ over the past 3 years is presented in Table IV.

Table IV.

Summary of studies on CD38 antibodies in patients with multiple myeloma and 1q21+ over the past 3 years.

Table IV.

Summary of studies on CD38 antibodies in patients with multiple myeloma and 1q21+ over the past 3 years.

DrugTreatment regimenNumber of patientsProportion of patients with 1q21+, %Detection threshold for 1q21+ by FISH, %CRPFSOS(Refs.)
Daratumumab4 cycles of D-RVD20730.0-RVD 1q+ vs. RVDD-RVD 1q+ vs.NA(132)
or RVD induction, 1q+: 57.6 vs. 57.1%RVd 1q+: NR vs.
ASCT, 2 cycles of (OR, 1.02; 95% CI,47.9 months
D-RVD or RVD 0.37–2.82)(OR, 0.42; 95% CI,
consolidation, and 0.14–1.27)
24 cycles of R ± D
maintenance therapy
DaratumumabDaratumumab23224.5-CR of patients with1q21+ vs. non-NA(134)
monotherapy, DRd, 1q21+ treated with1q21+: 24.5
DPd or DBd the Daratumumab-(95% CI,
based regimen:18.6–29.4) vs.
3 (5.26%);23.5 (95% CI,
There was no21.1–28.1)
significant differencemonths, P=0.392
in ORR between
patients with 1q21+
and patients without
1q21+ (P=0.09)
IsatuximabIsa-Pd vs. Pd30741.730NA1q21+: 9.5 vs.1q21+: 21.3 vs.(138)
3.8 months13.9 months
HR, 0.40 (95% CI,HR: 0.72 (95% CI,
0.25–0.63)0.48–1.07)
Non-1q21+: 11.6 vs. Non-1q21+:
9.5 months, HR,21.3 vs. 21.2
1.19 (95% CI,months, HR, 1.30
0.68–2.10)(95% CI,
0.80–2.14)
IsatuximabIsa-KD vs. KD3024130NA1q21+ Isa-KD vs.NA(138)
1q21+ KD: NR vs.
16.2 months
HR, 0.57 (95% CI,
0.33–0.98)

[i] FISH, fluorescence in situ hybridization; CR, complete response; PFS, progression-free survival; OS, overall survival; ASCT, autologous hematopoietic stem cell transplantation; R ± D, lenalidomide ± daratumumab; RVD, lenalidomide, bortezomib and dexamethasone; D-RVD, daratumumab combined with RVD; ORR, objective response rate; CI, confidence interval; HR, hazard ratio; OR, odds ratio; NR, not reached; DRd, daratumumab, lenalidomide and dexamethasone; DPd, daratumumab, pomalidomide and dexamethasone; DBd, daratumumab, bortezomib and dexamethasone; Isa-Pd, isatuximab, pomalidomide and dexamethasone; Pd, pomalidomide and dexamethasone; Isa-KD, isatuximab, carfilzomib and dexamethasone; KD, carfilzomib and dexamethasone.

Several studies have revealed that Daratumumab may improve the rate of MRD negativity and PFS in patients with newly diagnosed 1q21+, but has limited effect on patients with relapsed 1q21+. The GRIFFIN study included 207 transplantation-eligible patients with NDMM who were randomized to the D-RVD group (n=104) and the RVD group (n=103), and revealed that for patients with 1q21+, the rates of MRD negativity (10−5) in the D-RVD group and the RVD group were 61.8 and 28.6% (OR, 4.04; 95% CI, 1.38–11.81), respectively, and that the median PFS in the D-RVD group was higher (NR vs. 47.9 months) (OR, 0.42; 95% CI, 0.14–1.27) (132). This result suggests that the combination of D-RVD induction/consolidation with ASCT and R + DARA maintenance therapy increased the rate of MRD negativity and PFS in the 1q21+ subgroup. However, the poor prognosis of 1q21+ may be difficult to be overcome with Daratumumab in patients with MM and relapsed 1q21+. A study that investigated the outcomes of all patients with refractory MM receiving Daratumumab found that before initiating the treatment with Daratumumab, patients who were 1q21+ positive and classified as high-risk for GEP70 had the worst outcomes (0.3 and 0.8 years, respectively for PFS and OS), while patients without 1q21+ and low-risk for GEP70 did not reach the median PFS and OS (133). This observation indicates that a poor prognosis associated with 1q21+ may not be overcome by Daratumumab. Another multicenter retrospective study enrolled 232 RRMM patients who received a DARA-based regimen and underwent FISH following 1st-3rd line therapies (134). At a median follow-up of 35.7 months in the study, the median PFS for patients with 1q21+ using the DARA-based regimen was 24.6 months, and the ORR was 57.9%. There were no significant differences in PFS and ORR between patients with 1q21+ using the DARA regimen and patients without 1q21+. Similarly, there were also no significant differences in PFS or ORR among patients with 1q21+ using different treatment regimens. The prognosis and response profiles in patients with 1q21+ using the DARA regimen were similar to those with standard-risk cytogenetics (SRCyto) or other high-risk cytogenetics (HRCyto) (134). Although the prognosis in patients with 1q21+ using the DARA regimen in this study was similar to that in patients without 1q21+, given that median PFS and ORR were lower in patients with 1q21+ compared with historical controls (80,119,125) further studies are required to validate the role of Daratumumab in patients with relapsed 1q21+.

In patients with newly diagnosed high-risk 1q21+, Isatuximab may improve the rate of MRD negativity. Recently, an interim analysis of the GMMG-CONCEPT study was released. The aforementioned study included 153 high-risk patients with NDMM and aimed to evaluate the efficacy and safety of the Isa + KRD quadruplet (Isatuximab + carfilzomib + lenalidomide + dexamethasone) regimen in this population. The study considered patients with del17p or t(4;14) or t(14;16) or >3 copies of 1q21 with ISS II or III as high risk and enrolled 127 transplantation-eligible patients and 26 transplantation-ineligible patients, with 36.0% of patients with 1q21+. In the study, by the end of consolidation therapy, 63 (67.7%) cases in the transplantation-eligible (TE-ITT) group reached MRD negativity, 3 (3.2%) cases had MRD positivity, and 23 (24.7%) cases had not completed the consolidation therapy. In the transplantation-ineligible (TNE-ITT) group, 13 cases (54.2%) reached MRD negativity, no one had MRD positivity, and 11 (45.8%) cases had not completed the consolidation therapy. The ORR was 94.9% in the TE-ITT group, complete response (CR) was achieved in 57.7% of patients in the TNE-ITT group, and very good partial response (VGPR) was achieved in 30.8% of patients. Although no separate analysis was performed for patients with 1q21+, it still could be observed that the combination of Isatuximab based on KRD was effective in high-risk NDMM patients with 1q21+ and increased the rate of MRD negativity (135).

Unlike Daratumumab, the efficacy of Isatuximab in the treatment of relapsed/refractory MM with 1q21+ has been demonstrated in several studies. In the phase III studies ICARIA-MM and IKEMA, the addition of Isatuximab (Isa) to the backbone of pomalidomide-dexamethasone (PD) or carfilzomib-dexamethasone (KD), respectively, improved PFS among patients with relapsed/refractory MM, and subgroup analyses suggested benefit among patients with 1q21+ (136,137). Recently, four 1q21-related subgroups from ICARIA-MM and IKEMA were analyzed and showed improvement in median PFS and OS in patients with 1q21+ in the Isa-PD group when compared with the PD group. The median PFS was 9.5 and 3.8 months (HR, 0.40; 95% CI, 0.25–0.63) in patients with 1q21+, and the median OS was 21.3 and 13.9 months (HR, 0.72; 95% CI, 0.48–1.07). The median PFS and OS of patients with 1q21+ and patients without 1q21+ were similar in the Isa-PD group, with a median PFS of 9.5 and 11.6 months, respectively in both groups (HR=1.19; 95% CI, 0.68–2.10) and a median OS of 21.3 and 21.2 months, respectively (HR, 1.30; 95% CI, 0.80–2.14). In patients with 1q21+, the median PFS was improved in the Isa + KD group compared with the KD group (NR vs. 16.2 months, HR, 0.57; 95% CI, 0.33–0.98) (138). The aforementioned study has shown that Isatuximab combined with PD or KD can all improve the poor prognosis of 1q21+ in patients with relapsed/refractory MM.

Elotuzumab

Elotuzumab is a monoclonal antibody targeting signaling lymphocytic activation molecule family member 7 (SLAMF7), which is highly expressed by MM cells (139). Elotuzumab can not only directly act on SLAMF7 that is highly expressed on the surface of PCs to inhibit intercellular adhesion, thereby reducing the growth stimulation effect of stromal cells on myeloma cells, but also target, bind to and label SLAMF7 on the surface of myeloma cells, and enhance antibody-dependent cellular cytotoxicity in myeloma cells mediated by NK cells (140,141).

The SWOG-1211 study is a randomized, multicenter phase II clinical study that included 100 ASCT-ineligible patients with high-risk NDMM, where high-risk MM is defined as: High risk based on gene expression profiling analysis, t(14;16), t(14;20), del(17p) or 1q21+, primary plasma cell leukaemia and elevated serum LDH (≥2 times the upper limit of normal). The proportion of patients with 1q21+ was 47%. In the aforementioned study, there was no significant difference in prognosis between the RVD + elotuzumab group and the RVd group, with a PFS of 31 and 34 months, respectively (HR=0.968; 80% CI, 0.697–1.344; P=0.45). Similarly, in patients with 1q21+, the addition of elotuzumab did not improve the prognosis, with a median PFS of 41 months (95% CI, 22-NR) in the RVd group and 32 months (95% CI, 18-NR) in the RVD + elotuzumab group, with no statistical difference between the two groups (142).

Recently, a multicenter, single-arm phase II clinical study analyzed the efficacy and safety of elotuzumab combined with KRD (Elo-KRD) regimen in NDMM patients without an intent for ASCT. The study included 46 patients with NDMM, including 35% of patients with 1q21+. After 8 courses of Elo-KRD, a total of 26 (58%) patients achieved stringent complete response (sCR) and/or MRD negativity. Finally, 17 (38%) patients achieved sCR, 21 (47%) patients achieved CR or better, 38 (84%) patients achieved VGPR or better, and 39 (87%) patients achieved PR or better. In the evaluation of MRD, 26 (63%) patients of achieved MRD negativity following 8 courses of Elo-KRD, among whom 19 (44%) patients remained MRD negativity during the 8th to 12th courses and 15 (50%) patients sustained MRD negativity lasting for >1 year. This study demonstrated a significant increase in sCR in NDMM patients treated with elotuzumab + KRD but did not specifically evaluate the actual efficacy of elotuzumab in patients with 1q21+ (143).

Selective inhibitors of nuclear export

Selinexor is an oral selective inhibitor of nuclear export that has been approved by FDA for the treatment of relapsed/refractory MM. The target of Selinexor was exportin 1 (XPO1)/chromosome region maintenance 1, and selinexor reversibly inhibits tumor suppressor proteins (TSPs), growth regulators and oncogenic protein RNAs from the nucleus by blocking XPO1, which leads to nuclear accumulation of TSPs, reduction of oncoproteins such as c-myc and cyclinD1, cell cycle arrest and tumor cell apoptosis (144147).

The MARCH study is a multicenter, single-arm phase II clinical study that included 82 RRMM patients who had disease refractory to PI and IMiD, including 64.6% of patients with 1q21+ (148). In the aforementioned study, selinexor 80 mg combined with dexamethasone 20 mg was administered orally in patients on day 1 and day 3 of each week in 4-week cycle for multiple cycles. The results demonstrated an ORR of 29.3% (95% CI, 19.7–40.4), a median duration of response of 4.7 months, and median PFS and OS of 3.7 and 13.2 months, respectively (148). The aforementioned study did not analyze patients with 1q21+ separately but had a high proportion of patients with 1q21+. Similarly, the STOMP-XKd study, which involved 32 patients with RRMM, evaluated the safety and efficacy of selinexor (80 or 100 mg) + carfilzomib (56 or 70 mg/m2) + dexamethasone (40 mg) (XKD). The results showed that 53% of patients had high-risk cytogenetics del(17p), t(4;14), t(14;16) and/or gain 1q. The ORR was 78% after treatment with XKD, and median PFS was 15 months. In addition, XKD was well tolerated (149). Therefore, selinexor combined with dexamethasone could be used as a clinical option for the treatment of patients with RRMM, including 1q21+.

Conclusions

The interaction of MM cells with the bone marrow microenvironment generates a high-risk ecosystem that facilitates MM cell survival and immune response failure. 1q21+ is a secondary genomic event that occurs following primary IGH translocations. Therefore, the occurrence of 1q21+ is crucial for the evolution of myeloma cells to a high-risk state. In addition to the internal reasons such as genetic instability and gene driving in the pericentromeric region of chromosome 1, the genetic mechanism of 1q21+ also includes the protective effect of bone marrow microenvironment on MM cells. Therefore, the presence of 1q21+ predisposes to the risk of drug resistance and disease progression. The precise molecular mechanisms of acquiring 1q21+ and its effect on MM pathophysiology are yet to be fully elucidated. Further studies are required to assess the impact of the ecosystem formed by the bone marrow microenvironment and MM cells on 1q21+-induced resistance and aggressiveness.

As one of the most common cytogenetic abnormalities in MM, 1q21+ plays a role in the risk stratification of MM. In the past, both R-ISS and IMWG risk staging systems included 1q21+ in the intermediate-risk group (150,151). However, the 2016 IMWG consensus paper considered that patients with 1q21+ had a worse treatment outcome and 1q21+ was included in the high-risk group in the updated mSMART (152,153). Recently, both R2-ISS and MASS staging systems identified 1q21+ as a high-risk stratification factor (21,22). This indicates that the prognostic role of 1q21+ in MM has been widely recognized. In addition, although 1q21+ has been identified as a potentially poor prognostic factor, the effect of 1q21+ copy number on prognosis remains to be explored in more depth.

The application of CD138 immunomagnetic bead sorting combined with FISH technology is the most reliable method for the detection of 1q21+, but the threshold for 1q21+ in FISH testing varies significantly from <5-30% in various studies at present (89,154). EMN recommended 20% as the threshold for FISH testing (15), while most clinical trials generally do not report the threshold for 1q21+ by FISH testing or use a laboratory-defined threshold for evaluation. Inconsistencies in the thresholds for 1q21+ by FISH testing may result in differences in prognostic indicators such as PFS. If using 20% as the threshold for positive 1q21+, it might miss some patients with minor clones of 1q21+. For example, the retrospective study used 20 healthy donors' bone marrow specimens to set the thresholds for 1q21+ by MACS-FISH and direct-FISH and establish by a ‘mean + 3× standard deviation’ calculation, the cut-off values for each abnormality were 5% for 1q21+. The result indicated that 1q21+ clone sizes of 5–20% had clear adverse prognostic significance, and 5% was a reliable cut-off value for 1q21+ (89). If the EMN threshold had been applied to that study, then 25% of patients who tested positive for 1q21+ would have been considered negative. Therefore, there is currently a lack of reasonable threshold criteria for 1q21+ by FISH testing to improve evaluation of the prognostic status of patients with 1q21+.

The therapeutic limitation of MM is determined by clonal heterogeneity. 1q21+ is highly heterogeneous due to its association with the expression of multiple driver genes, posing numerous challenges to its treatment. IMiD/PI induction regimens alleviated the PFS disadvantage of 1q gain in some studies but did not improve the prognosis of 1q amplification. IMiD/PI induction, upfront high-dose therapy, and ASCT had a positive, but limited effect on the ORR and OS in patients with 1q21+. Daratumumab may improve the rate of MRD negativity and PFS in patients with newly diagnosed 1q21+, but there is a lack of clinical evidence that would be beneficial for patients with relapsed 1q21+. The actual efficacy in patients with 1q21+ was not specifically evaluated in studies of elotuzumab and selinexor. Isatuximab may improve the rate of MRD negativity in patients with newly diagnosed high-risk 1q21+, and in patients with RRMM, Isatuximab combined with PD or KD can all enhance the poor prognosis of 1q21+. Based on the existing results in the field of 1q21+, Isatuximab combined with IMiD and PI may be the therapy of the highest potential to improve the prognosis of patients with 1q21+.

In summary, as one of the most common cytogenetic abnormalities in MM, 1q21+ is closely related to the occurrence, progression, treatment, drug resistance and prognosis of MM. Patients with MM and 1q21+ tend to exhibit higher disease burden, more aggressive clinical manifestations, and more co-occurring high-risk cytogenetic abnormalities. Therefore, both R2-ISS and MASS staging systems identified 1q21+ as a high-risk stratification factor. The present review summarized the efficacy of ASCT, PI, IMiD, monoclonal antibodies and selective nuclear export inhibitors in the treatment of MM patients, especially in 1q21+ patients. Among them, isatuximab combined with IMiD or PI maybe the most potential therapy to improve the prognosis of patients with 1q21+ MM. However, for patients with 1q21+ who have no response to existing drug therapies, further studies on risk stratification are still needed to overcome the poor prognosis caused by clonal heterogeneity and provide personalized treatment options for them.

Acknowledgements

Not applicable.

Funding

The present study was supported by Sanofi.

Availability of data and materials

Not applicable.

Authors' contributions

LW was responsible for designing the theme of the review and ensuring that the descriptions are accurate and agreed by all authors. NL and ZX were responsible for ideas, collecting relevant studies from databases and writing of the manuscript. HL made critical modifications to important knowledge content within the manuscript. Data authentication is not applicable. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

ZX is a Sanofi employee and may hold shares and/or stock options in the company. The other authors declare that they have no competing interests.

References

1 

van de Donk NWCJ, Pawlyn C and Yong KL: Multiple myeloma. Lancet. 397:410–427. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Pop VS, Tomoaia G and Parvu A: Modern imaging techniques for monitoring patients with multiple myeloma. Med Pharm Rep. 95:377–384. 2022.PubMed/NCBI

3 

Huang J, Chan SC, Lok V, Zhang L, Lucero-Prisno DE III, Xu W, Zheng ZJ, Elcarte E, Withers M, Wong MCS, et al: The epidemiological landscape of multiple myeloma: A global cancer registry estimate of disease burden, risk factors, and temporal trends. Lancet Haematol. 9:e670–e677. 2022. View Article : Google Scholar : PubMed/NCBI

4 

Rajkumar SV: Multiple myeloma: 2022 Update on diagnosis, risk stratification, and management. Am J Hematol. 97:1086–1107. 2022. View Article : Google Scholar : PubMed/NCBI

5 

Hemminki K, Försti A, Houlston R and Sud A: Epidemiology, genetics and treatment of multiple myeloma and precursor diseases. Int J Cancer. 149:1980–1996. 2021. View Article : Google Scholar : PubMed/NCBI

6 

Dutta AK, Alberge JB, Sklavenitis-Pistofidis R, Lightbody ED, Getz G and Ghobrial IM: Single-cell profiling of tumour evolution in multiple myeloma-opportunities for precision medicine. Nat Rev Clin Oncol. 19:223–236. 2022. View Article : Google Scholar : PubMed/NCBI

7 

Bolli N, Avet-Loiseau H, Wedge DC, Van Loo P, Alexandrov LB, Martincorena I, Dawson KJ, Iorio F, Nik-Zainal S, Bignell GR, et al: Heterogeneity of genomic evolution and mutational profiles in multiple myeloma. Nat Commun. 5:29972014. View Article : Google Scholar : PubMed/NCBI

8 

Rajkumar SV: Updated diagnostic criteria and staging system for multiple myeloma. Am Soc Clin Oncol Educ Book. 35:e418–e423. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Perrot A, Lauwers-Cances V, Tournay E, Hulin C, Chretien ML, Royer B, Dib M, Decaux O, Jaccard A, Belhadj K, et al: Development and validation of a cytogenetic prognostic index predicting survival in multiple myeloma. J Clin Oncol. 37:1657–1665. 2019. View Article : Google Scholar : PubMed/NCBI

10 

Hanamura I: Multiple myeloma with high-risk cytogenetics and its treatment approach. Int J Hematol. 115:762–777. 2022. View Article : Google Scholar : PubMed/NCBI

11 

Caro J, Al Hadidi S, Usmani S, Yee AJ, Raje N and Davies FE: How to treat high-risk myeloma at diagnosis and relapse. Am Soc Clin Oncol Educ Book. 41:291–309. 2021. View Article : Google Scholar : PubMed/NCBI

12 

Neuse CJ, Lomas OC, Schliemann C, Shen YJ, Manier S, Bustoros M and Ghobrial IM: Genome instability in multiple myeloma. Leukemia. 34:2887–2897. 2020. View Article : Google Scholar : PubMed/NCBI

13 

Schmidt TM, Fonseca R and Usmani SZ: Chromosome 1q21 abnormalities in multiple myeloma. Blood Cancer J. 11:832021. View Article : Google Scholar : PubMed/NCBI

14 

Hanamura I, Stewart JP, Huang Y, Zhan F, Santra M, Sawyer JR, Hollmig K, Zangarri M, Pineda-Roman M, van Rhee F, et al: Frequent gain of chromosome band 1q21 in plasma-cell dyscrasias detected by fluorescence in situ hybridization: Incidence increases from MGUS to relapsed myeloma and is related to prognosis and disease progression following tandem stem-cell transplantation. Blood. 108:1724–1732. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Ross FM, Avet-Loiseau H, Ameye G, Gutiérrez NC, Liebisch P, O'Connor S, Dalva K, Fabris S, Testi AM, Jarosova M, et al: Report from the European myeloma network on interphase FISH in multiple myeloma and related disorders. Haematologica. 97:1272–1277. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Burroughs Garcìa J, Eufemiese RA, Storti P, Sammarelli G, Craviotto L, Todaro G, Toscani D, Marchica V and Giuliani N: Role of 1q21 in multiple myeloma: From pathogenesis to possible therapeutic targets. Cells. 10:13602021. View Article : Google Scholar : PubMed/NCBI

17 

Bisht K, Walker B, Kumar SK, Spicka I, Moreau P, Martin T, Costa LJ, Richter J, Fukao T, Macé S and van de Velde H: Chromosomal 1q21 abnormalities in multiple myeloma: A review of translational, clinical research, and therapeutic strategies. Expert Rev Hematol. 14:1099–1114. 2021. View Article : Google Scholar : PubMed/NCBI

18 

Kastritis E, Migkou M, Dalampira D, Gavriatopoulou M, Fotiou D, Roussou M, Kanellias N, Ntanasis-Stathopoulos I, Malandrakis P, Theodorakakou F, et al: Chromosome 1q21 aberrations identify ultra high-risk myeloma with prognostic and clinical implications. Am J Hematol. 97:1142–1149. 2022. View Article : Google Scholar : PubMed/NCBI

19 

Li X, Chen W, Wu Y, Li J, Chen L, Fang B, Feng Y, Liu J, Chen M, Gu J, et al: 1q21 gain combined with high-risk factors is a heterogeneous prognostic factor in newly diagnosed multiple myeloma: A multicenter study in China. Oncologist. 24:e1132–e1140. 2019. View Article : Google Scholar : PubMed/NCBI

20 

Garifullin A, Voloshin S, Shuvaev V, Martynkevich I, Kleina E, Chechetkin A, Bessmeltcev S, Kuzyaeva A, Sсhmidt A, Kuvshinov A and Pavlova I: Significance of modified risk stratification msmart 3.0 and autologous stem cell transplantation for patients with newly diagnosed multiple myeloma. Blood. 134 (Suppl 1):S55932019. View Article : Google Scholar

21 

Abdallah NH, Binder M, Rajkumar SV, Greipp PT, Kapoor P, Dispenzieri A, Gertz MA, Baughn LB, Lacy MQ, Hayman SR, et al: A simple additive staging system for newly diagnosed multiple myeloma. Blood Cancer J. 12:212022. View Article : Google Scholar : PubMed/NCBI

22 

D'Agostino M, Cairns DA, Lahuerta JJ, Wester R, Bertsch U, Waage A, Zamagni E, Mateos MV, Dall'Olio D, van de Donk NWCJ, et al: Second revision of the international staging system (R2-ISS) for overall survival in multiple myeloma: A European myeloma network (EMN) report within the HARMONY project. J Clin Oncol. 40:3406–3418. 2022. View Article : Google Scholar : PubMed/NCBI

23 

Pasvolsky O, Milton DR, Rauf M, Tanner MR, Bashir Q, Srour S, Tang G, Saini N, Ramdial J, Masood A, et al: Lenalidomide-based maintenance after autologous hematopoietic stem cell transplantation for patients with high-risk multiple myeloma. Transplant Cell Ther. 28:752.e1–752.e6. 2022. View Article : Google Scholar : PubMed/NCBI

24 

Villalba A, Gonzalez-Rodriguez AP, Arzuaga-Mendez J, Puig N, Arnao M, Arguiñano JM, Jimenez M, Canet M, Teruel AI, Sola M, et al: Single versus tandem autologous stem-cell transplantation in patients with newly diagnosed multiple myeloma and high-risk cytogenetics. A retrospective, open-label study of the PETHEMA/Spanish myeloma group (GEM). Leuk Lymphoma. 63:3438–3447. 2022. View Article : Google Scholar : PubMed/NCBI

25 

Barbieri E, Maccaferri M, Leonardi G, Giacobbi F, Corradini G, Lagreca I, Barozzi P, Potenza L, Marasca R and Luppi M: Adverse outcome associated with daratumumab-based treatments in relapsed/refractory multiple myeloma patients with amplification of chromosome arm 1q21: A single-center retrospective experience. Ann Hematol. 101:2777–2779. 2022. View Article : Google Scholar : PubMed/NCBI

26 

Chen L, Li Z, Li S, Fu W and Li R: Prognostic value and efficacy evaluation of novel drugs for multiple myeloma patients with 1q21 amplification (Amp1q21) only: A systematic review of randomized controlled trials. J Cancer. 11:2639–2644. 2020. View Article : Google Scholar : PubMed/NCBI

27 

Wang YT, Bao L, Chu B, Chen XH, Lu MQ, Shi L, Gao S, Fang LJ, Xiang QQ and Ding YH: Amp 1q21 is more predictable with dismal survival than gain 1q21 of newly diagnosed multiple myeloma in real-world analysis. J Clin Lab Anal. 36:e243752022. View Article : Google Scholar : PubMed/NCBI

28 

You H, Jin S, Wu C, Wang Q, Yan S, Yao W, Shi X, Shang J, Yan L, Yao Y, et al: The independent adverse prognostic significance of 1q21 gain/amplification in newly diagnosed multiple myeloma patients. Front Oncol. 12:9383922022. View Article : Google Scholar : PubMed/NCBI

29 

Manier S, Salem KZ, Park J, Landau DA, Getz G and Ghobrial IM: Genomic complexity of multiple myeloma and its clinical implications. Nat Rev Clin Oncol. 14:100–113. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Pawlyn C and Morgan GJ: Evolutionary biology of high-risk multiple myeloma. Nat Rev Cancer. 17:543–556. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Sawyer JR, Tricot G, Lukacs JL, Binz RL, Tian E, Barlogie B and Shaughnessy J Jr: Genomic instability in multiple myeloma: Evidence for jumping segmental duplications of chromosome arm 1q. Genes Chromosomes Cancer. 42:95–106. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Walker BA, Wardell CP, Murison A, Boyle EM, Begum DB, Dahir NM, Proszek PZ, Melchor L, Pawlyn C, Kaiser MF, et al: APOBEC family mutational signatures are associated with poor prognosis translocations in multiple myeloma. Nat Commun. 6:69972015. View Article : Google Scholar : PubMed/NCBI

33 

Saxe D, Seo EJ, Bergeron MB and Han JY: Recent advances in cytogenetic characterization of multiple myeloma. Int J Lab Hematol. 41:5–14. 2019. View Article : Google Scholar : PubMed/NCBI

34 

Sawyer JR, Tian E, Heuck CJ, Epstein J, Johann DJ, Swanson CM, Lukacs JL, Johnson M, Binz R, Boast A, et al: Jumping translocations of 1q12 in multiple myeloma: A novel mechanism for deletion of 17p in cytogenetically defined high-risk disease. Blood. 123:2504–2512. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Sawyer JR, Tricot G, Mattox S, Jagannath S and Barlogie B: Jumping translocations of chromosome 1q in multiple myeloma: Evidence for a mechanism involving decondensation of pericentromeric heterochromatin. Blood. 91:1732–1741. 1998. View Article : Google Scholar : PubMed/NCBI

36 

Sawyer JR, Tian E, Walker BA, Wardell C, Lukacs JL, Sammartino G, Bailey C, Schinke CD, Thanendrarajan S, Davies FE, et al: An acquired high-risk chromosome instability phenotype in multiple myeloma: Jumping 1q syndrome. Blood Cancer J. 9:622019. View Article : Google Scholar : PubMed/NCBI

37 

Bergsagel PL, Kuehl WM, Zhan F, Sawyer J, Barlogie B and Shaughnessy J Jr: Cyclin D dysregulation: An early and unifying pathogenic event in multiple myeloma. Blood. 106:296–303. 2005. View Article : Google Scholar : PubMed/NCBI

38 

Hose D, Rème T, Hielscher T, Moreaux J, Messner T, Seckinger A, Benner A, Shaughnessy JD Jr, Barlogie B, Zhou Y, et al: Proliferation is a central independent prognostic factor and target for personalized and risk-adapted treatment in multiple myeloma. Haematologica. 96:87–95. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Shaughnessy J: Amplification and overexpression of CKS1B at chromosome band 1q21 is associated with reduced levels of p27Kip1 and an aggressive clinical course in multiple myeloma. Hematology. 10 (Suppl 1):S117–S126. 2005. View Article : Google Scholar

40 

Ziccheddu B, Da Vià MC, Lionetti M, Maeda A, Morlupi S, Dugo M, Todoerti K, Oliva S, D'Agostino M, Corradini P, et al: Functional impact of genomic complexity on the transcriptome of multiple myeloma. Clin Cancer Res. 27:6479–6490. 2021. View Article : Google Scholar : PubMed/NCBI

41 

Barillé S, Bataille R and Amiot M: The role of interleukin-6 and interleukin-6/interleukin-6 receptor-alpha complex in the pathogenesis of multiple myeloma. Eur Cytokine Netw. 11:546–551. 2000.PubMed/NCBI

42 

Teoh PJ, Chung TH, Chng PYZ, Toh SHM and Chng WJ: IL6R-STAT3-ADAR1 (P150) interplay promotes oncogenicity in multiple myeloma with 1q21 amplification. Haematologica. 105:1391–1404. 2020. View Article : Google Scholar : PubMed/NCBI

43 

Gadó K, Domján G, Hegyesi H and Falus A: Role of interleukin-6 in the pathogenesis of multiple myeloma. Cell Biol Int. 24:195–209. 2000. View Article : Google Scholar : PubMed/NCBI

44 

Song Z, Ren D, Xu X and Wang Y: Molecular cross-talk of IL-6 in tumors and new progress in combined therapy. Thorac Cancer. 9:669–675. 2018. View Article : Google Scholar : PubMed/NCBI

45 

Quintanilla-Martinez L, Kremer M, Specht K, Calzada-Wack J, Nathrath M, Schaich R, Höfler H and Fend F: Analysis of signal transducer and activator of transcription 3 (Stat 3) pathway in multiple myeloma: Stat 3 activation and cyclin D1 dysregulation are mutually exclusive events. Am J Pathol. 162:1449–1461. 2003. View Article : Google Scholar : PubMed/NCBI

46 

Treon SP, Maimonis P, Bua D, Young G, Raje N, Mollick J, Chauhan D, Tai YT, Hideshima T, Shima Y, et al: Elevated soluble MUC1 levels and decreased anti-MUC1 antibody levels in patients with multiple myeloma. Blood. 96:3147–3153. 2000. View Article : Google Scholar : PubMed/NCBI

47 

Inoue J, Otsuki T, Hirasawa A, Imoto I, Matsuo Y, Shimizu S, Taniwaki M and Inazawa J: Overexpression of PDZK1 within the 1q12-q22 amplicon is likely to be associated with drug-resistance phenotype in multiple myeloma. Am J Pathol. 165:71–81. 2004. View Article : Google Scholar : PubMed/NCBI

48 

Legartova S, Krejci J, Harnicarova A, Hajek R, Kozubek S and Bartova E: Nuclear topography of the 1q21 genomic region and Mcl-1 protein levels associated with pathophysiology of multiple myeloma. Neoplasma. 56:404–413. 2009. View Article : Google Scholar : PubMed/NCBI

49 

Slomp A, Moesbergen LM, Gong JN, Cuenca M, von dem Borne PA, Sonneveld P, Huang DCS, Minnema MC and Peperzak V: Multiple myeloma with 1q21 amplification is highly sensitive to MCL-1 targeting. Blood Adv. 3:4202–4214. 2019. View Article : Google Scholar : PubMed/NCBI

50 

Walker BA, Leone PE, Chiecchio L, Dickens NJ, Jenner MW, Boyd KD, Johnson DC, Gonzalez D, Dagrada GP, Protheroe RK, et al: A compendium of myeloma-associated chromosomal copy number abnormalities and their prognostic value. Blood. 116:e56–e65. 2010. View Article : Google Scholar : PubMed/NCBI

51 

Shaughnessy JD Jr, Qu P, Usmani S, Heuck CJ, Zhang Q, Zhou Y, Tian E, Hanamura I, van Rhee F, Anaissie E, et al: Pharmacogenomics of bortezomib test-dosing identifies hyperexpression of proteasome genes, especially PSMD4, as novel high-risk feature in myeloma treated with total therapy 3. Blood. 118:3512–3524. 2011. View Article : Google Scholar : PubMed/NCBI

52 

Zhou W, Yang Y, Xia J, Wang H, Salama ME, Xiong W, Xu H, Shetty S, Chen T, Zeng Z, et al: NEK2 induces drug resistance mainly through activation of efflux drug pumps and is associated with poor prognosis in myeloma and other cancers. Cancer Cell. 23:48–62. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Marchesini M, Ogoti Y, Fiorini E, Aktas Samur A, Nezi L, D'Anca M, Storti P, Samur MK, Ganan-Gomez I, Fulciniti MT, et al: ILF2 is a regulator of RNA splicing and DNA damage response in 1q21-amplified multiple myeloma. Cancer Cell. 32:88–100.e6. 2017. View Article : Google Scholar : PubMed/NCBI

54 

Wu C, Yang T, Liu Y, Lu Y, Yang Y, Liu X, Liu X, Ye L, Sun Y, Wang X, et al: ARNT/HIF-1β links high-risk 1q21 gain and microenvironmental hypoxia to drug resistance and poor prognosis in multiple myeloma. Cancer Med. 7:3899–3911. 2018. View Article : Google Scholar : PubMed/NCBI

55 

Xiang J, Chen X, Chen M and Hou J: Increased expression of SETDB1 predicts poor prognosis in multiple myeloma. Biomed Res Int. 2022:33078732022. View Article : Google Scholar : PubMed/NCBI

56 

Sawyer JR, Tian E, Heuck CJ, Johann DJ, Epstein J, Swanson CM, Lukacs JL, Binz RL, Johnson M, Sammartino G, et al: Evidence of an epigenetic origin for high-risk 1q21 copy number aberrations in multiple myeloma. Blood. 125:3756–3759. 2015. View Article : Google Scholar : PubMed/NCBI

57 

Chatonnet F, Pignarre A, Sérandour AA, Caron G, Avner S, Robert N, Kassambara A, Laurent A, Bizot M, Agirre X, et al: The hydroxymethylome of multiple myeloma identifies FAM72D as a 1q21 marker linked to proliferation. Haematologica. 105:774–783. 2020. View Article : Google Scholar : PubMed/NCBI

58 

Trasanidis N, Katsarou A, Ponnusamy K, Shen YA, Kostopoulos IV, Bergonia B, Keren K, Reema P, Xiao X, Szydlo RM, et al: Systems medicine dissection of chr1q-amp reveals a novel PBX1-FOXM1 axis for targeted therapy in multiple myeloma. Blood. 139:1939–1953. 2022. View Article : Google Scholar : PubMed/NCBI

59 

Reid KBM: Complement component C1q: Historical perspective of a functionally versatile, and structurally unusual, serum protein. Front Immunol. 9:7642018. View Article : Google Scholar : PubMed/NCBI

60 

Peerschke EI and Ghebrehiwet B: cC1qR/CR and gC1qR/p33: Observations in cancer. Mol Immunol. 61:100–109. 2014. View Article : Google Scholar : PubMed/NCBI

61 

Xu J, Sun Y, Jiang J, Xu Z, Li J, Xu T and Liu P: Globular C1q receptor (gC1qR/p32/HABP1) suppresses the tumor-inhibiting role of C1q and promotes tumor proliferation in 1q21-amplified multiple myeloma. Front Immunol. 11:12922020. View Article : Google Scholar : PubMed/NCBI

62 

Chen H, Zhou N, Shi H, Yu W, Wu L and Zhou F: Presentation and outcomes of patients with multiple myeloma harboring gain or amplification of 1q21 and receiving novel agent therapies: Results from a single-center study. Hematology. 28:21779792023. View Article : Google Scholar : PubMed/NCBI

63 

Yang P, Chen H, Liang X, Xu W, Yu S, Huang W, Yi X, Guo Q, Tian M, Yue T, et al: Proposed risk-scoring model for estimating the prognostic impact of 1q gain in patients with newly diagnosed multiple myeloma. Am J Hematol. 98:251–263. 2023. View Article : Google Scholar : PubMed/NCBI

64 

Avet-Loiseau H, Attal M, Moreau P, Charbonnel C, Garban F, Hulin C, Leyvraz S, Michallet M, Yakoub-Agha I, Garderet L, et al: Genetic abnormalities and survival in multiple myeloma: The experience of the Intergroupe Francophone du Myélome. Blood. 109:3489–3495. 2007. View Article : Google Scholar : PubMed/NCBI

65 

Wu KL, Beverloo B, Lokhorst HM, Segeren CM, van der Holt B, Steijaert MM, Westveer PH, Poddighe PJ, Verhoef GE, Sonneveld P, et al: Abnormalities of chromosome 1p/q are highly associated with chromosome 13/13q deletions and are an adverse prognostic factor for the outcome of high-dose chemotherapy in patients with multiple myeloma. Br J Haematol. 136:615–623. 2007. View Article : Google Scholar : PubMed/NCBI

66 

Bang SM, Kim YR, Cho HI, Chi HS, Seo EJ, Park CJ, Yoo SJ, Kim HC, Chun HG, Min HC, et al: Identification of 13q deletion, trisomy 1q, and IgH rearrangement as the most frequent chromosomal changes found in Korean patients with multiple myeloma. Cancer Genet Cytogenet. 168:124–132. 2006. View Article : Google Scholar : PubMed/NCBI

67 

Bock F, Lu G, Srour SA, Gaballa S, Lin HY, Baladandayuthapani V, Honhar M, Stich M, Shah ND, Bashir Q, et al: Outcome of patients with multiple myeloma and CKS1B gene amplification after autologous hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 22:2159–2164. 2016. View Article : Google Scholar : PubMed/NCBI

68 

Weinhold N, Salwender HJ, Cairns DA, Raab MS, Waldron G, Blau IW, Bertsch U, Hielscher T, Morgan GJ, Jauch A, et al: Chromosome 1q21 abnormalities refine outcome prediction in patients with multiple myeloma-a meta-analysis of 2,596 trial patients. Haematologica. 106:2754–2758. 2021. View Article : Google Scholar : PubMed/NCBI

69 

You H, Jin S, Wu C, Wang Q, Yan S, Zhai Y, Yao W, Shi X, Shang J, Yan L, et al: The independent adverse prognostic significance of 1q21 gain/amplification in newly diagnosed multiple myeloma patients. Blood. 140 (Suppl 1):S10056–S10057. 2022. View Article : Google Scholar

70 

Kyle RA, Gertz MA, Witzig TE, Lust JA, Lacy MQ, Dispenzieri A, Fonseca R, Rajkumar SV, Offord JR, Larson DR, et al: Review of 1027 patients with newly diagnosed multiple myeloma. Mayo Clin Proc. 78:21–33. 2003. View Article : Google Scholar : PubMed/NCBI

71 

Krhovska P, Pika T, Proskova J, Balcarkova J, Zapletalova J, Bacovsky J and Minarik J: Bone metabolism parameters and their relation to cytogenetics in multiple myeloma. Eur J Haematol. 109:75–82. 2022. View Article : Google Scholar : PubMed/NCBI

72 

D'Oronzo S, Brown J and Coleman R: The role of biomarkers in the management of bone-homing malignancies. J Bone Oncol. 9:1–9. 2017. View Article : Google Scholar : PubMed/NCBI

73 

Kowalska M, Druzd-Sitek A, Fuksiewicz M, Kotowicz B, Chechlinska M, Syczewska M, Walewski J and Kaminska J: Procollagen I amino-terminal propeptide as a potential marker for multiple myeloma. Clin Biochem. 43:604–608. 2010. View Article : Google Scholar : PubMed/NCBI

74 

Croft J, Ellis S, Sherborne AL, Sharp K, Price A, Jenner MW, Drayson MT, Owen RG, Chown S, Lindsay J, et al: Copy number evolution and its relationship with patient outcome-an analysis of 178 matched presentation-relapse tumor pairs from the Myeloma XI trial. Leukemia. 35:2043–2053. 2021. View Article : Google Scholar : PubMed/NCBI

75 

Yan W, Fan H, Xu J, Liu J, Li L, Du C, Deng S, Sui W, Xu Y, Zou D, et al: Prognostic value of the second revision of the international staging system (R2-ISS) in a real-world cohort of patients with newly-diagnosed multiple myeloma. Chin Med J (Engl). 136:1744–1746. 2023. View Article : Google Scholar : PubMed/NCBI

76 

Nemec P, Zemanova Z, Greslikova H, Michalova K, Filkova H, Tajtlova J, Kralova D, Kupska R, Smetana J, Krejci M, et al: Gain of 1q21 is an unfavorable genetic prognostic factor for multiple myeloma patients treated with high-dose chemotherapy. Biol Blood Marrow Transplant. 16:548–554. 2010. View Article : Google Scholar : PubMed/NCBI

77 

Neben K, Lokhorst HM, Jauch A, Bertsch U, Hielscher T, van der Holt B, Salwender H, Blau IW, Weisel K, Pfreundschuh M, et al: Administration of bortezomib before and after autologous stem cell transplantation improves outcome in multiple myeloma patients with deletion 17p. Blood. 119:940–948. 2012. View Article : Google Scholar : PubMed/NCBI

78 

Nahi H, Våtsveen TK, Lund J, Heeg BM, Preiss B, Alici E, Møller MB, Wader KF, Møller HE, Grøseth LA, et al: Proteasome inhibitors and IMiDs can overcome some high-risk cytogenetics in multiple myeloma but not gain 1q21. Eur J Haematol. 96:46–54. 2016. View Article : Google Scholar : PubMed/NCBI

79 

Shah GL, Landau H, Londono D, Devlin SM, Kosuri S, Lesokhin AM, Lendvai N, Hassoun H, Chung DJ, Koehne G, et al: Gain of chromosome 1q portends worse prognosis in multiple myeloma despite novel agent-based induction regimens and autologous transplantation. Leuk Lymphoma. 58:1823–1831. 2017. View Article : Google Scholar : PubMed/NCBI

80 

Shah V, Sherborne AL, Walker BA, Johnson DC, Boyle EM, Ellis S, Begum DB, Proszek PZ, Jones JR, Pawlyn C, et al: Prediction of outcome in newly diagnosed myeloma: A meta-analysis of the molecular profiles of 1905 trial patients. Leukemia. 32:102–110. 2018. View Article : Google Scholar : PubMed/NCBI

81 

Chen D, Zhou D, Xu J, Zhou R, Ouyang J and Chen B: Prognostic value of 1q21 gain in multiple myeloma. Clin Lymphoma Myeloma Leuk. 19:e159–e164. 2019. View Article : Google Scholar : PubMed/NCBI

82 

Schmidt TM, Barwick BG, Joseph N, Heffner LT, Hofmeister CC, Bernal L, Dhodapkar MV, Gupta VA, Jaye DL, Wu J, et al: Gain of chromosome 1q is associated with early progression in multiple myeloma patients treated with lenalidomide, bortezomib, and dexamethasone. Blood Cancer J. 9:942019. View Article : Google Scholar : PubMed/NCBI

83 

Walker BA, Mavrommatis K, Wardell CP, Ashby TC, Bauer M, Davies F, Rosenthal A, Wang H, Qu P, Hoering A, et al: A high-risk, double-hit, group of newly diagnosed myeloma identified by genomic analysis. Leukemia. 33:159–170. 2019. View Article : Google Scholar : PubMed/NCBI

84 

Abdallah N, Greipp P, Kapoor P, Gertz MA, Dispenzieri A, Baughn LB, Lacy MQ, Hayman SR, Buadi FK, Dingli D, et al: Clinical characteristics and treatment outcomes of newly diagnosed multiple myeloma with chromosome 1q abnormalities. Blood Adv. 4:3509–3519. 2020. View Article : Google Scholar : PubMed/NCBI

85 

Gao W, Jian Y, Du J, Li X, Zhou H, Zhang Z, Yang G, Wang G, Tian Y, Li Y, et al: Gain of 1q21 is an adverse prognostic factor for multiple myeloma patients treated by autologous stem cell transplantation: A multicenter study in China. Cancer Med. 9:7819–7829. 2020. View Article : Google Scholar : PubMed/NCBI

86 

An G, Xu Y, Shi L, Shizhen Z, Deng S, Xie Z, Sui W, Zhan F and Qiu L: Chromosome 1q21 gains confer inferior outcomes in multiple myeloma treated with bortezomib but copy number variation and percentage of plasma cells involved have no additional prognostic value. Haematologica. 99:353–359. 2014. View Article : Google Scholar : PubMed/NCBI

87 

Du C, Mao X, Xu Y, Yan Y, Yuan C, Du X, Liu J, Fan H, Wang Q, Sui W, et al: 1q21 gain but not t(4;14) indicates inferior outcomes in multiple myeloma treated with bortezomib. Leuk Lymphoma. 61:1201–1210. 2020. View Article : Google Scholar : PubMed/NCBI

88 

Wang Y, Xu J, Xu B, Li P, Yang Y, Wang W, Xu T, Maihemaiti A, Lan T, Wang P, et al: The prognostic role of 1q21 gain/amplification in newly diagnosed multiple myeloma: The faster, the worse. Cancer. 129:1005–1016. 2023. View Article : Google Scholar : PubMed/NCBI

89 

Gao L, Liu Y, Li Y, Feng L, Wang Z, Wen L, Wang F, Huang X, Lu J and Lai Y: The importance of FISH signal cut-off value and copy number variation for 1q21 in newly diagnosed multiple myeloma: Is it underestimated? Clin Lymphoma Myeloma Leuk. 22:535–544. 2022. View Article : Google Scholar : PubMed/NCBI

90 

Liu X, Jia S, Chu Y, Tian B, Gao Y, Zhang C, Zheng Y, Jia W, Liu X, Yuan R, et al: Chromosome 1q21 gain is an adverse prognostic factor for newly diagnosed multiple myeloma patients treated with bortezomib-based regimens. Front Oncol. 12:9385502022. View Article : Google Scholar : PubMed/NCBI

91 

Schavgoulidze A, Perrot A, Cazaubiel T, Leleu X, Montes L, Jacquet C, Belhadj K, Brechignac S, Frenzel L, Chalopin T, et al: Prognostic impact of translocation t(14;16) in multiple myeloma according to the presence of additional genetic lesions. Blood Cancer J. 13:1602023. View Article : Google Scholar : PubMed/NCBI

92 

Pasvolsky O, Ghanem S, Milton DR, Rauf M, Tanner MR, Bashir Q, Srour S, Saini N, Lin P, Ramdial J, et al: Outcomes of patients with multiple myeloma and 1q gain/amplification receiving autologous hematopoietic stem cell transplant: The MD Anderson cancer center experience. Blood Cancer J. 14:42024. View Article : Google Scholar : PubMed/NCBI

93 

Cowan AJ, Green DJ, Kwok M, Lee S, Coffey DG, Holmberg LA, Tuazon S, Gopal AK and Libby EN: Diagnosis and management of multiple myeloma: A review. JAMA. 327:464–477. 2022. View Article : Google Scholar : PubMed/NCBI

94 

Rajkumar SV, Dimopoulos MA, Palumbo A, Blade J, Merlini G, Mateos MV, Kumar S, Hillengass J, Kastritis E, Richardson P, et al: International myeloma working group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol. 15:e538–e548. 2014. View Article : Google Scholar : PubMed/NCBI

95 

Kumar SK and Rajkumar SV: The multiple myelomas-current concepts in cytogenetic classification and therapy. Nat Rev Clin Oncol. 15:409–421. 2018. View Article : Google Scholar : PubMed/NCBI

96 

Bazarbachi AH, Al Hamed R, Malard F, Bazarbachi A, Harousseau JL and Mohty M: Induction therapy prior to autologous stem cell transplantation (ASCT) in newly diagnosed multiple myeloma: An update. Blood Cancer J. 12:472022. View Article : Google Scholar : PubMed/NCBI

97 

Al Hamed R, Bazarbachi AH, Malard F, Harousseau JL and Mohty M: Current status of autologous stem cell transplantation for multiple myeloma. Blood Cancer J. 9:442019. View Article : Google Scholar : PubMed/NCBI

98 

Varma A, Sui D, Milton DR, Tang G, Saini N, Hasan O, Mukherjee A, Joseph JJ, Bashir Q, Rondon G, et al: Outcome of multiple myeloma with chromosome 1q gain and 1p deletion after autologous hematopoietic stem cell transplantation: Propensity score matched analysis. Biol Blood Marrow Transplant. 26:665–671. 2020. View Article : Google Scholar : PubMed/NCBI

99 

Kumar SK, Buadi FK and Rajkumar SV: Pros and cons of frontline autologous transplant in multiple myeloma: The debate over timing. Blood. 133:652–659. 2019. View Article : Google Scholar : PubMed/NCBI

100 

Ntanasis-Stathopoulos I, Gavriatopoulou M, Kastritis E, Terpos E and Dimopoulos MA: Multiple myeloma: Role of autologous transplantation. Cancer Treat Rev. 82:1019292020. View Article : Google Scholar : PubMed/NCBI

101 

Dimopoulos MA, Moreau P, Terpos E, Mateos MV, Zweegman S, Cook G, Delforge M, Hájek R, Schjesvold F, Cavo M, et al: Multiple myeloma: EHA-ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 32:309–322. 2021. View Article : Google Scholar : PubMed/NCBI

102 

Brown PA, Shah B, Advani A, Aoun P, Boyer MW, Burke PW, DeAngelo DJ, Dinner S, Fathi AT, Gauthier J, et al: Acute lymphoblastic leukemia, version 2.2021, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 19:1079–1109. 2021. View Article : Google Scholar : PubMed/NCBI

103 

Ito T, Ando H, Suzuki T, Ogura T, Hotta K, Imamura Y, Yamaguchi Y and Handa H: Identification of a primary target of thalidomide teratogenicity. Science. 327:1345–1350. 2010. View Article : Google Scholar : PubMed/NCBI

104 

Lu G, Middleton RE, Sun H, Naniong M, Ott CJ, Mitsiades CS, Wong KK, Bradner JE and Kaelin WG Jr: The myeloma drug lenalidomide promotes the cereblon-dependent destruction of Ikaros proteins. Science. 343:305–309. 2014. View Article : Google Scholar : PubMed/NCBI

105 

Krönke J, Udeshi ND, Narla A, Grauman P, Hurst SN, McConkey M, Svinkina T, Heckl D, Comer E, Li X, et al: Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science. 343:301–305. 2014. View Article : Google Scholar : PubMed/NCBI

106 

Wang S and Jin FY: Advances on immunomodulatory drugs against multiple myeloma. Zhonghua Xue Ye Xue Za Zhi. 37:262–264. 2016.(In Chinese). PubMed/NCBI

107 

Abe Y and Ishida T: Immunomodulatory drugs in the treatment of multiple myeloma. Jpn J Clin Oncol. 49:695–702. 2019. View Article : Google Scholar : PubMed/NCBI

108 

Parman T, Wiley MJ and Wells PG: Free radical-mediated oxidative DNA damage in the mechanism of thalidomide teratogenicity. Nat Med. 5:582–585. 1999. View Article : Google Scholar : PubMed/NCBI

109 

Moreau P: How I treat myeloma with new agents. Blood. 130:1507–1513. 2017. View Article : Google Scholar : PubMed/NCBI

110 

Quach H, Ritchie D, Stewart AK, Neeson P, Harrison S, Smyth MJ and Prince HM: Mechanism of action of immunomodulatory drugs (IMiDS) in multiple myeloma. Leukemia. 24:22–32. 2010. View Article : Google Scholar : PubMed/NCBI

111 

Cao Y, Zhu H, He R, Kong L, Shao J, Zhuang R, Xi J and Zhang J: Proteasome, a promising therapeutic target for multiple diseases beyond cancer. Drug Des Devel Ther. 14:4327–4342. 2020. View Article : Google Scholar : PubMed/NCBI

112 

Thibaudeau TA and Smith DM: A practical review of proteasome pharmacology. Pharmacol Rev. 71:170–197. 2019. View Article : Google Scholar : PubMed/NCBI

113 

Nunes AT and Annunziata CM: Proteasome inhibitors: Structure and function. Semin Oncol. 44:377–380. 2017. View Article : Google Scholar : PubMed/NCBI

114 

Ito S: Proteasome inhibitors for the treatment of multiple myeloma. Cancers (Basel). 12:2652020. View Article : Google Scholar : PubMed/NCBI

115 

Moreau P, Richardson PG, Cavo M, Orlowski RZ, San Miguel JF, Palumbo A and Harousseau JL: Proteasome inhibitors in multiple myeloma: 10 Years later. Blood. 120:947–959. 2012. View Article : Google Scholar : PubMed/NCBI

116 

Herndon TM, Deisseroth A, Kaminskas E, Kane RC, Koti KM, Rothmann MD, Habtemariam B, Bullock J, Bray JD, Hawes J, et al: U.S. food and drug administration approval: Carfilzomib for the treatment of multiple myeloma. Clin Cancer Res. 19:4559–4563. 2013. View Article : Google Scholar : PubMed/NCBI

117 

Chen Q, Han X, Zheng G, Yang Y, Li Y, Zhang E, Yang L, Dong M, He D, He J and Cai Z: The adverse impact of a gain in chromosome 1q on the prognosis of multiple myeloma treated with bortezomib-based regimens: A retrospective single-center study in China. Front Oncol. 12:10846832022. View Article : Google Scholar : PubMed/NCBI

118 

Kapoor P, Schmidt TM, Jacobus S, Wei Z, Fonseca R, Callander NS, Lonial S, Rajkumar SV and Kumar S: OAB-052: Impact of chromosome 1 abnormalities on newly diagnosed multiple myeloma treated with proteasome inhibitor, immunomodulatory drug, and dexamethasone: Analysis from the ENDURANCE ECOG-ACRIN E1A11 trial. Clin Lymphoma Myeloma Leuk. 21 (Suppl 2):S33–S34. 2021. View Article : Google Scholar

119 

Tang HKK, Fung CY, Morgan GJ, Kumar S, Siu L, Ip HWA, Yip SF, Lau KNH, Lau CK, Lee H, et al: The impact of bortezomib-based induction in newly diagnosed multiple myeloma with chromosome 1q21 gain. Ther Adv Hematol. 13:204062072210820432022. View Article : Google Scholar : PubMed/NCBI

120 

Panopoulou A, Cairns DA, Holroyd A, Nichols I, Cray N, Pawlyn C, Cook G, Drayson M, Boyd K, Davies FE, et al: Optimizing the value of lenalidomide maintenance by extended genetic profiling: An analysis of 556 patients in the Myeloma XI trial. Blood. 141:1666–1674. 2023. View Article : Google Scholar : PubMed/NCBI

121 

D'Agostino M, Ruggeri M, Aquino S, Giuliani N, Arigoni M, Gentile M, Olivero M, Vincelli ID, Capra A, Mussatto C, et al: Impact of gain and amplification of 1q in newly diagnosed multiple myeloma patients receiving carfilzomib-based treatment in the forte trial. Blood. 136 (Suppl 1):S38–S40. 2020. View Article : Google Scholar

122 

Misiewicz-Krzeminska I, de Ramón C, Corchete LA, Krzeminski P, Rojas EA, Isidro I, García-Sanz R, Martínez-López J, Oriol A, Bladé J, et al: Quantitative expression of Ikaros, IRF4, and PSMD10 proteins predicts survival in VRD-treated patients with multiple myeloma. Blood Adv. 4:6023–6033. 2020. View Article : Google Scholar : PubMed/NCBI

123 

Moreau P, Masszi T, Grzasko N, Bahlis NJ, Hansson M, Pour L, Sandhu I, Ganly P, Baker BW, Jackson SR, et al: Oral ixazomib, lenalidomide, and dexamethasone for multiple myeloma. N Engl J Med. 374:1621–1634. 2016. View Article : Google Scholar : PubMed/NCBI

124 

Avet-Loiseau H, Bahlis NJ, Chng WJ, Masszi T, Viterbo L, Pour L, Ganly P, Palumbo A, Cavo M, Langer C, et al: Ixazomib significantly prolongs progression-free survival in high-risk relapsed/refractory myeloma patients. Blood. 130:2610–2618. 2017. View Article : Google Scholar : PubMed/NCBI

125 

Gay F, Musto P, Rota-Scalabrini D, Bertamini L, Belotti A, Galli M, Offidani M, Zamagni E, Ledda A, Grasso M, et al: Carfilzomib with cyclophosphamide and dexamethasone or lenalidomide and dexamethasone plus autologous transplantation or carfilzomib plus lenalidomide and dexamethasone, followed by maintenance with carfilzomib plus lenalidomide or lenalidomide alone for patients with newly diagnosed multiple myeloma (FORTE): A randomised, open-label, phase 2 trial. Lancet Oncol. 22:1705–1720. 2021. View Article : Google Scholar : PubMed/NCBI

126 

Wu HT and Zhao XY: Regulation of CD38 on multiple myeloma and NK cells by monoclonal antibodies. Int J Biol Sci. 18:1974–1988. 2022. View Article : Google Scholar : PubMed/NCBI

127 

van de Donk NWCJ, Richardson PG and Malavasi F: CD38 antibodies in multiple myeloma: Back to the future. Blood. 131:13–29. 2018. View Article : Google Scholar : PubMed/NCBI

128 

Krejcik J, Casneuf T, Nijhof IS, Verbist B, Bald J, Plesner T, Syed K, Liu K, van de Donk NW, Weiss BM, et al: Daratumumab depletes CD38+ immune regulatory cells, promotes T-cell expansion, and skews T-cell repertoire in multiple myeloma. Blood. 128:384–394. 2016. View Article : Google Scholar : PubMed/NCBI

129 

van de Donk NWCJ and Usmani SZ: CD38 antibodies in multiple myeloma: Mechanisms of action and modes of resistance. Front Immunol. 9:21342018. View Article : Google Scholar : PubMed/NCBI

130 

Gozzetti A, Ciofini S, Simoncelli M, Santoni A, Pacelli P, Raspadori D and Bocchia M: Anti CD38 monoclonal antibodies for multiple myeloma treatment. Hum Vaccin Immunother. 18:20526582022. View Article : Google Scholar : PubMed/NCBI

131 

Jiang H, Acharya C, An G, Zhong M, Feng X, Wang L, Dasilva N, Song Z, Yang G, Adrian F, et al: SAR650984 directly induces multiple myeloma cell death via lysosomal-associated and apoptotic pathways, which is further enhanced by pomalidomide. Leukemia. 30:399–408. 2016. View Article : Google Scholar : PubMed/NCBI

132 

Chari A, Kaufman JL, Laubach JP, Sborov DW, Reeves B, Rodriguez C, Silbermann R, Costa LJ, Anderson LD Jr, Nathwani N, et al: Daratumumab plus lenalidomide, bortezomib, and dexamethasone (D-RVd) in transplant-eligible newly diagnosed multiple myeloma (NDMM) patients (Pts): Final analysis of griffin among clinically relevant subgroups. Blood. 140 (Suppl 1):S7278–S7281. 2022. View Article : Google Scholar

133 

Mohan M, Weinhold N, Schinke C, Thanedrarajan S, Rasche L, Sawyer JR, Tian E, van Rhee F and Zangari M: Daratumumab in high-risk relapsed/refractory multiple myeloma patients: Adverse effect of chromosome 1q21 gain/amplification and GEP70 status on outcome. Br J Haematol. 189:67–71. 2020. View Article : Google Scholar : PubMed/NCBI

134 

Parrondo RD, Gardner LB, Alhaj Moustafa M, Roy V, Sher T, Rasheed A, Warsame RM, Larsen JT, Gonsalves EI, Kourelis T, et al: Therapeutic outcomes of relapsed-refractory multiple myeloma patients with 1q21+treated with daratumumab-based regimens: A retrospective analysis. Blood. 140 (Suppl 1):S7237–S7238. 2022. View Article : Google Scholar

135 

Weisel K, Besemer B, Haenel M, Lutz R, Mann C, Munder M, Goerner M, Reinhardt HC, Nogai A, Ko YD, et al: Isatuximab, carfilzomib, lenalidomide, and dexamethasone (Isa-KRd) in patients with high-risk newly diagnosed multiple myeloma: Planned interim analysis of the GMMG-concept trial. Blood. 140 (Suppl 1):S1836–S1838. 2022. View Article : Google Scholar

136 

Harrison SJ, Perrot A, Alegre A, Simpson D, Wang MC, Spencer A, Delimpasi S, Hulin C, Sunami K, Facon T, et al: Subgroup analysis of ICARIA-MM study in relapsed/refractory multiple myeloma patients with high-risk cytogenetics. Br J Haematol. 194:120–131. 2021. View Article : Google Scholar : PubMed/NCBI

137 

Spicka I, Moreau P, Martin TG, Facon T, Martinez G, Oriol A, Koh Y, Lim A, Mikala G, Rosiñol L, et al: Isatuximab plus carfilzomib and dexamethasone in relapsed multiple myeloma patients with high-risk cytogenetics: IKEMA subgroup analysis. Eur J Haematol. 109:504–512. 2022. View Article : Google Scholar : PubMed/NCBI

138 

Martin T, Richardson PG, Facon T, Moreau P, Perrot A, Spicka I, Bisht K, Inchauspé M, Casca F, Macé S, et al: Primary outcomes by 1q21+ status for isatuximab-treated patients with relapsed/refractory multiple myeloma: Subgroup analyses from ICARIA-MM and IKEMA. Haematologica. 107:2485–2491. 2022. View Article : Google Scholar : PubMed/NCBI

139 

Hsi ED, Steinle R, Balasa B, Szmania S, Draksharapu A, Shum BP, Huseni M, Powers D, Nanisetti A, Zhang Y, et al: CS1, a potential new therapeutic antibody target for the treatment of multiple myeloma. Clin Cancer Res. 14:2775–2784. 2008. View Article : Google Scholar : PubMed/NCBI

140 

Ishibashi M, Morita R and Tamura H: Immune functions of signaling lymphocytic activation molecule family molecules in multiple myeloma. Cancers (Basel). 13:2792021. View Article : Google Scholar : PubMed/NCBI

141 

Pazina T, James AM, MacFarlane AW IV, Bezman NA, Henning KA, Bee C, Graziano RF, Robbins MD, Cohen AD and Campbell KS: The anti-SLAMF7 antibody elotuzumab mediates NK cell activation through both CD16-dependent and -independent mechanisms. Oncoimmunology. 6:e13398532017. View Article : Google Scholar : PubMed/NCBI

142 

Usmani SZ, Hoering A, Ailawadhi S, Sexton R, Lipe B, Hita SF, Valent J, Rosenzweig M, Zonder JA, Dhodapkar M, et al: Bortezomib, lenalidomide, and dexamethasone with or without elotuzumab in patients with untreated, high-risk multiple myeloma (SWOG-1211): Primary analysis of a randomised, phase 2 trial. Lancet Haematol. 8:e45–e54. 2021. View Article : Google Scholar : PubMed/NCBI

143 

Derman BA, Kansagra A, Zonder J, Stefka AT, Grinblatt DL, Anderson LD Jr, Gurbuxani S, Narula S, Rayani S, Major A, et al: Elotuzumab and weekly carfilzomib, lenalidomide, and dexamethasone in patients with newly diagnosed multiple myeloma without transplant intent: A phase 2 measurable residual disease-adapted study. JAMA Oncol. 8:1278–1286. 2022. View Article : Google Scholar : PubMed/NCBI

144 

Turner JG, Kashyap T, Dawson JL, Gomez J, Bauer AA, Grant S, Dai Y, Shain KH, Meads M, Landesman Y and Sullivan DM: XPO1 inhibitor combination therapy with bortezomib or carfilzomib induces nuclear localization of IkappaBalpha and overcomes acquired proteasome inhibitor resistance in human multiple myeloma. Oncotarget. 7:78896–78909. 2016. View Article : Google Scholar : PubMed/NCBI

145 

Gandhi UH, Cornell RF, Lakshman A, Gahvari ZJ, McGehee E, Jagosky MH, Gupta R, Varnado W, Fiala MA, Chhabra S, et al: Outcomes of patients with multiple myeloma refractory to CD38-targeted monoclonal antibody therapy. Leukemia. 33:2266–2275. 2019. View Article : Google Scholar : PubMed/NCBI

146 

Tai YT, Landesman Y, Acharya C, Calle Y, Zhong MY, Cea M, Tannenbaum D, Cagnetta A, Reagan M, Munshi AA, et al: CRM1 inhibition induces tumor cell cytotoxicity and impairs osteoclastogenesis in multiple myeloma: Molecular mechanisms and therapeutic implications. Leukemia. 28:155–165. 2014. View Article : Google Scholar : PubMed/NCBI

147 

Argueta C, Kashyap T, Klebanov B, Unger TJ, Guo C, Harrington S, Baloglu E, Lee M, Senapedis W, Shacham S and Landesman Y: Selinexor synergizes with dexamethasone to repress mTORC1 signaling and induce multiple myeloma cell death. Oncotarget. 9:25529–25544. 2018. View Article : Google Scholar : PubMed/NCBI

148 

Qiu L, Xia Z, Fu C, Chen W, Chang C, Fang B, An G, Wei Y, Cai Z, Gao S, et al: Selinexor plus low-dose dexamethasone in Chinese patients with relapsed/refractory multiple myeloma previously treated with an immunomodulatory agent and a proteasome inhibitor (MARCH): a phase II, single-arm study. BMC Med. 20:1082022. View Article : Google Scholar : PubMed/NCBI

149 

Gasparetto C, Schiller GJ, Tuchman SA, Callander NS, Baljevic M, Lentzsch S, Rossi AC, Kotb R, White D, Bahlis NJ, et al: Once weekly selinexor, carfilzomib and dexamethasone in carfilzomib non-refractory multiple myeloma patients. Br J Cancer. 126:718–725. 2022. View Article : Google Scholar : PubMed/NCBI

150 

Chng WJ, Dispenzieri A, Chim CS, Fonseca R, Goldschmidt H, Lentzsch S, Munshi N, Palumbo A, Miguel JS, Sonneveld P, et al: IMWG consensus on risk stratification in multiple myeloma. Leukemia. 28:269–277. 2014. View Article : Google Scholar : PubMed/NCBI

151 

Palumbo A, Avet-Loiseau H, Oliva S, Lokhorst HM, Goldschmidt H, Rosinol L, Richardson P, Caltagirone S, Lahuerta JJ, Facon T, et al: Revised international staging system for multiple myeloma: A report from international myeloma working group. J Clin Oncol. 33:2863–2869. 2015. View Article : Google Scholar : PubMed/NCBI

152 

Sonneveld P, Avet-Loiseau H, Lonial S, Usmani S, Siegel D, Anderson KC, Chng WJ, Moreau P, Attal M, Kyle RA, et al: Treatment of multiple myeloma with high-risk cytogenetics: A consensus of the International myeloma working group. Blood. 127:2955–2962. 2016. View Article : Google Scholar : PubMed/NCBI

153 

Rajkumar SV: Multiple myeloma: 2020 Update on diagnosis, risk-stratification and management. Am J Hematol. 95:548–567. 2020. View Article : Google Scholar : PubMed/NCBI

154 

Hanamura I: Gain/amplification of chromosome arm 1q21 in multiple myeloma. Cancers (Basel). 13:2562021. View Article : Google Scholar : PubMed/NCBI

155 

Goldman-Mazur S, Vesole DH and Jurczyszyn A: Clinical implications of cytogenetic and molecular aberrations in multiple myeloma. Acta Haematol Pol. 52:18–28. 2021. View Article : Google Scholar

Related Articles

Journal Cover

June-2024
Volume 27 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu N, Xie Z, Li H and Wang L: The numerous facets of 1q21<sup>+</sup> in multiple myeloma: Pathogenesis, clinicopathological features, prognosis and clinical progress (Review). Oncol Lett 27: 258, 2024
APA
Liu, N., Xie, Z., Li, H., & Wang, L. (2024). The numerous facets of 1q21<sup>+</sup> in multiple myeloma: Pathogenesis, clinicopathological features, prognosis and clinical progress (Review). Oncology Letters, 27, 258. https://doi.org/10.3892/ol.2024.14391
MLA
Liu, N., Xie, Z., Li, H., Wang, L."The numerous facets of 1q21<sup>+</sup> in multiple myeloma: Pathogenesis, clinicopathological features, prognosis and clinical progress (Review)". Oncology Letters 27.6 (2024): 258.
Chicago
Liu, N., Xie, Z., Li, H., Wang, L."The numerous facets of 1q21<sup>+</sup> in multiple myeloma: Pathogenesis, clinicopathological features, prognosis and clinical progress (Review)". Oncology Letters 27, no. 6 (2024): 258. https://doi.org/10.3892/ol.2024.14391