Our cohort consisted of 12/38 patients with high-grade OS (31.6%) with initial bone metastasis (group A) and 26/38 (68.4%) with initial pulmonary metastasis (group B), of whom 15/38 (39.5%) had paired samples of primary lesions and metastatic lesions (Table 1 and Supplementary Table 1 and Fig. 1a-c). For our discovery set, WES was at a median depth of ∽591× (range, 268∽1239) using paired constitutional DNA from peripheral blood (Supplementary Table 2). Five of 15 (33.3%) paired tumor samples were derived from pretherapeutic biopsies. In other words, these samples presented the initial characteristics of their tumor status. We first assessed genomic complexity within this cohort along with clinical progression in these cases. These data were then partly integrated with transcriptome and immunostaining results to characterize the genomic landscape of our OS cohort.
Table 1. Clinical demographics of the 38 metastatic osteosarcomas.
Characteristic
|
Group A
|
Group B
|
Statistical method
|
p value
|
n (%)
|
12 (31.6)
|
26 (68.4)
|
|
|
Age at diagnosis (in years)
|
|
|
T test
|
0.060
|
Range
|
6-61
|
6-57
|
|
|
Median (Q1, Q3)
|
19.5 (13.25, 38.75)
|
14 (11, 17.5)
|
|
|
Gender, n (%)
|
|
|
Chi-square
|
1.000
|
Male
|
9 (23.7)
|
20 (52.6)
|
|
|
Female
|
3 (7.9)
|
6 (15.8)
|
|
|
Paired primary osteosarcoma, n (%)
|
|
|
Chi-square
|
0.033
|
Yes
|
8 (21.1)
|
7 (18.4)
|
|
|
No
|
4 (10.5)
|
19 (50.0)
|
|
|
Primary sample site, n (%)
|
|
|
Chi-square
|
0.280
|
Femur
|
8 (21.1)
|
16 (42.1)
|
|
|
Humerus
|
0 (0.0)
|
3 (7.9)
|
|
|
Tibia
|
2 (5.3)
|
6 (15.8)
|
|
|
Pelvis
|
1 (2.6)
|
1 (2.6)
|
|
|
Spine
|
1 (2.6)
|
0 (0.0)
|
|
|
Metastatic sample site, n (%)
|
|
|
Chi-square
|
0.000*
|
Femur
|
2 (5.3)
|
3 (7.9)
|
|
|
Fibula
|
2 (5.3)
|
1 (2.6)
|
|
|
Tibia
|
0 (0.0)
|
2 (5.3)
|
|
|
Pelvis
|
4 (10.5)
|
0 (0.0)
|
|
|
Rib
|
1 (2.6)
|
0 (0.0)
|
|
|
Sacrum
|
1 (2.6)
|
0 (0.0)
|
|
|
Spine
|
2 (5.3)
|
1 (2.6)
|
|
|
Lung
|
0 (0.0)
|
19 (50.0)
|
|
|
Genomic type, n (%)
|
|
|
Chi-square
|
0.0180a
|
SNV
|
10 (26.3)
|
11 (28.9)
|
|
|
SV or mixed
|
2 (5.3)
|
15 (39.5)
|
|
|
TMB
|
|
|
Wilcoxon
|
0.010*
|
Range
|
1.1-32.9
|
0-14.2
|
|
|
Median (Q1, Q3)
|
4.85 (2.75, 11.98)
|
2.4 (1.38, 4.45)
|
|
|
MSI status, n (%)
|
|
|
Chi-square
|
0.316
|
MSS
|
11 (29.0)
|
26 (68.4)
|
|
|
MSI-H
|
1 (2.6)
|
0 (0.0)
|
|
|
Neoantigen
|
|
|
Wilcoxon
|
0.002*
|
Range
|
57-2084
|
0-1539
|
|
|
Median (Q1, Q3)
|
743 (316.5, 1034.5)
|
128.5 (49, 200.5)
|
|
|
HED status, n (%)
|
|
|
Chi-square
|
1.000
|
Low
|
10 (26.3)
|
22 (57.9)
|
|
|
High
|
2 (5.3)
|
4 (10.5)
|
|
|
Adjuvant therapy, n (%)
|
|
|
|
|
First-line treatment
|
11 (29.0)
|
25 (65.8)
|
Chi-square
|
0.538
|
Second-line treatment
|
9 (23.7)
|
22 (57.9)
|
Chi-square
|
0.656
|
Third-line treatment
|
5 (13.2)
|
10 (26.3)
|
Chi-square
|
1.000
|
Fourth-line treatment
|
2 (5.3)
|
5 (13.2)
|
Chi-square
|
1.000
|
Vital status, n (%)
|
|
|
Chi-square
|
0.108
|
Alive
|
7 (18.4)
|
22 (57.9)
|
|
|
Dead
|
5 (13.2)
|
4 (10.5)
|
|
|
PFS (mo)
|
|
|
T test
|
0.596
|
Range
|
0-48.23
|
2.23-34.43
|
|
|
Median (Q1, Q3)
|
12.4 (7.45, 21.22)
|
13.95 (8.00, 19.75)
|
|
|
OS or follow-up time (mo)
|
|
|
T test
|
0.856
|
Range
|
7-59
|
9-54
|
|
|
Median (Q1, Q3)
|
23.5 (15.25, 46.5)
|
27 (16.75, 38.25)
|
|
|
Abbreviations: SNV, single nucleotide variation; Amp, amplification; Fus, fusion; TMB, tumor mutation burden; MSI, microsatellite instability; MSS, microsatellite stability; MSI-H, microsatellite instability-high; PFS, progression free survival; Mo, month; OS, overall survival.
HED, human leukocyte antigen-I evolutionary divergence. HED score> 7.887, defined as HED high; HED score ≤ 7.887, defined as HED low.
[Insert Table 1 here]
Clinicopathologic characteristics and genomic landscape
The clinical demographics of the 38 high-grade OS patients are summarized in Table 1. The clinical, pathologic, and predominant genomic landscape characteristics of all OSs with DNA sequencing are shown in Fig. 1a. The median age at diagnosis was 19.5 years for group A (Q1, Q3, 13.3, 38.8) and 14.0 years for group B (Q1, Q3, 11.0, 17.5). Males comprised the majority of the studied population, with 75.0% and 77.0% in both groups, respectively. For most of our patients, the primary lesions were located in the femur: 66.7% in group A and 61.5% in group B. For resected and metastatic samples, 4/12 (33.3%) were located in the pelvis, 2/12 (16.7%) in the femur, fibula and spine separately and 1/12 (8.3%) in the rib and sacrum in group A. When those patients developed pulmonary metastasis, it was usually multiple and scattered, such that the possibility for metastasectomy was low. In group B, although 19/26 (73.1%) were pulmonary lesions, 3/26 (11.5%), 2/26 (7.7%), 1/26 (3.8%) were later-developed femoral, tibial, fibular and spinal lesions respectively. Regarding the last-collected samples, 91.6% in group A showed progression upon first-line chemotherapy and 75.0% upon second-line systemic therapy; for group B, 96.2% progressed on first-line therapy and 84.6% on second-line therapy (detailed information in Table 1 and Supplementary Table 1). Therefore, subsequent WES data analyses were performed on 58 OS samples from 38 patients. With a median follow-up time of 26.0 months (Q1, Q3, 16.8, 39.0), 7/12 (58.3%) in group A were alive with disease while 22/26 (84.6%) in group B were alive. We did not find any obvious significant difference in progression-free survival (p=0.33) or overall survival (p=0.06) (Supplementary Fig. 1).
We defined driver mutations in established cancer genes, which mainly include SNVs and SVs, to describe the genomic landscape for the whole population. It is interesting that our cohorts seemed to show diverse biological behaviors based on the main genomic manifestations. Our population contained more point mutations other than historical high-risk OS genetic sequencings published [17, 18, 26, 27]. Consequently, we adopted a novel strategy to re-subclassify our tumors, restricting our focus to high-confidence variants (see Methods). A large portion of the group A were of the SNV type. Group B showed similar manifestation as reported for high-risk OS, involving genomic amplifications as well as fusions or mixed type (which we called the SV type), with few recurrent point mutations (Fig. 1d).
Somatic alterations detected by our methods in the 38 metastatic patients are shown in Fig. 1a and listed in Supplementary Table 2. Among genes commonly mutated, TP53 and MYC were identified in 22/58 and 14/58 patients respectively (57.9% and 36.8%; Fig. 1a; Supplementary Table 2). However, for this cohort of OS patients, we did not specifically design our panel to identify TP53 intron 1 rearrangements, as has recently been reported in OS [17], and more TP53 mutation might be missing due to the WES approach. We also identified alterations in MUC16 (17 mutations in 11 patients, 25.9%), NCOR1 (2 mutations in 9 patients, 23.7%), RAD21 (2 mutations in 9 patients, 23.7%), PTK2 (1 mutations in 8 patients, 21.1%), CDK4 (1 mutations in 7 patients, 18.4%), GLI1(2 mutations in 7 patients, 18.4%) and IGF1R (1 mutations in 7 patients, 18.4%) (Supplementary Table 2). Rb1 loss was only observed in 13.8% (8/58) of samples. Nevertheless, we noticed that for WES, only 14% (4399/31364) of the genomic alterations are reported mutations, whereas 86% (26965/31364) are not reported in the literatures (Fig. 1b); it is unclear whether these alterations influence the occurrence and evolution of OS.
Approximately 89.7% (52/58) of samples had CNAs and structural rearrangements. With respect to the former (Fig. 1a and Supplementary Table 2), amplifications at NCOR1 (n=13/58 samples; 22.4%), RAD21 (n=12/58 samples; 20.7%), CDK4 (n=10/58 samples; 17.2%) and MYC (n=19/58 samples; 32.8%) were the most frequent CNAs. Furthermore, PIK3CG mutation was mutually exclusive of RB1 alteration; overall, this pathway was altered in approximately 67.2% (39/58) of OS samples. Other less common regions of recurrent amplification are provided in Fig. 1a and Supplementary Table 2. We did not observe MDM2 amplification, as expected [26].
The burden of coding indels and substitutions across the 58 tumor samples varied from 2 to 1482 mutations per tumor (median 84; Supplementary Table 2). The highest coding mutation burden (32.9 mut/Mb) was observed in patient WJY, who initially presented with bone metastasis with a time interval from diagnosis to metastasis longer than 24 months and whose tumor exhibited MSI.
Diverse genomic characterization between osteosarcoma initially metastatic to bones and to lungs
We showed the mutation profiles between group A and group B in Fig. 2a. Surprisingly, group A mainly exhibited gene substitutions and short indels (green mark); group B mainly presented with gene amplifications, deletions and rearrangements (red mark). To describe this phenomenon more precisely, we reclassified our population into two genomic types: SNVs, accounting for more than 40% of all genomic alterations, which included gene substitutions and short indels; SVs, accounting for more than 40% of all genomic alterations, which included amplifications, homozygous deletions and breakpoints that disrupt genes or generate gene fusions. For those with both SNVs and SVs of more than 40%, we classified them as the mixed type. In fact, a large portion of our population had the mixed type (17/38, 44.7%) (Fig. 1c). Thus, to highlight patients with distinct genomic manifestations from group A, we further integrated the mixed type into the SV type. We compared all these patients’ genomic changes (see Table 2). Based on chi-square analysis, we observed obverse statistical disequilibrium for the differences between two groups (p=0.018), which suggests that the biological behavior from the perspective of genomic manifestations was distinct for these two groups of population. It should be noted that we further compared the TMB, neoantigen, MSI and HED status between the two groups. However, due to small sample sizes, we only observed diverse differences in TMB (p=0.01) and neoantigen status (p=0.0028) respectively (Fig. 2c and d).
Table 2. The difference of osteosarcoma patients’ genomic alterations between group A and group B.
|
Group A
|
Group B
|
Statistical method
|
p value
|
n (%)
|
12 (31.6)
|
26 (68.4)
|
|
|
Genomic type, n (%)
|
|
|
Chi-square
|
0.0180a
|
SNV
|
10 (26.3)
|
11 (28.9)
|
|
|
SV or mixed
|
2 (5.3)
|
15 (39.5)
|
|
|
TMB
|
|
|
Wilcoxon
|
0.0100a
|
Range
|
1.1-32.9
|
0-14.2
|
|
|
Median (Q1, Q3)
|
4.85 (2.75, 11.98)
|
2.4 (1.38, 4.45)
|
|
|
Neoantigen
|
|
|
Wilcoxon
|
0.0028a
|
Range
|
57-2084
|
0-1539
|
|
|
Median (Q1, Q3)
|
743 (316.5, 1034.5)
|
128.5 (49, 200.5)
|
|
|
MSI, n (%)
|
|
|
Chi-square
|
0.3160
|
MSS
|
11 (29.0)
|
26 (68.4)
|
|
|
MSI-H
|
1 (2.6)
|
0 (0.0)
|
|
|
HED-status, n (%)
|
|
|
Chi-square
|
1.0000
|
Low
|
10 (26.3)
|
22 (57.9)
|
|
|
High
|
2 (5.3)
|
4 (10.5)
|
|
|
Abbreviations: SNV, single nucleotide variation; SV, structural variants; Amp, amplification; Fus, fusion; TMB, tumor mutation burden; MSI, microsatellite instability; MSS, microsatellite stability; MSI-H, microsatellite instability-high.
HED human leukocyte antigen-I evolutionary divergence. HED score> 7.887, defined as HED high; HED score ≤ 7.887, defined as HED low.
The copy number variation region was further analyzed, and we found that CNV amplifications and deletions were more frequent in group B than in group A (Fig. 2b). The most common amplified regions involved 17p11.2, 5p15.2, 9q22.32, 1q21.2, 2q11.1, 8q24.21, 19q12, 9p24.1, 2q11.2, and 15q26.3, including the common driver genes MYC, CCNE1, JAK2, and IGF1R. In addition, 16p13.3, 22q13.33, 17p13.1, 17q25.3, 10q26.3, 19p13.11, 16q24.3, 8p21.3, 9q33.3, and 11p15.5 were the most common regions with copy number loss. The regions with increased copy number only included the driver genes of PTK2, JAK2, and CDK4 in group A.
The mutation burden of OS was similar reported as pancancer analysis [28]. However, the median TMB was 4.85 (Q1, Q3, 2.75, 11.98) and 2.4 (Q1, Q3, 1.38, 4.45) for group A and group B, respectively (Fig. 2c). Tumor neoantigen status can reflect immunogenicity, though the emergence of neoantigens was less than that in melanoma, and the median amount of neoantigen in group A was almost six times more than that in group B, at 743 (316.5, 1034.5) versus 128.5 (49, 200.5) (p=0.0028) (Fig. 2d). We compared all mutations in cell signaling pathway between the two groups and found differences, with most of the genes participating in the DDR pathway (p=0.03), and Notch pathway (p=0.03), with no difference in the cell cycle, TP53, IGF-beta, MYC, HIPPO, RTK or RAS pathways (Fig. 2e).
We analyzed the gene signature profiles of all 38 patients based on the literature for renal cell carcinoma or non-small cell lung cancer. However, mutation spectra revealed significant differences for signature 1 between the groups, mainly referring to age (Supplementary Fig. 2). Thus, we further compared the age distribution between these groups of patients with the Mann–Whitney U test but did not observe any significant difference, with a p value of 0.06 (Supplementary Fig. 3).
For SNVs, 65.8% (25/38) were nonsynonymous mutations, which might partly correlate with the higher expression of neoantigens in group A (Fig. 1b). In four of these SNV-type cases, the paired metastatic lesions were of the SV type. The number of SNVs called with high confidence ranged from 7 to 1482 (median, 150) per exome. Of these, a median of 30 SNV changes (range 4–272) within protein-coding regions potentially result in a functional product. For SV, 44.7% (17/38) were mixed mutations, including amplifications, fusions and long indels; only 1 sample carried a TP53 gene rearrangement, and 14 other rearrangements involved TP53, RB1, NTRK3, LRP1B and other genes in 9 samples. Regarding somatic CNAs, 248 amplification events in 22 samples with a median copy number of 7 (range 3~77) were detected. There were 20 events in 10 samples of gene deletion, including 15 gene deletions and 5 partial deletions in a genomic region of 7236~196391 bp.
High conservation of reported genetic sequencing over time in most osteosarcomas
For the 15 patients with paired primary and metastatic samples, we performed tumor phylogenetics to identify the basis of metastasis. Of the 15 patients, 8/15 (53.3%) were in group A and 7/15 (46.7%) in group B; their genomic manifestations of non-silent mutations are depicted in Fig. 1c. Other than prominent mutation landscape diversity between group A and group B (Fig. 3a), we did not observe any difference between primary and metastatic specimens for genomic landscapes (Fig. 1c), TMB (Supplementary Fig. 6a), neoantigens (Supplementary Fig. 6b) or profiling pathways (Supplementary Fig. 6c). We observed high conservation of reported genetic sequencing over time in both group A and group B. However, it was difficult to precisely define whether these cases involved linear or parallel progression models [29] (Supplementary Figs. 4 and 5). Most of our cases, regardless of group A or group B, were more similar to tumor self-seeding model, in which the primary tumor continually emits cells to a metastasis that was in fact seeded at early stages of tumorigenesis, masking evidence of parallel progression [29]. Phylogenetic trees inferred from genomic data may represent the evolutionary life history of individual tumors, as discussed in the literature [29]. In the present study, the phylogenetic relationship between the regional biopsies of a given sample was inferred from mutation data using LICHeE (Lineage Inference for Cancer Heterogeneity and Evolution) [30], PyClone [24] and CITUP (Conality Inference in Tumors Using Phylogeny) [25]. We employed two methods to describe these phylogenetics to identify the origin of metastasis (Supplementary Figs. 4 and 5), which exhibited multi-sample lineage trees based on the VAF of somatic SNVs and the subclonal evolution model (mutation- and copy number-based) respectively. Overall, 3/8 (37.5%) patients in group A had a more linear evolutionary pattern, whereas 7/7 (100%) patients in group B had a more parallel evolutionary pattern (Supplementary Figs. 4 and 5). We utilized the root/branch ratio (dN/dS ratio) for calculation [31] and found that groups A and B were not statistical different (p=0.46).
Treatment-induced amplifications
In this subset of the population, 3 patients (WS, HXD and WJY) in group A had treatment-naive primary and paired metastatic samples. For these three patients, two of their metastatic samples (WS and HXD) were resected after first-line chemotherapy according to our protocols [32]. The other patient, who was also the only patient (WJY) with a high TMB >30 mut/Mb, did not receive any systemic treatment other than resection during the whole disease process. In addition, the primary samples of two patients in group B (LZZ and SQZP) were treatment naive and derived from open incision biopsy before chemotherapy. However, these two cases both progressed so rapidly during treatment that the metastatic lesions had both progressed on first-line chemotherapy at the time of resection. Based on their genomic landscapes (Fig. 1c and Supplementary Figs. 4 and 5), we noticed that more genomic amplifications appeared after systemic treatment. For WS, bone metastatic lesions newly presented with 4 gene amplifications (ADGRA2, FGFR1, NSD3, POLB); for HXD, samples after treatment newly presented with PTK2 amplification, which did not occur in WJY, who did receive any systemic treatment for she was older than 40 years with relatively indolent disease process. For LZZ and SQZP, genomic amplifications were the major genomic alterations. Samples after systemic treatment showed a tendency toward a decline in SNVs. For the other paired samples, the primary lesions were resected after neoadjuvant chemotherapy; thus, we could not determine the influence of chemotherapy. It is suspected that systemic treatment, especially chemotherapy for OS, might contribute to genomic amplifications, which further might be the epitome of chromothripsis during tumor evolution.
We also compared their phylogenetic trees as well as clonal prevalence before and after chemotherapy (Supplementary Figs. 4 and 5) and did not find any relationship between evolutionary patterns and prognosis.
A distinctive immune ecosystem in osteosarcoma with initially bone metastasis based on multiplex immunohistochemistry
TLSs (tertiary lymphoid structures) are an important biomarker for identifying selected soft tissue sarcomas that might show a response to ICIs. On the basis of our patients’ genomic profiles, we next assessed tumor samples histologically to gain insight into the density and distribution of B cells as well as their relationship to other checkpoint molecules. Surprisingly, according to m-IHC of 3 randomly selected patient samples from group A, one (HXD) showed TLSs at the edges and margins of the tumor (shown in Fig. 3a2); 3 from group B lacked TLSs or CD3+CD8+ T cells (Fig. 3b2). It is also interesting that all patients in the vacant district of PD-L1 (programmed cell death ligand 1) expression usually showed high intensity and strongly positive expression of B7-H3 (Fig. 3a3, b3 and c1, p=0.0043). However, the patient (HXD) with multiple TLS districts of an immunogenic state exhibited poor staining of B7-H3 (Fig. 3a3), the mechanism of which should be further investigated. Notably, architectural analysis showed that CD20+ B cells were localized in TLSs and colocalized with CD4+, CD8+ and CD56+ T cells. Findings between gene expression profilings and immunohistochemistry analysis were complementary and showed modest correlation, which suggested that group A patients seemed to be in a more immunogenic state than group B. We later tried a single anti-PD-1 antibody (camrelizumab, Jiangsu HengRui Medicine Co., 200 mg ivgtt. Q2W) for another patient in our clinical practice in a multiple bone metastatic state but without any pulmonary metastasis after 4 lines of systemic treatment. Surprisingly, after just 4 infusions, a PET/CT scan showed an obvious a partial metabolic response (PMR) by PET Response Criteria in Solid Tumors (PERCIST) [33] (detailed case report in Supplementary Data 6 and Supplementary Fig. 7). Longer follow-up is needed to confirm the duration of the response and overall survival.
Tumor-associated macrophages (TAMs), one of the most abundant immune components in OS, are difficult to characterize due to their heterogeneity. Although multiple approaches have been used, the spatial distribution of TAMs in situ remains unclear. Thus, to examine the TAM distribution within the microenvironment, we analyzed the spatial density of the populations characterized above within different ROIs. The individual TAMs for all patients were then plotted based on the intensity of CD68, CD163, and CD206 (Fig. 4d and e), providing evidence of a spectrum of macrophage populations. The average intensity of each marker was determined on a per-patient basis, confirming that these populations are represented within each individual patient sample and ROI. An increase in overall macrophage density was observed within tumor regions (edge and margin, but not core, detailed in Supplementary Table 3) compared with adjacent normal tissue (Supplementary Fig. 8a, p=0.056).