CCR 24 0545 PDF
CCR 24 0545 PDF
4 Jian Cui1,2#, Xiaoyun Li1,2#, Shuhui Deng1,2,3#, Chenxing Du1,2, Huishou Fan4, Wenqiang
5 Yan1,2, Jingyu Xu1,2, Xiaoqing Li5, Tengteng Yu1,2, Shuaishuai Zhang1,2, Rui Lv1,2, Weiwei
6 Sui1,2, Mu Hao1,2, Xin Du5, Yan Xu1,2, Shuhua Yi1,2, Dehui Zou1,2, Tao Cheng1,2, Lugui
21 #These authors contributed equally to this work: Jian Cui, Xiaoyun Li and Shuhui Deng.
22
23 Running title:
24 scRNA-seq of therapy-induced clonal evolution in MM
25
63 Word count:
64 Text word count: 7220
65 Abstract word count: 247
66 Figure count: 6
3
69 Translational relevance
70 Therapy-induced clonal evolution is one of the most important hindrances toward a cure for
71 multiple myeloma. Through single-cell sequencing of paired diagnostic and post-treatment
72 bone marrow samples, we examined how therapy-induced clonal evolution and resistance
73 pathways allow minimal residual clones to survive induction treatment. We demonstrated
74 three primary trajectories of transcriptional evolution after treatment: clonal elimination in
4
83 Abstract
84 Purpose: In multiple myeloma (MM), therapy-induced clonal evolution is associated with
85 treatment resistance and is one of the most important hindrances toward a cure for MM. To
86 further understand the molecular mechanisms controlling the clonal evolution of MM, we
87 applied single-cell RNA-sequencing (scRNA-seq) to paired diagnostic and post-treatment
88 bone marrow (BM) samples.
109 Keywords:
110 Single-cell RNA sequencing, multiple myeloma, clonal evolution, transcriptional evolution,
111 tumor microenvironment
112
5
113 Introduction
114 The advent of novel therapeutics, such as proteasome inhibitors (PIs) and immunomodulatory
115 drugs (IMiDs), has significantly enhanced survival outcomes for patients with multiple
116 myeloma (MM) over the past two decades (1). Presently, the treatment objective for MM is
117 transitioning from achieving complete response (CR), traditionally assessed using
118 conventional biochemical tests, to realizing undetectable minimal residual disease (MRD-)
125 However, recent studies exploring MRD dynamics have highlighted that only a subset of
126 patients (those who have undergone autologous stem-cell transplant (5) or received
127 continuous maintenance treatment (6)) experience a transition from detectable MRD (MRD+)
128 to MRD-. This underscores the urgency of characterizing the biological properties of
129 persistent MRD tumor cells. Such efforts can unveil new insights into the chemoresistance
130 mechanisms of these cells and potentially disclose hitherto unknown vulnerabilities that can
131 be exploited to eradicate persistent MRD tumor cells (4, 7).
132
133 A high degree of intraclonal heterogeneity is a key feature of MM. Patients with newly
134 diagnosed multiple myeloma (NDMM) invariably harbor multiple clones with distinct genetic
135 backgrounds (8). Moreover, treatments exert varying impacts on intraclonal diversity, leading
136 to the elimination of sensitive clones and the selection of resistant ones (7, 8). While our
137 current understanding of the genetic and non-genetic biology of MRD tumor cells has been
138 predominantly gleaned from bulk-level analysis (9-11). The advent of single-cell technologies,
139 especially multi-omics single-cell technologies, has offered us a novel perspective on
140 understanding the clonal evolution of MM (12). A recent single-cell study has revealed that
141 high-risk major clones observed at relapse are already present at diagnosis as minor,
142 undetectable subclones (13). Moreover, a subgroup of plasma cells (PCs) exhibiting 1q
6
143 alterations, TP53 mutations, and elevated expression of the PHF19 has been identified to be
144 associated with the progression of MM (14). By integrating single-cell RNA sequencing
145 (scRNA-seq) with various sequencing platforms, co-evolution of tumor cells and the tumor
146 microenvironment (TME) has also been explored during progression of MM (15).
147
148 However, two main limitations exist in previous MM clonal evolution studies. Firstly, the
163 To fill these knowledge gaps, our study examined the transcriptional dynamics of PCs from
164 diagnosis through to post-treatment. We collected and sequenced a total of 38 paired bone
165 marrow (BM) samples. Our findings revealed three primary trajectories of transcriptional
166 evolution post-treatment: clonal elimination in MRD- patients, as well as clonal stabilization
167 and clonal selection in MRD+ patients. Additionally, we observed disparate resistance
168 pathways in residual malignant PCs (mPCs). A comparison of the genetic and transcriptional
169 dynamics unveiled a strong correlation between genetic and non-genetic factors driving the
170 clonal evolution of MM. Lastly, we identified variations in cellular interactions between
171 mPCs and the TME, with selective PCs exhibiting the strongest interactions with the TME.
172
7
173 Taken together, our study offered a deeper understanding of the clonal evolution and
174 resistance mechanisms of MM. By examining these processes at a single-cell resolution on a
175 large scale, we confirmed the link between genetic and non-genetic mechanisms driving
176 clonal evolution, providing insights into potential therapeutic strategies aimed at disrupting
177 the connections between MRD tumor cells and the TME.
178
189 Bone marrow mononuclear cells (BMMCs) were isolated from the BM aspirates using the
190 density gradient centrifugation method with Ficoll-Paque PLUS lymphocyte isolation sterile
191 solution (Cytiva, Sweden). For patients MM1-MM8, CD138+ plasma cells (PCs) were
192 isolated using magnetic-activated cell sorting with CD138+ magnetic beads (Miltenyi Biotec,
193 Paris, France). As indicated by several previous studies and ours, a ≥90% plasma cell purity
194 could be achieved after cell sorting (20-22). All samples were viably cryopreserved in
195 dimethyl sulfoxide at a final concentration of 10%. This study was approved by the local
196 institutional ethics committees under the oversight of the Institute of Hematology & Blood
197 Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College.
198 In accordance with the guidelines of the Declaration of Helsinki, all patients and the HD
199 provided informed consent prior to enrollment.
200
212 Single-cell RNA library construction and sequencing using the 10x Genomics platform.
213 Frozen BM cells were rapidly thawed, washed, counted, and resuspended in PBS and 0.04%
214 bovine serum albumin to a final concentration of 1,000 cells per μl. In this study, samples
215 were sequenced in two batches. Samples MM1 to MM8 constituted the first batch, while
216 samples MM9 to MM19 comprised the second batch. The Single Cell 3' Library Kit V3 (10x
217 Genomics, CA, USA) was used for the cDNA synthesis of individual cells. These cells were
218 then barcoded and ready for library construction following the manufacturer's instructions.
219 Upon preparation, the indexed cDNA libraries underwent sequencing on an Illumina NovaSeq
220 6000 platform using a paired-end mode. Novogene Co., Ltd. (Beijing, China) performed the
221 sequencing according to the manufacturer's guidelines, targeting an approximate sequencing
222 depth of 20,000 reads per cell.
223
252 Cell types were determined by evaluating the average expression levels of established
253 markers. These included SDC1, IGHG1, and MZB1 for PCs; CD79A, CD79B, and MS4A1
254 for B cells; S100A8, S100A9, and LYZ for myeloid cells; GNLY, NKG7, CD3D, and CD3E
255 for NK cells and/or T cells; CD34 for hematopoietic stem and progenitor cells (HSPCs); and
256 HBA1, HBA2, HBB, and PF4 for erythrocytes and/or megakaryocytes, as previously
257 described (28-32).
258
259 Following this, PCs were isolated based on the expression of known marker genes such as
260 SDC1, MZB1, and XBP1. A reclustering process was then implemented. The methods
261 described above were used to cluster PCs into 22 subsets from both HD and patients.
262 Malignant PCs from each patient, both pre- and post-treatment, were integrated for
10
263 subsequent analysis.
264
273 For the differential analysis depicted in Fig. 3, we used a pct threshold of 0.6 to identify
274 DEGs across patients. For other differential analyses, a min.pct threshold of 0.3 was used.
275 These DEGs were then uploaded to the Metascape (33) website (https://metascape.org), and
276 default parameters were used to perform pathway enrichment analyses on the 50 hallmark
277 pathways (34).
278
285 To identify somatic large-scale CNVs, the gene count matrix was used as the input with the
286 following parameters: cluster_by_groups = False, cutoff = 0.1, analysis_mode = “subclusters”,
287 BayesMaxPNormal = 0.25. This allowed us to identify regions of the genome where there
288 were significant increases or decreases in gene copy numbers, which can often be indicative
289 of cancer-associated genetic alterations.
290
388 Results
397 We analyzed 161,403 CD138+ PCs and bone marrow mononuclear cells (BMMCs) derived
398 from HD, MM pre-, and post-treatment samples, all of which passed stringent quality controls
399 (Fig. S1A, S1B; Table S2). Additionally, we integrated our dataset with the scRNA-seq
400 dataset from eight HDs from the Human Cell Atlas (HCA) (40). After normalizing the gene
401 expression matrix, we identified six major cell subsets in the BM using graph-based clustering
402 (Fig. S1C, S1D). We observed a distinct separation of plasma/myeloma cells from immune
403 cells, with notable enrichment of CD138+ cells in the MM TME (Fig. 1B-1D). In total, we
404 profiled 76,036 PCs (median = 902) and analyzed a number of immune cells per sample
405 ranging from 839 to 10,137, with a median of 3,059 cells (Table S3).
406
407 Single-cell transcriptional profiles reveal patient-specific patterns and common driver
408 events.
409 To delve deeper into the inter- and intra-patient tumor heterogeneity in MM, we carried out
410 unsupervised clustering for samples with a sufficient number of PCs (≥ 30 PCs). This process
411 yielded 22 clusters of PCs. Interestingly, most PCs derived from HDs were clustered into
412 Cluster 1 (C1), a group that also contained PCs from various patients. Consequently, PCs in
15
413 C1 were identified as HD/normal-like PCs (nlPCs) (Fig. 1E). We observed significant
414 transcriptional differences among patients, with the majority of PC clusters being composed
415 exclusively of cells from a single patient. However, there were some exceptions: Cluster 15
416 (C15), for instance, contained PCs from two patients with distinct translocations (MM12 with
417 t(6;14), and MM13 with t(4;14)) (Fig. 1F; Table S1). Furthermore, longitudinal samples from
418 the same patients were grouped into separate clusters (MM5: C2 and C11; MM6: C5 and C19)
451 To further understand the transcriptional differences between nlPCs and mPCs, we
452 subsequently performed unsupervised clustering on the nlPCs and mPCs from MM2_pre. Our
453 results unveiled more transcriptional clusters than copy number variation (CNV) clones.
454 Moreover, nlPCs and mPCs were distinctly separated, while the transcriptome-based
455 clustering displayed a high degree of consistency with CNV clones and cell groups (Fig. 2A).
456 Further analysis revealed unique CNA profiles among three CNV subclones, with S1 and S3
457 demonstrating similar CNA profiles, and S2 exhibiting a seemingly normal CNA profile (Fig.
458 2B). In addition, cellular differentiation states inferred using CytoTRACE indicated a less
459 differentiated state in mPCs and a more differentiated state in nlPCs (Fig. 2C). Further
460 analysis, which included HD PCs, indicated an intermediate differentiated state of nlPCs (Fig.
461 S2E). DEG analysis in nlPCs and mPCs from MM2_pre compared to HD PCs revealed 55
462 shared upregulated DEGs. These genes included some previously described MM-specific
463 targets, such as SLAMF7 (43) and MCL1 (44) (Fig. 2D; Table S5). Pathway enrichment
464 analysis for the shared upregulated DEGs demonstrated significant enrichment for the NF-κB
465 pathway (Fig. 2E). On the other hand, compared to nlPCs, mPCs showed upregulated
466 expression of endoplasmic reticulum (ER) and unfolded protein response (UPR) pathway
467 genes PPIB, mitochondrial stress genes COX7C, COX6B1, and a previously reported
468 MM-related resistance gene MIF (45) (Fig. 2E; Table S5). Furthermore, activation of the
469 MYC signaling pathway was observed in mPCs (Fig. 2F). Selected MM-related driver genes,
470 including ITGB7, LAMP5, KRAS, and BRAF, were expressed at the highest levels in mPCs,
471 while nlPCs only exhibited low-level expression of these genes (Fig. 2G). Analysis of all
472 nlPCs grouped in the C1 cluster demonstrated similar seemingly normal CNA profiles (Fig.
17
473 S2F), while the expression of MM-related genes was exclusively detected in patient-derived
474 nlPCs (Fig. 2H). Further analysis of nlPCs from MM9_pre, MM3_post and MGUS also
475 demonstrated similar seemingly normal CNA profiles, whereas mPCs from MM3_post
476 displayed various loci involved by CNV (Fig. S3). Therefore, nlPCs might represent a
477 transitional state of PCs, evolving from normal PCs to mPCs.
478
487 Several distinguishing features were observed in this cluster: it lacked secondary genetic
488 events as shown in the CNA profile, exhibited monoclonal immunoglobulin expression or
489 light chain restriction, and despite clustering with HD PCs at the single-cell level, the nlPCs
490 demonstrated significant transcriptional alterations. Our results suggested that nlPCs
491 represented a transitional state of PCs, transitioning from normal PCs to mPCs. Given the
492 multi-step disease progression characteristic of MM, such nlPCs might represent a potential
493 target for early intervention in MM and warrant further investigation.
494
507 However, the percentage of mPCs within the total PC population does not fully reflect the
508 transcriptional changes that occur in mPCs. Thus, we compared the dynamic shifts in mPC
516 Thus, we categorized mPCs into three groups based on their response to treatment: sensitive
517 PCs, such as those in clusters C0, C1, C3, and C5 in MM5, which were nearly eliminated
518 following treatments; resistant PCs, which did not respond or only partially responded to
519 treatment, such as those in clusters C0, C1, C2, and C4 in MM19; and selective PCs, which
520 showed a growth advantage and became the predominant cluster post-treatment, including C2
521 in MM5, C1 in MM6, and C1 in MM14 (Fig. S6B, S6C).
522
523 Although most of the resistant clusters remained in a state of cell-cycle arrest post-treatment,
524 we identified a cell-cycle-related resistant cluster in five out of seven patients who received
525 bortezomib and lenalidomide in combination with dexamethasone treatment (Fig. 3B). To
526 delve deeper into these cycling-resistant clusters, we carried out a DEG analysis between HD,
527 sensitive, and cycling-resistant PCs. Interestingly, our results demonstrated that
528 cycling-resistant PCs highly expressed several genes previously linked to myeloma drug
529 resistance, such as CKS1B, STMN1, TUBA1B, and TYMS (47-50). We also noted high
530 expression of PHF19, a gene recently implicated in the malignant progression of MM and PC
531 leukemia (14).
532
19
533 The pathways enriched in cycling-resistant cells were associated with UPR, hypoxia, and
534 cell-cycle/proliferation pathways. Notably, in line with recent reports that indicate an
535 upregulation of antioxidant gene programs in cycling persister cells (51), we found that
536 cycling-resistant cells also highly expressed MIF, a key antioxidant gene known to mediate
537 MM cell resistance to PIs (45) (Fig. 3C, 3D; Table S6). However, our analysis of
538 noncycling-resistant PCs highlighted only a few upregulated genes, such as S100A4,
541 Compared to both HD PCs and sensitive PCs, the genes displaying upregulation in selective
542 PCs were found to be significantly enriched in pathways such as NF-κB, as demonstrated by
543 genes like FOSB, JUN, and EGR1. Furthermore, they were also enriched in the anti-apoptotic
544 pathway, exemplified by genes such as MCL1. The gene PPP1R15A, crucial for controlling
545 cell viability amid protein folding stress (52), was also observed to be upregulated in selective
546 PCs. Notably, this gene has been previously associated with the progression from MGUS to
547 MM (53). Furthermore, CD74, which is the primary surface receptor for MIF, was
548 significantly upregulated in selective PCs (Fig. 3E, 3F; Table S8). As previous studies have
549 suggested that stimulation of surface CD74 contributes to NF-κB pathway activation (54), we
550 thus hypothesized that CD74 played an important role in the interaction between selective
551 cells and the TME.
552
553 Next, we investigated transcription factors (TFs) that could regulate different drug resistance
554 pathways in cycling-resistant and selective cells. Our analysis indicated that EZH2, E2F1, and
555 E2F2, all of which are cell-cycle/proliferation-regulating TFs, were upregulated in
556 cycling-resistant PCs (Fig. 3G). Conversely, the genes highly expressed in selective PCs were
557 regulated by TFs such as FOS and JUN. It has been reported that the TF AP-1, formed by
558 FOS and JUN, plays a crucial role in enhancing survival but decreasing the proliferation of
559 MM cells (15) (Fig. 3G).
560
561 Lastly, we compared the DEGs between selective PCs and cycling-resistant PCs (Table S9).
562 We found that selective PCs were enriched for pathways including NF-κB, apoptosis and P53
20
563 pathways, while cycling-resistant PCs were enriched for pathways including MYC, E2F target,
564 and UPR pathways (Fig. 3H).
565
581 The adverse prognostic value of the cycling-resistant signature was further validated by
582 utilizing three independent datasets (GSE2658, GSE9782, and GSE57317) consisting of 868
583 MM cases (Fig. 4C). And our results indicated a slightly longer OS for patients with a high
584 selective score compared to those without a high selective score, though the difference did not
585 reach statistical significance in the Log-rank analysis (Fig. 4D).
586
587 Longitudinal single-cell analysis of MM patients pre- and post-treatment reveals genetic
588 evolution and different patterns of metabolic adaptation.
589 To gain deeper insights into the interplay between genetic and non-genetic evolution, we
590 applied inferCNV (55) to delineate the clonal architecture of genetic events in patients. A
591 representative patient, MM5, exhibited branching evolution (16), with analysis of pre- and
592 post-treatment mPCs identifying six CNV subclones among the eight transcriptional clusters
21
593 (Fig. 5A). Consistent with iFISH results (Table S1), we observed a common 13q deletion
594 (13q-) in all six CNV subclones (Fig. 5B).
595
596 To investigate the clonal evolution pattern in MM5, we considered the clonal structure of each
597 CNV subclone. Early genetic events were found in all or most of the CNV subclones, while
598 later events occurred in only some of the subgroups. Additionally, ancestral malignant
607 Further analysis revealed a high degree of concordance between transcriptional clusters and
608 genetic subclones, such as C2 (transcriptional cluster) and S4 (genetic subclone), or between
609 C4 (transcriptional cluster) and S6 (genetic subclone) (Fig. 5D). This observation suggested a
610 strong link between genetic and non-genetic mechanisms driving the clonal evolution of MM.
611 Similarly, the transcriptional selection observed in MM5 post-treatment could also be
612 interpreted from a genetic perspective. Notably, two groups of tiny subclones at diagnosis,
613 namely S4 and S6, became the dominant subclones post-treatment, whereas S1, S2, and S3
614 were virtually eliminated, and S5 was only partly preserved (Fig. 5E).
615
616 Viewed through the genetic lens of clonal evolution, S4 (corresponding to C2) emerged as the
617 predominant major CNV subclone post-treatment, followed by S6 (corresponding to C4).
618 Interestingly, MM5 exhibited a significantly shorter survival compared to other patients with
619 MRD+ status, with an OS of only 10 months. We hypothesized that both the non-genetic
620 resistance mechanisms of C2 and C4 contributed to the inferior outcome in MM5. To
621 investigate this further, we inferred the cellular differentiation states using CytoTRACE,
622 revealing a more differentiated state in C2 and a less differentiated state in C4. However, both
22
623 C2 and C4 displayed higher differentiation compared to other transcriptional clusters (Fig.
624 5F).
625
626 Further DEG analysis between C2 and C4 clusters highlighted significant differences in gene
627 expression patterns. C2 showed high expression of genes related to the "Hypoxia" pathway,
628 such as PPP1R15A and ATF3, while C4 exhibited elevated expression of genes involved in
636 Similar genetic selection was observed in MM6 and MM16 (Fig. 5I; Fig. S7), while MM10,
637 MM16, MM17, and MM19 exhibited genetic stabilization (Fig. 5I; Fig. S8). In summary, our
638 findings demonstrated a close link between the genetic landscape and non-genetic
639 mechanisms driving myeloma cell treatment resistance. Moreover, the distinct patterns of
640 metabolic adaptation played crucial roles in driving the clonal selection of residual malignant
641 cells.
642
654 To understand the functional implications of these changes, we conducted a DEG analysis on
655 CD14+ monocytes post-treatment. The results revealed significantly elevated expression of
656 chemokines, such as CCL3, CCL4, and CXCL2, by CD14+ monocytes (Fig. 6D; Table S11).
657 This heightened expression of chemokines suggested an enhanced recruitment effect of
658 CD14+ monocytes on T cells. Conversely, genes highly expressed by CD16+ monocytes
663 Taken together, our results indicated that the treatment not only remodeled the composition of
664 the MM TME but also altered the transcriptional status of immune cells, reflecting a complex
665 interplay between the immune system and myeloma progression.
666
681 Moreover, we found enhanced interactions between selective mPCs and NK cells, mediated
682 by HLA-E/CD94:NKG2A interactions, which represent inhibitory signals to NK cells (Fig.
24
683 S9B). Additionally, HLA-F/LILRB1 interactions, representing an inhibitory signaling axis in
684 the TME (57), was also enriched between selective mPCs and myeloid cells (Fig. 6I). These
685 findings raised intriguing questions about the potential involvement of these differential
686 cellular interactions in the observed patterns of transcriptional evolution and distinct
687 resistance pathways among the residual mPCs. Further studies are warranted to elucidate the
688 functional implications of these interactions in the immune escape and survival of selective
691 Discussion
692 This scRNA-seq study of paired diagnostic and post-treatment samples represented the most
693 extensive single-cell dataset to date. It elucidated transcriptional dynamics and uncovered
694 resistance pathways within MRD tumor cells in patients with MM. There were five main
695 findings in this study. Firstly, a population of nlPCs was identified in MM. Secondly, three
696 main trajectories of transcriptional evolution post-treatment were observed. These were clonal
697 elimination in MRD- patients, as well as clonal stabilization and clonal selection in MRD+
698 patients. Thirdly, a close link between genetic and non-genetic mechanisms behind the clonal
699 evolution of MM was confirmed, which further supported the multidimensional background
700 of clonal evolution. Fourthly, transcriptional and metabolic adaptations were observed as
701 important activated prosurvival pathways in MRD tumor cells. Finally, differential cellular
702 interactions between mPCs and TME cells were observed, showing that the TME might
703 provide shelter for MRD tumor cells.
704
705 Genetic studies employing iFISH and whole-exome sequencing have demonstrated that MM
706 is not a single disease entity but also a spectrum of molecular conditions, each with distinct
707 clinical outcomes (58). Moreover, gene expression profiling has revealed various subgroups
708 within these conditions, defined by the cytogenetic features of PCs and the aberrant
709 expression of a D-group cyclin (41). Previous phenotypic studies have identified several
710 subclones within patients with MM, each associated with unique chemoresistant profiles,
711 cytogenetic characteristics, and clonogenic potentials (7). Recently, an MGUS-like phenotype
712 has been described in MM patients based on the prevalence of BM PC and clonal PC within
25
713 the BM PC compartment (59). These findings underscore the remarkable inter-tumor
714 heterogeneity of MM and suggest the existence of a cell population evolving from normal to
715 malignant PCs identifiable at the early stages of MM pathogenesis. In alignment with
716 previous scRNA-seq studies (29, 32, 46), our study identified a PC cluster composed of PCs
717 from HDs and multiple MM patients. Interestingly, even clonal PCs from MM patients
718 co-clustered with HD PCs. We designated such clonal PCs as nlPCs. Further analyses
723 Therapy-induced clonal evolution plays a crucial role in MM drug resistance (58). As
724 treatments exert selective pressure, subclones either carry or acquire specific genetic
725 alterations, conferring a competitive fitness that enables these PCs to evade therapeutic
726 pressure. Utilizing quantitative multigene fluorescence in situ hybridization, we have charted
727 the phylogeny of tumor subclones and identified four patterns of evolutionary architecture in
728 129 longitudinal samples from 57 MM patients. We found that patients exhibiting clonal
729 stabilization had superior OS, while those with linear evolution had the worst outcomes (16).
730 Increasingly, studies are focusing on clonal evolution in MRD tumor cells (6, 7). Previous
731 research indicates that standard-risk patients are more susceptible to clonal selection events,
732 whereas high-risk patients more often acquire mutations (4). A recent single-cell study has
733 revealed that genetic abnormalities observed at relapse are already present at diagnosis within
734 subclonal populations (13). Based on these findings, we investigated the transcriptional
735 dynamics between disease diagnosis and post-treatment. Our results revealed three main
736 trajectories of transcriptional evolution post-treatment: clonal elimination in MRD- patients,
737 as well as clonal stabilization and clonal selection in MRD+ patients. Furthermore, by
738 delineating the clonal architecture of genetic events per patient using inferCNV (55), we
739 found a high correlation between genetic and non-genetic mechanisms underpinning the
740 clonal evolution of MM. We proposed further investigations to elucidate the clinical
741 significance of nlPCs.
742
26
743 There is growing evidence suggesting that drug resistance in cancer cannot be solely
744 attributed to simple genetic causes (19). The concept that non-genetic mechanisms contribute
745 to therapeutic resistance is increasingly gaining recognition (19). In the context of MM,
746 subclones with identical genetic backgrounds can result in multiple transcriptional clusters
747 (29), which points to the non-genetic mechanisms that influence transcriptional status. A
748 recent scRNA-seq study has reported upregulation of the UPR, ER, and mitochondrial stress
755 MM progression involves aberrant interactions between PCs and the TME (60).
756 Comprehensive evidence suggests that both hematopoietic and non-hematopoietic cells,
757 including regulatory T cells (61), regulatory B cells (62), myeloid-derived suppressor cells (63,
758 64), and stromal cells (56), contribute to PC proliferation. Our study identified a potential
759 supportive role of the TME for MRD tumor cells. The most pronounced interaction between
760 MRD tumor cells and the TME was observed in selective PCs. Furthermore, the upregulated
761 expression of CD74 by selective PCs and the potential contribution of CD74-related
762 PC-myeloid cell interactions to their immune evasion were noted. Given that myeloid cells or
763 monocyte-derived cells, including osteoclasts (65) and DCs (66), comprise the primary
764 cellular component of the microenvironment that promotes immunosuppression, disrupting
765 the interactions between selective PCs and myeloid cells by blocking CD74 could be a
766 potential therapeutic approach to eradicate MRD tumor cells.
767
768 This study also highlights several remaining challenges. First, since only one MGUS sample
769 was sequenced in our study, the interpretation of the results of our study needs to take into
770 account the limited number of MGUS patients in our cohort. Nevertheless, the identification
771 of nlPCs added substantial weight to the concept of inter-tumor heterogeneity in MM.
772 Moreover, the inclusion of steroids in the induction regimen may also lead to transcriptional
27
773 and metabolic adaptation in residual mPCs. Further experimental studies are necessary to
774 confirm whether the transcriptional and metabolic adaptation in mPCs is in response to a
775 specific drug or represent a general phenomenon.
776
777 In conclusion, single-cell profiling of MRD tumor cells could provide novel insights into the
778 transcriptional evolution and molecular mechanisms underlying therapeutic resistance. This
782 Acknowledgments:
783 We thank Prof. Nikhil C. Munshi for his insightful suggestions to this study. We thank all the
784 patients and families for participation in this research study. Fig. 1A, and the Graphic abstract
785 were created with the assistance of BioRender. This study was supported by the National
786 Natural Science Foundation (grants 82270218, U22A20291 to G. An), Youth Fund of the
787 National Natural Science Foundation of China (grants 81900214, to S. Deng), the National
788 Natural Science Foundation (grants 32370722 to X. Gao), the CAMS Innovation Fund for
789 Medical Sciences (CIFMS) (grants 2022-I2M-1-022, to L. Qiu; grants 2021-I2M-C&T-B-079,
790 to G. An, and grants 2021-I2M-1-041), the International Cooperation Projects of National
791 Natural Science Foundation (grants 81920108006, to L. Qiu). J. Cui is supported by grant
792 from International Myeloma Society.
793
28
794 References
795
796 1. Rodriguez-Otero P, Paiva B, San-Miguel JF. Roadmap to cure multiple myeloma. Cancer Treat Rev. 2021. 100:
797 102284.
798 2. Anderson KC, Auclair D, Adam SJ, Agarwal A, Anderson M, Avet-Loiseau H, et al. Minimal Residual Disease in
799 Myeloma: Application for Clinical Care and New Drug Registration. Clin Cancer Res. 2021. 27(19): 5195-5212.
800 3. Quach H. MRD end point in myeloma: ready for prime time. Blood. 2022. 139(6): 799-802.
801 4. Goicoechea I, Puig N, Cedena M, Burgos L, Cordón L, Vidriales M, et al. Deep MRD profiling defines outcome and
29
837 resistance mechanism to CAR T cell therapy in a patient with multiple myeloma. Nat Commun. 2021. 12(1): 868.
838 19. Marine J, Dawson S, Dawson MA. Non-genetic mechanisms of therapeutic resistance in cancer. Nat Rev Cancer.
839 2020. 20(12): 743-756.
840 20. Corre J, Perrot A, Caillot D, Belhadj K, Hulin C, Leleu X, et al. del(17p) without TP53 mutation confers a poor
841 prognosis in intensively treated newly diagnosed patients with multiple myeloma. Blood. 2021. 137(9): 1192-1195.
842 21. Shah V, Sherborne AL, Walker BA, Johnson DC, Boyle EM, Ellis S, et al. Prediction of outcome in newly diagnosed
843 myeloma: a meta-analysis of the molecular profiles of 1905 trial patients. Leukemia. 2018. 32(1): 102-110.
844 22. An G, Li Z, Tai YT, Acharya C, Li Q, Qin X, et al. The impact of clone size on the prognostic value of chromosome
845 aberrations by fluorescence in situ hybridization in multiple myeloma. Clin Cancer Res. 2015. 21(9): 2148-2156.
30
881 Bioinformatics. 2013. 14: 7.
882 39. Efremova M, Vento-Tormo M, Teichmann SA, Vento-Tormo R. CellPhoneDB: inferring cell–cell communication
883 from combined expression of multi-subunit ligand–receptor complexes. Nat Protoc. 2020. 15(4): 1484-1506.
884 40. Rozenblatt-Rosen O, Stubbington M, Regev A, Teichmann SA. The Human Cell Atlas: from vision to reality. Nature.
885 2017. 550(7677): 451-453.
886 41. Zhan F, Huang Y, Colla S, Stewart JP, Hanamura I, Gupta S, et al. The molecular classification of multiple myeloma.
887 Blood. 2006. 108(6): 2020-2028.
888 42. Merz M, Merz A, Wang J, Wei L, Hu Q, Hutson N, et al. Deciphering spatial genomic heterogeneity at a single cell
889 resolution in multiple myeloma. Nat Commun. 2022. 13(1): 807.
31
925 57. Barkal AA, Weiskopf K, Kao KS, Gordon SR, Rosental B, Yiu YY, et al. Engagement of MHC class I by the
926 inhibitory receptor LILRB1 suppresses macrophages and is a target of cancer immunotherapy. Nat Immunol. 2018.
927 19(1): 76-84.
928 58. Pawlyn C, Morgan GJ. Evolutionary biology of high-risk multiple myeloma. Nat Rev Cancer. 2017. 17(9): 543-556.
929 59. Burgos L, Tamariz-Amador L, Puig N, Cedena M, Guerrero C, Jelínek T, et al. Definition and Clinical Significance of
930 the Monoclonal Gammopathy of Undetermined Significance–Like Phenotype in Patients With Monoclonal
931 Gammopathies. J Clin Oncol. 2023: JCO.22.01916.
932 60. Palumbo A, Anderson K. Multiple Myeloma. N Eng J Med. 2011. 364(11): 1046-1060.
933 61. Feng X, Zhang L, Acharya C, An G, Wen K, Qiu L, et al. Targeting CD38 Suppresses Induction and Function of T
945
32
946 Author contributions:
947 J. Cui: Conceptualization, data curation, formal analysis, investigation, validation,
948 visualization, writing-original draft, writing-review & editing. X. Li: Conceptualization, data
949 curation, formal analysis, investigation, software, visualization, writing-original draft,
950 writing-review & editing. Shuhui Deng: Conceptualization, funding acquisition,
951 investigation, project administration, writing-original draft, writing-review & editing. C. Du:
33
970 Figure legends
971 Figure 1. Single-cell transcriptional landscape of HD and MM patients.
972 (A) The workflow illustrates the strategy for cell collection from matching diagnosis-treated
973 paired samples for scRNA-seq. (B) Six major single-cell clusters from BM samples of HD,
974 MGUS, NDMM (pre-treatment), and MM patients during induction therapy (post-treatment).
975 Annotation of cell clusters is marked in the bottom part with color codes. Cell embeddings are
37
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/doi/10.1158/1078-0432.CCR-24-0545/3464142/ccr-24-0545.pdf by guest on 26 June 2024
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/doi/10.1158/1078-0432.CCR-24-0545/3464142/ccr-24-0545.pdf by guest on 26 June 2024
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/doi/10.1158/1078-0432.CCR-24-0545/3464142/ccr-24-0545.pdf by guest on 26 June 2024
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/doi/10.1158/1078-0432.CCR-24-0545/3464142/ccr-24-0545.pdf by guest on 26 June 2024
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/doi/10.1158/1078-0432.CCR-24-0545/3464142/ccr-24-0545.pdf by guest on 26 June 2024
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/doi/10.1158/1078-0432.CCR-24-0545/3464142/ccr-24-0545.pdf by guest on 26 June 2024