Although neoadjuvant chemotherapy is a standard component of breast cancer treatment, recent evidence suggests that chemotherapeutic drugs can promote metastasis through poorly defined mechanisms. Here we utilize xenograft mouse models of triple-negative breast cancer to explore the importance of chemotherapy-induced tumor-derived small extracellular vesicles (sEV) in metastasis. Doxorubicin (DXR) enhanced tumor cell sEV secretion to accelerate pulmonary metastasis by priming the premetastatic niche. Proteomic analysis and CRISPR/Cas9 gene editing identified the inflammatory glycoprotein PTX3 enriched in DXR-elicited sEV as a critical regulator of chemotherapy-induced metastasis. Both genetic inhibition of sEV secretion from primary tumors and pharmacologic inhibition of sEV uptake in secondary organs suppressed metastasis following chemotherapy. Taken together, this research uncovers a mechanism of chemotherapy-mediated metastasis by which drug-induced upregulation of sEV secretion and PTX3 protein cargo primes the premetastatic niche and suggests that inhibition of either sEV uptake in secondary organs or secretion from primary tumor cells may be promising therapeutic strategies to suppress metastasis.

Significance:

These findings show that chemotherapy-induced small extracellular vesicles accelerate breast cancer metastasis, and targeted inhibition of tumor-derived vesicles may be a promising therapeutic strategy to improve the efficacy of chemotherapy treatment.

Breast cancer is the most commonly diagnosed malignancy and the leading cause of cancer-associated death in women worldwide, and is responsible for nearly 15% of all new cancer cases and 40,000 deaths per year in the United States (1). Approximately 12% to 17% of newly diagnosed breast cancer cases are classified as triple negative (TNBC) due to the absence of cellular hormone and growth factor receptors (2). Despite the relatively low incidence, TNBC is associated with higher rates of recurrence and lower overall survival than receptor-positive tumors (3). Specifically, patients diagnosed with TNBC have an overall 77% five-year survival rate, compared with 93% for other breast cancer subtypes, and those with metastatic disease have a 22% five-year survival and 80% five-year recurrence rate (4).

Neoadjuvant chemotherapy treatment (NAC), which is typically composed of an anthracycline, alkylator, and taxane regimen, is the standard of care for patients with locally advanced TNBC (4). Despite the ubiquitous use of NAC, only ∼30% of TNBC patients achieve a pathologic complete response (pCR), defined as an absence of residual invasive disease, following treatment (4–6). Patients who do not achieve pCR have significantly decreased three-year overall and disease-free survival rates compared with patients who do achieve pCR (5), and evidence suggests that in a subset of patients, chemotherapy treatment itself may actually promote tumor metastasis (7–12). Therefore, an urgent need exists to understand the molecular mechanisms underlying TNBC metastasis, and how chemotherapy treatment accelerates metastasis, in order to facilitate the development of more effective therapeutic options.

Small extracellular vesicles (sEV) have recently emerged as important regulators of cancer progression and metastasis through the horizontal transfer of cellular material (13). These nanovesicles are secreted by almost all cell types and contain a variety of proteins, RNA, and DNA, which are packaged into intraluminal vesicles within the late endosome and released into the extracellular environment following fusion of the multivesicular body (MVB) with the plasma membrane (14). Uptake of tumor-derived sEVs in secondary tissues has been shown to promote tumor cell invasiveness, facilitate epithelial-to-mesenchymal transition, suppress the host immune system, and prime the premetastatic niche to enhance metastasis (15–20). Despite the well-defined and multifaceted role of sEVs in cancer progression, the effect of chemotherapy treatment on tumor-derived sEV secretion and function is poorly understood. In this study, we establish that the anthracycline doxorubicin (DXR) upregulates breast cancer cell sEV secretion and differentially regulates sEV protein content, and that sEVs derived from DXR-treated cells accelerate metastasis in models of both highly metastatic and poorly metastatic TNBC, demonstrating that sEV secretion is one mechanism by which chemotherapy treatment promotes metastasis. Moreover, we identify the glycoprotein pentraxin-related protein 3 (PTX3) as a novel regulator of sEV-induced breast cancer metastasis, which accelerates tumor progression through priming of the premetastatic niche. We further show that inhibition of both sEV uptake in secondary organs and secretion by primary tumor cells suppresses chemotherapy-induced metastasis, suggesting that targeting tumor-derived sEVs may be an effective therapeutic strategy to suppress metastasis in TNBC patients.

Cell lines

The human breast cancer cell lines MDA-MB-231 (ATCC HTB-26), MDA-MB-468 (ATCC HTB-132), and MCF7 (ATCC HTB-22) were purchased from ATCC. SV40 large T antigen-immortalized mouse embryonic fibroblasts were generated in our lab as previously described (21). Luciferase reporter cells were generated with a pCDH1-SV40-Luc2 construct and selected with hygromycin (200 μg/mL). CRISPR/Cas9-mediated PTX3 knockout (KO) was performed by subcloning three single-guide RNAs (gRNA) targeting PTX3 (5′-AGTGAACTTACGGTCCTCAG-3′, 5′-CCACCACAAACACTATGGAT-3′, 5′-GACGCGAGCCGACCTGCACG-3′) into pLenti-CRISPR-V2 (Addgene 52961). Lentiviral packaging in 293T cells and transduction of pLenti-CRISPR-PTX3 into MDA-MB-231 cells were performed as previously described (22). Puromycin selection (1 μg/mL) was used to isolate single clones, which were screened using immunoblotting for gene disruption, and four validated single clones were pooled. CRISPR/Cas9-mediated RAB27A KO was performed by transducing MDA-MB-231 cells with lentiCas9-Blast (Addgene 52962) and using Blasticidin selection (4 μg/mL) to create a Cas9-stable control cell line. Three gRNAs targeting RAB27A (5′-CCTCTGCCAGTGGCTCCATC-3′, 5′-CTCCAAATTTATCACAACAG-3′, 5′-TGCAGTTATGGGACACAGCA-3′) were subcloned into LRG (Addgene 65656). LRG-Rab27a was delivered by lentiviral transduction to MDA-MB-231 Cas9 cells followed by FACS sorting as previously described (22). Single clones were validated by immunoblotting for gene disruption and two clones pooled. All cell lines in the lab are passaged for less than 6 months before use and periodically authenticated by Mycoplasma testing, morphologic inspection, and STR analysis.

Small extracellular vesicle isolation

To make sEV-depleted FBS, heat-inactivated FBS was centrifuged twice at 120,000 RCF in an SW32Ti rotor (Beckman) for 12 hours at 4°C and filtered through a 0.2 μm filter. For sEV isolation, conditioned media were collected from cells cultured for 24 hours in sEV-depleted media with 400 nmol/L (MDA-MB-231 and MCF7) or 40 nmol/L (MDA-MB-468) DXR, or with a volume of DMSO equivalent to 10% the volume of DXR. Conditioned media were centrifuged at 500 RCF for 10 minutes at 4°C, filtered through a 0.2-μm syringe filter (VWR 28145-501), and concentrated using Pierce 100K MWCO protein concentrators (Thermo Fisher 88533) by centrifuging at 4,000 RCF and 4°C. The concentrated supernatant was centrifuged at 10,000 RCF for 20 minutes at 4°C and then centrifuged twice at 120,000 RCF for 2.5 hours at 4°C and once for 12 hours at 120,000 RCF and 4°C in an SW55Ti rotor (Beckman). The sEV pellet was resuspended in phosphate-buffered saline (PBS) and stored at −20°C. Density gradient ultracentrifugation was performed using a protocol adapted from Kowal and colleagues (23).

Nanoparticle tracking analysis

sEV quantification was performed using a NanoSight NS300 (Malvern Instruments Ltd.). EV samples were diluted to a volume of 1 mL (1:500 to 1:1,000) in particle-free water and five, 60-second videos were generated at 24.98 frames/sec in scatter mode, as previously described (24). Particle size and concentration were calculated using NanoSight software version 3.2 (Malvern Instruments Ltd.). To quantify vesicle secretion per cell, cells were treated with DMSO or DXR in sEV-depleted media for 24 hours and trypsinized using Trypsin-EDTA 0.05% without Ca or Mg (VWR Scientific 45000-660) at the time of conditioned media collection. Cells were counted using a Cellometer Auto 2000 Cell Viability Counter (Nexcelom Bioscience), and total vesicle concentration in each sample was divided by the final cell count.

Transmission electron microscopy

sEV electron microscopy was performed using a protocol adapted from Rikkert and colleagues (25). Briefly, sEVs were diluted in PBS, and 10 μL sample was pipetted onto a 400 mesh copper grid with carbon-coated formvar film and imaged using a JEOL JEM1400 transmission electron microscope (JEOL USA Inc.).

Proteomic analysis

For mass spectrometry (MS) analysis, 50 μg sEVs were resuspended in PBS and quantified using tandem MS at the University of Michigan with a TMT-10plex Isobaric Label Reagent Set (Thermo Fisher 90110) according to the manufacturer's instructions. Samples were analyzed using an Orbitrap Velos mass spectrometer (Thermo Fisher Scientific), and data processing was performed using Proteome Discover and SequestHT. Proteins were filtered against a human database using a false discovery rate of less than 5%. Pathway analysis was performed using Advaita iPathway Guide (Advaita Bioinformatics). The proteomic heat map was generated using the R complex-heat map package.

Orthotopic xenograft mouse model

All animal studies were performed according to guidelines established and approved by the Institutional Animal Care and Use Committee at the Penn State College of Medicine. An orthotopic xenograft breast cancer model was generated by injecting 1.0 × 106 cells into the left fifth inguinal mammary fat pad of 6- to 8-week-old female NOD SCID gamma (NSG, Jackson 005557) or female NOD Rag gamma (NRG, Jackson 007799) mice. Cells were injected in a 50:50 mixture of PBS and Matrigel basement membrane matrix (Fisher CB-40234). For sEV treatment experiments, mice received tail-vein injections of 10 μg purified sEVs in 100 μL PBS, or an equivalent volume of PBS, three times per week, with treatments beginning 1 week after xenograft. For chemotherapy treatment experiments, mice received tail-vein injections of 5 mg/kg DXR (NRG and NSG mice) or 2.5 mg/kg DXR (NSG mice) as indicated, or an equivalent volume of PBS on days 7 and 12 after tumor xenograft. As indicated, chemotherapy-treated mice received daily subcutaneous injections of 0.2 mg/kg heparin in 100 μL PBS, or an equivalent volume of PBS, beginning 1 week after tumor xenograft and continuing for the duration of the experiment. Metastatic tumor growth was tracked by imaging mice on a weekly basis using an IVIS Lumina III in vivo bioluminescent imaging system. Mice were anesthetized using 2.5% isoflurane (IsoSol Isoflurane, USP, VEDCO) and injected with 5 μL/g body weight of 30 mg/mL D-luciferin dissolved in PBS 5 minutes prior to imaging. Photon flux was calculated using region of interest (ROI) measurements around the primary tumor site or the anterior thoracic area for lung and liver metastasis. Primary tumor volume was measured using calipers and calculated as volume = π(length*width2)/6. At the experiment endpoint, mice were humanely euthanized and tumor, lung, and liver tissue were harvested for ex vivo analysis and histology.

Tail-vein injection metastasis mouse model

A metastatic model was established by injecting female 6- to 8-week-old NSG mice via the tail vein with 10 μg purified sEVs in 100 μL PBS, or an equivalent volume of PBS, on days 0, 2, and 4, followed by tail-vein injection of 2.5 × 105 cells on day 5. Metastatic tumor growth was quantified by weekly IVIS imaging. Whole-body ROI measurements were used to quantify metastatic tumor growth. At the experimental endpoint, mice were euthanized, and lung and liver tissue were harvested for ex vivo analysis.

Statistical analysis

GraphPad Prism 7.0 was used for statistical analysis of data. Two-tailed Student t tests were used for single comparisons, and group differences were evaluated using one-way or two-way ANOVA. Statistical significance was set to P < 0.05.

Doxorubicin promotes metastasis and upregulates small extracellular vesicle secretion in TNBC

To confirm that chemotherapy treatment accelerates breast cancer metastasis, we generated orthotopic xenografts of MDA-MB-231 cells expressing a luciferase reporter in the mammary fat pad of female NSG mice, and then treated mice twice over the course of 5 days with a low dose of the topoisomerase II inhibitor DXR or an equivalent volume of PBS, in line with established treatment models (Supplementary Fig. S1A; ref. 26). Bioluminescent imaging of the anterior thoracic region, which masks the luminescent signal produced by the primary tumor in order to more clearly visualize metastatic radiance in the pulmonary and hepatic regions, confirmed that mice treated with DXR demonstrated a significant acceleration in tumor cell metastasis compared with mice treated with PBS (Supplementary Fig. S1B and S1C). Primary tumor volume was significantly decreased in mice treated with DXR compared with PBS (Supplementary Fig. S1D and S1E), confirming that DXR decreases primary tumor growth while promoting metastasis.

We hypothesized that chemotherapy treatment altered sEV secretion from primary tumors, and that sEVs derived from chemotherapy-treated tumor cells promote metastasis. In order to determine the effect of chemotherapy treatment on sEV secretion, we selected concentrations of DXR that inhibit less than 10% of cell viability in vitro in the highly metastatic MDA-MB-231 and the poorly metastatic MDA-MB-468 human TNBC cells lines (Supplementary Fig. S1F). Differential ultracentrifugation was used to isolate sEVs from the conditioned media of cells treated for 24 hours with DXR or DMSO, the vehicle control. Using electron microscopy, immunoblotting, and nanoparticle tracking analysis (NTA), we confirmed that isolated particles displayed the characteristic size, morphology, and surface markers of sEVs (Fig. 1AC). We found that low-dose DXR treatment significantly upregulated both the number of sEVs and the amount of sEV protein secreted per cell from both breast cancer cell lines in vitro (Fig. 1D and E). However, the amount of protein secreted per sEV was not significantly affected by DXR treatment (Fig. 1F). Importantly, we found that the number of circulating sEVs was significantly upregulated in the plasma of tumor-bearing mice treated with DXR compared with mice treated with PBS (Fig. 1G), demonstrating that DXR upregulates sEV secretion both in vitro and in vivo.

Figure 1.

Doxorubicin treatment upregulates sEV secretion. A, Representative electron microscope images of DMSO- and DXR-EVs. Scale bar, 200 nm. B, Immunoblot of DMSO- and DXR-EVs probed for positive and negative sEV markers. C, Representative NTA graphs of DMSO- and DXR-EV size and concentration. D, sEV secretion per cell normalized to the DMSO-EV average for each cell line. MDA-MB-231, n = 6; MDA-MB-468, n = 9. Student t test; mean ± SEM; *, P < 0.05; ***, P < 0.005. E, sEV protein secretion per cell normalized to the DMSO-EV average for each cell line. MDA-MB-231, n = 11; MDA-MB-468, n = 11. Student t test; mean ± SEM; *, P < 0.05; ****, P < 0.001. F, Protein secretion per sEV normalized to the DMSO-EV average for each cell line. MDA-MB-231, n = 3; MDA-MB-468, n = 4. Student t test; mean ± SEM; ns, not significant. G, NTA quantification of sEVs per mL plasma derived from MDA-MB-231, crNT tumor-bearing mice treated twice with PBS or DXR. PBS, n = 8; DXR, n = 8. Student t test; mean ± SEM; **, P < 0.005.

Figure 1.

Doxorubicin treatment upregulates sEV secretion. A, Representative electron microscope images of DMSO- and DXR-EVs. Scale bar, 200 nm. B, Immunoblot of DMSO- and DXR-EVs probed for positive and negative sEV markers. C, Representative NTA graphs of DMSO- and DXR-EV size and concentration. D, sEV secretion per cell normalized to the DMSO-EV average for each cell line. MDA-MB-231, n = 6; MDA-MB-468, n = 9. Student t test; mean ± SEM; *, P < 0.05; ***, P < 0.005. E, sEV protein secretion per cell normalized to the DMSO-EV average for each cell line. MDA-MB-231, n = 11; MDA-MB-468, n = 11. Student t test; mean ± SEM; *, P < 0.05; ****, P < 0.001. F, Protein secretion per sEV normalized to the DMSO-EV average for each cell line. MDA-MB-231, n = 3; MDA-MB-468, n = 4. Student t test; mean ± SEM; ns, not significant. G, NTA quantification of sEVs per mL plasma derived from MDA-MB-231, crNT tumor-bearing mice treated twice with PBS or DXR. PBS, n = 8; DXR, n = 8. Student t test; mean ± SEM; **, P < 0.005.

Close modal

Small extracellular vesicles derived from DXR-treated cells accelerate breast cancer metastasis

To determine the effect of sEVs derived from DXR-treated breast cancer cells on tumor growth and progression, we generated orthotopic xenografts of MDA-MB-231 and MDA-MB-468 cells in NSG mice. One-week after xenograft, we began treating mice three times per week with tail-vein injections of 10 μg purified sEVs derived from DMSO- or DXR-treated breast cancer cells, or with an equivalent volume of PBS (Fig. 2A). Bioluminescent imaging of the anterior thoracic region demonstrated that treatment with DXR-EVs significantly accelerated metastatic tumor growth compared with treatment with DMSO-EVs or PBS in both the highly metastatic and poorly metastatic xenograft models (Fig. 2B and C; Supplementary Fig. S2A and S2B). Histologic scoring of lung metastatic tumor burden using hematoxylin and eosin–stained lung sections demonstrated that DXR-EV treatment significantly enhanced pulmonary metastasis, while quantification of total liver tumor area demonstrated a slight, but not statistically significant, increase in liver metastasis in mice treated with DXR-EVs (Fig. 2DF; Supplementary Fig. S2C–S2E). Notably, sEV treatment did not significantly affect primary tumor growth in either cell line, as demonstrated by bioluminescent imaging (Fig. 2G; Supplementary Fig. S2F and S2G), weekly caliper measurements (Fig. 2H), and quantification of tumor volume ex vivo (Supplementary Fig. S2H–S2K). These results suggest that DXR-EVs accelerate pulmonary metastasis without affecting primary breast tumor growth.

Figure 2.

sEVs derived from DXR-treated cells accelerate breast cancer metastasis. A, Experimental design. B, Representative IVIS images of metastatic signal in mice treated with MDA-MB-231 sEVs or PBS. C, ROI quantification of MDA-MB-231 metastatic tumor radiance. Data represent two independent experiments. PBS, n = 17; 231 DMSO-EV, n = 16; 231 DXR-EV, n = 19. Two-way ANOVA, mean ± SEM; **, P < 0.01; ***, P < 0.005. D, Representative hematoxylin and eosin sections of fixed lung and liver tissue taken from mice sacrificed 8 weeks after initiation of sEV treatment. Liver metastases are outlined. E and F, Average lung (E) and liver (F) metastatic tumor burden from mice treated with MDA-MB-231 sEVs. Data represent one experiment. PBS, n = 6; 231; DMSO-EV, n = 6; 231 DXR-EV, n = 8. One-way ANOVA; mean ± SEM; *, P < 0.05; **, P < 0.01; ns, not significant. G, Representative IVIS images of mice with MDA-MB-231 tumor xenografts. H, MDA-MB-231 tumor volume taken using weekly caliper measurements.

Figure 2.

sEVs derived from DXR-treated cells accelerate breast cancer metastasis. A, Experimental design. B, Representative IVIS images of metastatic signal in mice treated with MDA-MB-231 sEVs or PBS. C, ROI quantification of MDA-MB-231 metastatic tumor radiance. Data represent two independent experiments. PBS, n = 17; 231 DMSO-EV, n = 16; 231 DXR-EV, n = 19. Two-way ANOVA, mean ± SEM; **, P < 0.01; ***, P < 0.005. D, Representative hematoxylin and eosin sections of fixed lung and liver tissue taken from mice sacrificed 8 weeks after initiation of sEV treatment. Liver metastases are outlined. E and F, Average lung (E) and liver (F) metastatic tumor burden from mice treated with MDA-MB-231 sEVs. Data represent one experiment. PBS, n = 6; 231; DMSO-EV, n = 6; 231 DXR-EV, n = 8. One-way ANOVA; mean ± SEM; *, P < 0.05; **, P < 0.01; ns, not significant. G, Representative IVIS images of mice with MDA-MB-231 tumor xenografts. H, MDA-MB-231 tumor volume taken using weekly caliper measurements.

Close modal

DXR-EVs accelerate metastasis by priming the premetastatic niche

We hypothesized that DXR-EVs accelerate breast tumor metastasis either by promoting primary tumor cell migration or invasion or by priming the premetastatic niche in secondary organs. To test the hypothesis that DXR-EVs enhance primary tumor cell movement, we performed migration and invasion assays in vitro using an IncuCyte Live-Cell Analysis System. MDA-MB-231 and MDA-MB-468 cells treated with 10 μg/mL DXR-EVs demonstrated no significant increase in migration or invasion in vitro compared with cells treated with DMSO-EVs or PBS (Supplementary Fig. S3A–S3D), suggesting that uptake of DXR-EVs by primary tumor cells may not significantly enhance metastasis.

To test the hypothesis that DXR-EVs prime the premetastatic niche, we first used whole-organ fluorescent imaging to demonstrate that both MDA-MB-231– and MDA-MB-468–derived Vybrant DiD-labeled DMSO- and DXR-EVs can localize to the lungs and liver in vivo within the first 12 to 24 hours after injection (Supplementary Fig. S4A–S4J). To determine how uptake of sEVs in these organs affects tumor progression, we experimentally modeled metastatic dissemination by injecting female NSG mice three times over the course of 5 days with 10 μg DXR-EV or DMSO-EVs, or an equivalent volume of PBS, followed by tail-vein injection of MDA-MB-231 or MDA-MB-468 cells (Fig. 3A). We found that whole-body metastatic tumor growth was significantly enhanced in mice pretreated with DXR-EVs compared with mice pretreated with DMSO-EVs or PBS in both tumor models (Fig. 3B and C; Supplementary Fig. S5A and S5B). Of note, bone marrow metastatic bioluminescent signaling was increased in mice treated with MDA-MB-468 DXR-EVs, highlighting differences in EV organotropism between the two cell lines. Lung section scoring confirmed that tumor burden was significantly enhanced in mice pretreated with DXR-EVs compared with DMSO-EVs or PBS, while the total liver tumor area was not statistically different between treatment groups, although an increase trend in liver metastasis was observed in mice pretreated with MDA-MB-231 DXR-EVs (Fig. 3DF; Supplementary Fig. S5C and S5D). These results suggest that sEVs derived from DXR-treated breast cancer cells promote pulmonary metastasis by priming the premetastatic niche.

Figure 3.

sEVs derived from DXR-treated cells prime the premetastatic niche. A, Experimental design. B, Representative IVIS images of MDA-MB-231 metastatic luciferase signal in mice from indicated treatment groups. C, ROI quantification of MDA-MB-231 whole-body metastatic tumor radiance. Data represent two independent experiments. PBS, n = 8; 231 DMSO-EV, n = 9; 231 DXR-EV, n = 11. Two-way ANOVA; mean ± SEM; **, P < 0.01; ****, P < 0.001. D, Representative hematoxylin and eosin sections of fixed lung and liver tissue from mice treated with MDA-MB-231 sEVs sacrificed 6 weeks after tumor cell injection. Liver metastases are outlined. E and F, Average lung (E) and liver (F) metastatic tumor burden for mice treated with MDA-MB-231 sEVs. Data represent two independent experiments. PBS, n = 8; 231 DMSO-EV, n = 9; 231 DXR-EV, n = 11. One-way ANOVA; mean ± SEM; *, P < 0.05; **, P < 0.01; ns, not significant.

Figure 3.

sEVs derived from DXR-treated cells prime the premetastatic niche. A, Experimental design. B, Representative IVIS images of MDA-MB-231 metastatic luciferase signal in mice from indicated treatment groups. C, ROI quantification of MDA-MB-231 whole-body metastatic tumor radiance. Data represent two independent experiments. PBS, n = 8; 231 DMSO-EV, n = 9; 231 DXR-EV, n = 11. Two-way ANOVA; mean ± SEM; **, P < 0.01; ****, P < 0.001. D, Representative hematoxylin and eosin sections of fixed lung and liver tissue from mice treated with MDA-MB-231 sEVs sacrificed 6 weeks after tumor cell injection. Liver metastases are outlined. E and F, Average lung (E) and liver (F) metastatic tumor burden for mice treated with MDA-MB-231 sEVs. Data represent two independent experiments. PBS, n = 8; 231 DMSO-EV, n = 9; 231 DXR-EV, n = 11. One-way ANOVA; mean ± SEM; *, P < 0.05; **, P < 0.01; ns, not significant.

Close modal

DXR differentially regulates small extracellular vesicle protein cargo

To establish how uptake of DXR-EVs primes the pulmonary premetastatic niche, we performed unbiased proteomic analysis of MDA-MB-231 DMSO- and DXR-EVs. Of the 1,758 EV proteins found in our screen, we identified 40 significantly upregulated and 167 significantly downregulated proteins in DXR-EVs compared with DMSO-EVs (Fig. 4AC). We were particularly interested in the inflammatory glycoprotein PTX3, which was one of the most highly upregulated proteins in our screen. High PTX3 levels are associated with a significantly decreased probability of relapse-free survival in breast cancer patients who have received NAC (Fig. 4D), making it a promising target. To further confirm that PTX3 is a sEV-associated protein, we performed density-gradient ultracentrifugation and found strong colocalization between PTX3 and the sEV marker CD63 (Supplementary Fig. S6A). We used CRISPR/Cas9 to KO PTX3 (crPTX3) in MDA-MB-231 cells (Fig. 4E) and confirmed that PTX3 KO did not affect tumor cell migration in vitro compared with CRISPR nontargeting (crNT) MDA-MB-231 cells (Supplementary Fig. S6B). We isolated DMSO- and DXR-EVs from crNT and crPTX3 cells and used immunoblotting to confirm that PTX3 is absent in crPTX3 sEVs, and that DXR treatment upregulates PTX3 expression approximately 2-fold in crNT sEVs (Fig. 4F and G). This phenotype is unlikely unique to the MDA-MB-231 cell line, as PTX3 was also upregulated approximately 1.5-fold in DXR-EVs derived from MDA-MB-468 and MCF7 cells (Supplementary Fig. S6C and S6D). We further characterized crNT and crPTX3 sEVs using electron microscopy and NTA to confirm that PTX3 KO did not affect sEV size or morphology (Fig. 4H and I). NTA demonstrated that DXR significantly increased sEV secretion per cell in both crNT and crPTX3 cells compared with DMSO treatment, and no significant differences in sEV secretion were found between crNT and crPTX3 cells (Fig. 4I and J). We used dot plots with or without detergent treatment, which disrupts the vesicle membrane to expose internal proteins, to confirm that PTX3 is upregulated in DXR-EVs and to demonstrate that PTX3 localizes to the sEV surface (Fig. 4K).

Figure 4.

DXR treatment regulates sEV protein content. A, Volcano plot of MDA-MB-231 DMSO-EV and DXR-EV proteins. Red, upregulated in MDA-MB-231 DXR-EVs; blue, downregulated in MDA-MB-231 DXR-EVs. LogFC >0.585 or ≤0.585, −Log10(adj P) >1.303. PTX3 (yellow) is highly enriched in MDA-MB-231 DXR-EVs; logFC = 1.072, P = 1.0e−6. B, 1,758 proteins were identified in MDA-MB-231 cell–derived sEVs. Forty proteins were significantly upregulated, and 167 were significantly downregulated in MDA-MB-231 DXR-EVs. C, Heat map of relative protein abundance in MDA-MB-231 DXR and DMSO-EVs. N = 4 per treatment condition. Top 15 most abundant proteins in DXR and DMSO sEVs are indicated. D, Kaplan–Meier relapse-free survival curve of breast cancer patients who received neoadjuvant chemotherapy treatment, stratified by PTX3 expression. PTX3 low, n = 112; PTX3 high, n = 111. Created using kmplot.com. E, Immunoblot of MDA-MB-231 crNT and crPTX3 cells probed for indicated proteins. F, Immunoblot of sEVs derived from crNT and crPTX3 MDA-MB-231 cells treated with DMSO or DXR. G, Quantification of PTX3 expression in crNT DMSO- and DXR-EVs, normalized to CD63. N = 4; mean ± SEM; Student t test; *, P < 0.05. H, Representative electron microscope images of crNT and crPTX3 sEVs. Scale bar, 200 nm. I, Representative NTA graphs of crNT and crPTX3 EVs. J, sEV secretion per cell normalized to crNT DMSO-EV. N = 3; mean ± SEM; two-way ANOVA; **, P < 0.01. K, Dot plot of crNT sEVs probed for indicated proteins. Membranes were treated with or without Tween-20 during primary antibody incubation to expose internal sEV proteins.

Figure 4.

DXR treatment regulates sEV protein content. A, Volcano plot of MDA-MB-231 DMSO-EV and DXR-EV proteins. Red, upregulated in MDA-MB-231 DXR-EVs; blue, downregulated in MDA-MB-231 DXR-EVs. LogFC >0.585 or ≤0.585, −Log10(adj P) >1.303. PTX3 (yellow) is highly enriched in MDA-MB-231 DXR-EVs; logFC = 1.072, P = 1.0e−6. B, 1,758 proteins were identified in MDA-MB-231 cell–derived sEVs. Forty proteins were significantly upregulated, and 167 were significantly downregulated in MDA-MB-231 DXR-EVs. C, Heat map of relative protein abundance in MDA-MB-231 DXR and DMSO-EVs. N = 4 per treatment condition. Top 15 most abundant proteins in DXR and DMSO sEVs are indicated. D, Kaplan–Meier relapse-free survival curve of breast cancer patients who received neoadjuvant chemotherapy treatment, stratified by PTX3 expression. PTX3 low, n = 112; PTX3 high, n = 111. Created using kmplot.com. E, Immunoblot of MDA-MB-231 crNT and crPTX3 cells probed for indicated proteins. F, Immunoblot of sEVs derived from crNT and crPTX3 MDA-MB-231 cells treated with DMSO or DXR. G, Quantification of PTX3 expression in crNT DMSO- and DXR-EVs, normalized to CD63. N = 4; mean ± SEM; Student t test; *, P < 0.05. H, Representative electron microscope images of crNT and crPTX3 sEVs. Scale bar, 200 nm. I, Representative NTA graphs of crNT and crPTX3 EVs. J, sEV secretion per cell normalized to crNT DMSO-EV. N = 3; mean ± SEM; two-way ANOVA; **, P < 0.01. K, Dot plot of crNT sEVs probed for indicated proteins. Membranes were treated with or without Tween-20 during primary antibody incubation to expose internal sEV proteins.

Close modal

Small extracellular vesicle-associated PTX3 accelerates breast cancer metastasis

To determine how sEV-associated PTX3 affects breast tumor progression, we first confirmed that MDA-MB-231 cell migration and invasion were not enhanced by treatment with crPTX3 sEVs compared with crNT EVs or PBS (Supplementary Fig. S6E and S6F). To establish whether sEV-associated PTX3 accelerates breast cancer metastasis, we repeated our premetastatic niche model by treating female NSG mice three times with 10 μg crNT DXR-EVs or crPTX3 DXR-EVs, or an equivalent volume of PBS, followed by tail-vein injection of MDA-MB-231 cells (Fig. 5A). As expected, bioluminescent imaging confirmed that mice pretreated with crNT DXR-EVs demonstrated a significant acceleration in whole-body metastasis compared with PBS (Fig. 5B and C). However, mice pretreated with crPTX3 DXR-EVs showed a drastic reduction in tumor burden compared with crNT DXR-EV pretreatment, with levels of metastatic progression equivalent to mice pretreated with PBS (Fig. 5B and C). Histologic scoring of lung tumor burden corroborated this finding, despite no significant differences in liver tumor burden between treatment groups (Fig. 5DF). We found no significant differences between crNT and crPTX3 EV uptake in cells in vitro (Supplementary Fig. S6G), suggesting that differences in EV uptake are unlikely responsible for the observed decrease in metastasis.

Figure 5.

Small extracellular vesicle-associated PTX3 accelerates breast cancer metastasis. A, Experimental design. B, Representative IVIS images of MDA-MB-231 metastatic luciferase signal in mice from indicated treatment groups. C, ROI quantification of MDA-MB-231 metastatic tumor radiance. Data represent two independent experiments. PBS, n = 13; crNT DXR-EV, n = 12; crPTX3 DXR-EV, n = 13. Two-way ANOVA, mean ± SEM; ****, P < 0.001. D, Representative hematoxylin and eosin-stained sections of lung and liver tissue from mice treated with crNT and crPTX3 sEVs sacrificed 5 weeks after tumor cell injection. Liver metastases are outlined. E and F, Average lung (E) and liver (F) metastatic tumor burden from mice treated with crNT and crPTX3 sEVs. Data represent one experiment. PBS, n = 8; MDA-MB-231 crNT DXR-EV, n = 7; MDA-MB-231 crPTX3 DXR-EV, n = 8. One-way ANOVA, mean ± SEM; *, P < 0.05; **, P < 0.01; ns, not significant. G, Experimental design. H, Representative IVIS images of metastatic luciferase signal in mice with MDA-MB-231 crNT (top) or crPTX3 (bottom) primary tumors receiving PBS or 5 mg/kg DXR. I, ROI quantification of MDA-MB-231 crNT and crPTX3 metastatic tumor radiance normalized to cell luciferase expression. Data represent one experiment. crNT + PBS, n = 5; crNT + DXR, n = 7; crPTX3 + PBS, n = 7; crPTX3 + DXR, n = 7. Two-way ANOVA; mean ± SEM; ****, P < 0.0001. J, Quantification of PTX3 expression per mL plasma in sEVs derived from crNT tumor-bearing mice treated with PBS or DXR normalized to crNT + PBS average. Data represent one experiment. crNT + PBS, n = 6; crNT + DXR, n = 6. Student t test with Mann–Whitney correction; mean ± SEM; **, P < 0.01.

Figure 5.

Small extracellular vesicle-associated PTX3 accelerates breast cancer metastasis. A, Experimental design. B, Representative IVIS images of MDA-MB-231 metastatic luciferase signal in mice from indicated treatment groups. C, ROI quantification of MDA-MB-231 metastatic tumor radiance. Data represent two independent experiments. PBS, n = 13; crNT DXR-EV, n = 12; crPTX3 DXR-EV, n = 13. Two-way ANOVA, mean ± SEM; ****, P < 0.001. D, Representative hematoxylin and eosin-stained sections of lung and liver tissue from mice treated with crNT and crPTX3 sEVs sacrificed 5 weeks after tumor cell injection. Liver metastases are outlined. E and F, Average lung (E) and liver (F) metastatic tumor burden from mice treated with crNT and crPTX3 sEVs. Data represent one experiment. PBS, n = 8; MDA-MB-231 crNT DXR-EV, n = 7; MDA-MB-231 crPTX3 DXR-EV, n = 8. One-way ANOVA, mean ± SEM; *, P < 0.05; **, P < 0.01; ns, not significant. G, Experimental design. H, Representative IVIS images of metastatic luciferase signal in mice with MDA-MB-231 crNT (top) or crPTX3 (bottom) primary tumors receiving PBS or 5 mg/kg DXR. I, ROI quantification of MDA-MB-231 crNT and crPTX3 metastatic tumor radiance normalized to cell luciferase expression. Data represent one experiment. crNT + PBS, n = 5; crNT + DXR, n = 7; crPTX3 + PBS, n = 7; crPTX3 + DXR, n = 7. Two-way ANOVA; mean ± SEM; ****, P < 0.0001. J, Quantification of PTX3 expression per mL plasma in sEVs derived from crNT tumor-bearing mice treated with PBS or DXR normalized to crNT + PBS average. Data represent one experiment. crNT + PBS, n = 6; crNT + DXR, n = 6. Student t test with Mann–Whitney correction; mean ± SEM; **, P < 0.01.

Close modal

To confirm that PTX3 is involved in chemotherapy-induced metastasis, we performed orthotopic xenografts of crNT and crPTX3 MDA-MB-231 cells in female NRG mice. One week after xenograft, on day 0, we treated mice with DXR or PBS followed by a second treatment on day 5 (Fig. 5G). We found that anterior thoracic metastatic tumor growth was significantly increased in mice with crNT primary tumors treated with DXR compared with PBS, while mice with crPTX3 primary tumors treated with DXR did not show a comparable increase in metastasis (Fig. 5H and I; Supplementary Fig. S6H). Importantly, we found that sEVs isolated from the plasma of crNT tumor-bearing mice treated with DXR demonstrated a significant increase in PTX3 compared with sEVs isolated from mice treated with PBS, confirming that DXR treatment upregulates circulating sEV-associated PTX3 in vivo (Fig. 5J; Supplementary Fig. S6I). Consistently, crNT and crPTX3 tumors treated with DXR were significantly smaller than crNT tumors treated with PBS at the experiment endpoint (Supplementary Fig. S6J–S6M). Taken together, these results demonstrate that DXR treatment significantly upregulates sEV-associated PTX3, which is critically involved in chemotherapy-induced breast cancer metastasis.

Inhibition of small extracellular vesicle secretion and uptake suppresses chemotherapy-induced metastasis

After confirming that sEVs derived from DXR-treated tumor cells accelerate metastasis, we sought to determine whether inhibiting sEV uptake and secretion would suppress chemotherapy-induced metastasis. Heparan sulfate proteoglycans on the surface of secondary cells serve as receptors for sEV binding and internalization, and heparin treatment has been shown to block sEV transfer between cells in vitro (27, 28). We first confirmed that heparin treatment suppresses EV uptake in vivo by treating mice with Vybrant DiD-labeled EVs preincubated with or without 0.2 mg/kg heparin and performing whole-organ fluorescent imaging 24 hours after injection (Fig. 6AC). After imaging we digested whole lung tissue and used flow cytometry to confirm that heparin pretreatment suppressed EV uptake in vivo (Supplementary Fig. S7A and S7B), and used the IncuCyte system to confirm that heparin treatment does not suppress tumor cell migration in vitro (Supplementary Fig. S7C).

Figure 6.

Inhibition of sEV uptake and secretion suppresses chemotherapy-induced metastasis. A and B, Quantification of 24-hour lung (A) and liver (B) uptake of Vybrant DiD-labeled MDA-MB-231 DMSO- and DXR-EVs preincubated with 0.2 mg/kg heparin or PBS, normalized to PBS average. Data represent one experiment, n = 3 samples per treatment condition. Mean ± SEM one-way ANOVA; *, P < 0.05; ****, P < 0.0001; ns, not significant. C, Representative IVIS images of fluorescent DMSO and DXR-EV signal in lung and liver tissue. D, Immunoblot of MDA-MB-231 control (cntrl) and Rab27a KO cells probed for indicated proteins. E, Representative NTA graph of control and crRab27a sEVs. F, Representative electron microscope images of control and crRab27a EVs. Scale bar, 200 nm. G, Experimental design. H, Representative IVIS images of control and crRab27a metastatic tumor growth in mice treated with PBS or 2.5 mg/kg DXR, with or without subcutaneous PBS or 0.2 mg/kg heparin treatment. crRab27a + DXR radiance normalized to relative luciferase expression. I, ROI quantification of MDA-MB-231 cntrl and crRab27a metastatic tumor radiance. crRab27a + DXR radiance normalized to relative luciferase expression. Data represent one experiment. Cntrl + PBS, n = 9; cntrl + DXR + PBS, n = 10; cntrl + DXR + Heparin, n = 10; crRab27a + DXR, n = 10 for days 0 and 5. Cntrl + PBS, n = 9; cntrl + DXR + PBS, n = 9; cntrl + DXR + Heparin, n = 4; crRab27a + DXR, n = 8 for day 10. Two-way ANOVA, mean ± SEM; *, P < 0.05; ***, P < 0.005.

Figure 6.

Inhibition of sEV uptake and secretion suppresses chemotherapy-induced metastasis. A and B, Quantification of 24-hour lung (A) and liver (B) uptake of Vybrant DiD-labeled MDA-MB-231 DMSO- and DXR-EVs preincubated with 0.2 mg/kg heparin or PBS, normalized to PBS average. Data represent one experiment, n = 3 samples per treatment condition. Mean ± SEM one-way ANOVA; *, P < 0.05; ****, P < 0.0001; ns, not significant. C, Representative IVIS images of fluorescent DMSO and DXR-EV signal in lung and liver tissue. D, Immunoblot of MDA-MB-231 control (cntrl) and Rab27a KO cells probed for indicated proteins. E, Representative NTA graph of control and crRab27a sEVs. F, Representative electron microscope images of control and crRab27a EVs. Scale bar, 200 nm. G, Experimental design. H, Representative IVIS images of control and crRab27a metastatic tumor growth in mice treated with PBS or 2.5 mg/kg DXR, with or without subcutaneous PBS or 0.2 mg/kg heparin treatment. crRab27a + DXR radiance normalized to relative luciferase expression. I, ROI quantification of MDA-MB-231 cntrl and crRab27a metastatic tumor radiance. crRab27a + DXR radiance normalized to relative luciferase expression. Data represent one experiment. Cntrl + PBS, n = 9; cntrl + DXR + PBS, n = 10; cntrl + DXR + Heparin, n = 10; crRab27a + DXR, n = 10 for days 0 and 5. Cntrl + PBS, n = 9; cntrl + DXR + PBS, n = 9; cntrl + DXR + Heparin, n = 4; crRab27a + DXR, n = 8 for day 10. Two-way ANOVA, mean ± SEM; *, P < 0.05; ***, P < 0.005.

Close modal

To determine whether heparin treatment suppresses chemotherapy-induced metastasis in vivo, we performed orthotopic xenografts of MDA-MB-231 cells in female NSG mice and treated mice twice over the course of 5 days with tail-vein injections of DXR or PBS. Mice treated with DXR received daily subcutaneous injections of 0.2 mg/kg heparin or an equivalent volume of PBS (Fig. 6G). Quantification of anterior thoracic bioluminescent imaging confirmed that DXR treatment accelerated metastasis compared with treatment with PBS (Fig. 6H and I). Significantly, mice treated with daily subcutaneous doses of heparin demonstrated a complete suppression of DXR-induced metastasis (Fig. 6H and I). Of note, both groups of mice treated with DXR showed a decrease in primary tumor growth compared with mice treated with PBS, with no difference in tumor growth between the two groups, indicating that DXR significantly decreased primary tumor growth and that heparin treatment suppressed chemotherapy-induced metastasis without affecting primary tumor growth (Supplementary Fig. S7D and S7E).

To validate that primary tumor-derived sEVs are involved in chemotherapy-induced metastasis, we used CRISPR/Cas9 to KO RAB27A, a Rab-GTPase involved in shuttling the MVB to the plasma membrane for sEV secretion, in MDA-MB-231 cells (crRab27a; Fig. 6D). We confirmed that RAB27A KO does not affect MDA-MB-231 proliferation in vitro (Supplementary Fig. S7F), and used NTA and electron microscopy to demonstrate that RAB27A KO decreases sEV secretion (Fig. 6E and F). We performed xenografts of crRab27a MDA-MB-231 cells in female NSG mice in parallel with control MDA-MB-231 xenografts, and treated mice twice with DXR (Fig. 6G). Using bioluminescent imaging, we confirmed that genetic inhibition of sEV secretion significantly decreased metastatic tumor growth compared with control tumors treated with DXR (Fig. 6H and I; Supplementary Fig. S7G) and found that DXR decreased both control and crRab27a primary tumor size compared with PBS treatment (Supplementary Fig. S7D and S7E). These results demonstrate that sEVs derived from chemotherapy-treated primary tumor cells are critically involved in promoting breast cancer metastasis.

Literature reports that chemotherapeutic drugs can decrease primary tumor growth while promoting metastasis (8, 26, 29, 30). In this study, we investigated the role of chemotherapy-induced, primary tumor-derived sEVs in cancer progression. We first confirmed that DXR treatment accelerated metastasis in a mouse xenograft model of human TNBC and found that low doses of DXR significantly enhanced sEV secretion in TNBC cell lines in vitro as well as sEV circulation in tumor-bearing mice in vivo, corroborating literature accounts that sEV secretion is upregulated in response to cellular stress (31, 32). We found that DXR-EV treatment accelerated pulmonary metastasis in mouse breast cancer xenograft models compared with control sEV treatment, without affecting primary tumor growth. In agreement with previous reports (33), we found that both DMSO- and DXR-EVs are taken up by lung and liver tissue in vivo and saw no significant differences in the level of sEV uptake between these two groups, suggesting that the sEV cargo, rather than differences in uptake, was primarily responsible for the increase in pulmonary metastasis. We observed a significant acceleration in whole-body and pulmonary metastatic tumor growth in mice that had been pretreated with DXR-EVs compared with DMSO-EVs prior to tumor cell tail-vein injection, demonstrating that DXR-EVs prime the premetastatic niche to accelerate metastasis. While liver metastasis was not significantly affected by sEV treatment, we did observe a slight increase in liver metastasis following MDA-MB-231 DXR-EV treatment in both our primary tumor and premetastatic niche models, suggesting that DXR-EVs may prime the hepatic premetastatic niche to a lesser extent than the pulmonary niche. Similarly, mice pretreated with MDA-MB-468 sEVs demonstrated an increase in bone marrow uptake, highlighting differences in EV organotropism between the two breast cancer cell lines. Proteomic analysis of MDA-MB-231 sEVs revealed over 200 differentially expressed proteins in DXR-EVs. We focused on the upregulated TNF-inducible pattern recognition molecule PTX3, which we found predicts decreased event-free survival when elevated in breast cancer patients following NAC. Mice pretreated with crPTX3 DXR-EVs showed reduced pulmonary metastasis compared with mice pretreated with crNT DXR-EVs prior to tail-vein injection of MDA-MB-231 cells, demonstrating a critical role for PTX3 in sEV-mediated metastasis. Strikingly, while DXR treatment enhanced metastasis in crNT xenografts, metastasis was attenuated in crPTX3 xenografts treated with DXR. Taken together, these data suggest a model whereby DXR treatment upregulates both the secretion of primary breast tumor sEVs and the amount of PTX3 sEV cargo. Uptake of these tumor-derived DXR-EVs in secondary tissues, primarily the lung, primes the premetastatic niche, creating a favorable environment for metastatic breast tumor cell colonization.

One limitation of our study is that we utilized immunodeficient NSG and NRG mice to study the effect of sEVs on human TNBC metastasis. Although chemotherapy treatment weakens the host immune system, immunodeficient mice are not a truly representative model of breast cancer patients, and future studies are necessary to determine how sEV-associated PTX3 regulates metastasis in an immunocompetent model. However, in their 2019 paper, Keklikoglou and colleagues reported that chemotherapy treatment is able to enhance metastasis in both immunodeficient and immunocompetent transgenic mouse models of breast cancer (26). They found that paclitaxel and DXR-treated breast cancer cells release sEVs enriched in annexin A6 (ANXA6), which they found promotes lung metastasis through the activation of endothelial cells, induction of Ccl2, and expansion of monocytes within the premetastatic niche (26). Although we identified ANXA6 in our proteomic screen, it was not significantly enriched in our DXR-EVs, suggesting that different chemotherapeutic drugs likely regulate sEV protein cargo in a dose- and context-dependent manner. Because sEVs contain thousands of proteins and RNAs, this also suggests that ANXA6 is not the only sEV protein responsible for accelerating pulmonary metastasis, and that sEVs derived from chemotherapy-treated cells are likely capable of promoting tumor progression through multiple pathways, either in parallel or in concert. In our model, we found that PTX3 expression is upregulated approximately 2-fold in DXR-EVs derived from the highly metastatic MDA-MB-231 cell line, which expresses a high level of PTX3 mRNA, and upregulated approximately 1.5-fold in DXR-EVs derived from the poorly metastatic MDA-MB-468 and MCF7 cell lines, which express low levels of PTX3 mRNA. While this demonstrates that upregulation of sEV-associated PTX3 is a common mechanism of DXR treatment, it remains an interesting question whether highly metastatic cell lines are more reliant on PTX3-mediated priming of the premetastatic niche than poorly metastatic lines. Indeed, literature demonstrates that different chemotherapeutic agents upregulate various prometastatic sEV cargo, including heparanase (34) and miR-126a (30), highlighting the need to fully investigate the various mechanisms by which different drug-induced cargo accelerate metastasis.

PTX3 has a complex role in tumor progression (35) and has been shown to promote tumor metastasis and chemoresistance following treatment with the chemotherapeutic drugs cisplatin and 5-fluorouracil (36). As a pattern-recognition molecule induced by inflammation, PTX3 plays a critical role in host defense against infectious pathogens, tissue repair, and tumor-related inflammation (37). PTX3 has been shown to interact with Factor H and C1q to regulate complement activation, leading to leukocyte recruitment, proinflammatory cytokine production, and angiogenesis within the tumor microenvironment (38). Although the role of PTX3 in tumor inflammation is well characterized, our model of human TNBC utilizes immunodeficient NSG and NRG mice, which lack T, B, and NK cells, and have deficient cytokine signaling pathways, suggesting that cytokine-mediated inflammation may not be the primary mechanism by which DXR-EV-associated PTX3 accelerates metastasis. In addition to its role in complement activation, PTX3 is also associated with vascular endothelial dysfunction in a variety of pathologies (39, 40). PTX3 has been shown to induce morphologic changes that lead to dysfunction in the endothelial layer through the P-selectin and matrix metalloproteinase-1 (MMP-1) pathway, another protein that was found to be highly upregulated in DXR-EVs in our proteomic screen (41, 42). PTX3 is able to bind to P-selectin on the surface of vascular endothelial cells, leading to reduced nitric oxide production, inhibition of cell proliferation, increased expression of MMP-1, and endothelial cell dysfunction (41). Vessels exposed to PTX3 exhibit broken plasma membranes, diluted cytoplasm, and increased vascular leakage (41). We found that sEV-associated PTX3 localizes to the surface of the vesicle, suggesting that DXR EV-associated PTX3 may bind to P-selectin on the surface of vascular endothelial cells, promoting endothelial dysfunction and vascular leakiness that would enhance the ability of circulating tumor cells (CTC) and prometastatic immune cells to extravasate into the lungs. Chemotherapy treatment has previously been shown to promote metastasis by enhancing the ability of CTCs to intravasate into the vasculature (8), but the role of PTX3 in creating microenvironments of metastasis is unknown. Future research is necessary to determine the role of DXR-EVs on vascular dysfunction in both immunodeficient and immunocompetent mouse models, and to determine whether PTX3 may be a suitable therapeutic target to suppress chemotherapy-induced metastasis and enhance the efficacy of NAC.

Finally, we hypothesized that if DXR-EVs accelerate metastasis, inhibition of sEV secretion from the primary tumor or uptake in secondary tissues would suppress metastasis. The anticoagulant heparin has been shown to inhibit the binding and uptake of sEVs in secondary cells, so we hypothesized that heparin treatment could suppress sEV-mediated metastasis (28). Importantly, we found that mice cotreated with DXR and daily subcutaneous injections of heparin demonstrated an almost complete suppression in metastasis compared with mice treated with DXR and subcutaneous injections of PBS, suggesting that inhibition of primary tumor-derived DXR-EV uptake in secondary tissues does decrease metastasis. To confirm that inhibition of sEV uptake, rather than an off-target effect of heparin, was responsible for the observed suppression in metastasis, we knocked out RAB27A, a Rab-GTPase necessary for sEV secretion, in MDA-MB-231 cells. We found that mice xenografted with crRab27a primary tumors did not demonstrate an increase in metastasis following DXR treatment compared with mice bearing control tumors, reinforcing the finding that primary tumor-derived DXR-EVs play a critical role in chemotherapy-mediated metastasis. Heparin has long been thought to suppress metastasis in various cancer models, but its clinical use has been limited due to the increased risk of patient bleeding (43–45). Our research demonstrates a novel mechanism by which heparin is able to suppress chemotherapy-induced metastasis through the inhibition of DXR-EV uptake in secondary tissues, and suggests that further studies are warranted to determine whether heparin and its derivatives could be utilized as a therapeutic tool for high-risk TNBC patients.

H.G. Wang reports grants from NIH during the conduct of the study. No disclosures were reported by the other authors.

C.A. Wills: Conceptualization, data curation, formal analysis, validation, investigation, visualization, methodology, writing–original draft, writing–review and editing. X. Liu: Data curation and investigation. L. Chen: Resources, data curation, investigation, and methodology. Y. Zhao: Investigation and methodology. C.M. Dower: Conceptualization, resources, and methodology. J. Sundstrom: Resources, software, supervision, investigation, and methodology. H.-G. Wang: Conceptualization, resources, supervision, funding acquisition, project administration, writing–review and editing.

We acknowledge all members of the Wang and Sundstrom labs, the Penn State College of Medicine Department of Pediatrics, Department of Ophthalmology, Department of Comparative Medicine, Bioluminescent Imaging Core, Transmission Electron Microscopy Core, and Flow Cytometry Core, as well as the University of Michigan Proteomics Resource Facility. This work was supported in part by the Lois High Berstler Research Endowment Fund, the Four Diamonds Fund, and the NIH Grant 5T32CA060395.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Siegel
RL
,
Miller
KD
,
Jemal
A
. 
Cancer statistics, 2017
.
CA Cancer J Clin
2017
;
67
:
7
30
.
2.
Li
Y
,
Yang
D
,
Chen
P
,
Yin
X
,
Sun
J
,
Li
H
, et al
Efficacy and safety of neoadjuvant chemotherapy regimens for triple-negative breast cancer: a network meta-analysis
.
Aging
2019
;
11
:
6286
311
.
3.
Bianchini
G
,
Balko
JM
,
Mayer
IA
,
Sanders
ME
,
Gianni
L
. 
Triple-negative breast cancer: challenges and opportunities of a heterogeneous disease
.
Nat Rev Clin Oncol
2016
;
13
:
674
90
.
4.
Kassam
F
,
Enright
K
,
Dent
R
,
Dranitsaris
G
,
Myers
J
,
Flynn
C
, et al
Survival outcomes for patients with metastatic triple-negative breast cancer: implications for clinical practice and trial design
.
Clin Breast Cancer
2009
;
9
:
29
33
.
5.
Liedtke
C
,
Mazouni
C
,
Hess
KR
,
André
F
,
Tordai
A
,
Mejia
JA
, et al
Response to neoadjuvant therapy and long-term survival in patients with triple-negative breast cancer
.
J Clin Oncol
2008
;
26
:
1275
81
.
6.
von Minckwitz
G
,
Untch
M
,
Blohmer
J-U
,
Costa
SD
,
Eidtmann
H
,
Fasching
PA
, et al
Definition and impact of pathologic complete response on prognosis after neoadjuvant chemotherapy in various intrinsic breast cancer subtypes
.
J Clin Oncol
2012
;
30
:
1796
804
.
7.
Ratajczak
MZ
,
Jadczyk
T
,
Schneider
G
,
Kakar
SS
,
Kucia
M
. 
Induction of a tumor-metastasis-receptive microenvironment as an unwanted and underestimated side effect of treatment by chemotherapy or radiotherapy
.
J Ovarian Res
2013
;
6
:
95
.
8.
Karagiannis
GS
,
Pastoriza
JM
,
Wang
Y
,
Harney
AS
,
Entenberg
D
,
Pignatelli
J
, et al
Neoadjuvant chemotherapy induces breast cancer metastasis through a TMEM-mediated mechanism
.
Sci Transl Med
2017
;
9
:
eaan0026
.
9.
Daenen
LGM
,
Roodhart
JML
,
van Amersfoort
M
,
Dehnad
M
,
Roessingh
W
,
Ulfman
LH
, et al
Chemotherapy enhances metastasis formation via VEGFR-1-expressing endothelial cells
.
Cancer Res
2011
;
71
:
6976
85
.
10.
Hung
C-M
,
Hsu
Y-C
,
Chen
T-Y
,
Chang
C-C
,
Lee
M-J
. 
Cyclophosphamide promotes breast cancer cell migration through CXCR4 and matrix metalloproteinases
.
Cell Biol Int
2017
;
41
:
345
52
.
11.
Park
SI
,
Liao
J
,
Berry
JE
,
Li
X
,
Koh
AJ
,
Michalski
ME
, et al
Cyclophosphamide creates a receptive microenvironment for prostate cancer skeletal metastasis
.
Cancer Res
2012
;
72
:
2522
32
.
12.
Karagiannis
GS
,
Condeelis
JS
,
Oktay
MH
. 
Chemotherapy-induced metastasis: mechanisms and translational opportunities
.
Clin Exp Metastasis
2018
;
35
:
269
84
.
13.
Steinbichler
TB
,
Dudás
J
,
Riechelmann
H
,
Skvortsova
I-I
. 
The role of exosomes in cancer metastasis
.
Semin Cancer Biol
2017
;
44
:
170
81
.
14.
Milane
L
,
Singh
A
,
Mattheolabakis
G
,
Suresh
M
,
Amiji
MM
. 
Exosome mediated communication within the tumor microenvironment
.
J Control Release
2015
;
219
:
278
94
.
15.
Peinado
H
,
Alečković
M
,
Lavotshkin
S
,
Matei
I
,
Costa-Silva
B
,
Moreno-Bueno
G
, et al
Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET
.
Nat Med
2012
;
18
:
883
91
.
16.
Greening
DW
,
Gopal
SK
,
Xu
R
,
Simpson
RJ
,
Chen
W
. 
Exosomes and their roles in immune regulation and cancer
.
Semin Cell Dev Biol
2015
;
40
:
72
81
.
17.
Rana
S
,
Malinowska
K
,
Zöller
M
. 
Exosomal tumor microRNA modulates premetastatic organ cells
.
Neoplasia
2013
;
15
:
281
95
.
18.
Luga
V
,
Zhang
L
,
Viloria-Petit
AM
,
Ogunjimi
AA
,
Inanlou
MR
,
Chiu
E
, et al
Exosomes mediate stromal mobilization of autocrine Wnt-PCP signaling in breast cancer cell migration
.
Cell
2012
;
151
:
1542
56
.
19.
Rahman
MA
,
Barger
JF
,
Lovat
F
,
Gao
M
,
Otterson
GA
,
Nana-Sinkam
P
. 
Lung cancer exosomes as drivers of epithelial mesenchymal transition
.
Oncotarget
2016
;
7
:
54852
66
.
20.
Ribeiro
MF
,
Zhu
H
,
Millard
RW
,
Fan
G-C
. 
Exosomes function in pro- and anti-angiogenesis
.
Curr Angiogenes
2013
;
2
:
54
9
.
21.
Takahashi
Y
,
Hori
T
,
Cooper
TK
,
Liao
J
,
Desai
N
,
Serfass
JM
, et al
Bif-1 haploinsufficiency promotes chromosomal instability and accelerates Myc-driven lymphomagenesis via suppression of mitophagy
.
Blood
2013
;
121
:
1622
32
.
22.
Tang
Z
,
Takahashi
Y
,
Chen
C
,
Liu
Y
,
He
H
,
Tsotakos
N
, et al
Atg2A/B deficiency switches cytoprotective autophagy to non-canonical caspase-8 activation and apoptosis
.
Cell Death Differ
2017
;
24
:
2127
38
.
23.
Kowal
J
,
Arras
G
,
Colombo
M
,
Jouve
M
,
Morath
JP
,
Primdal-Bengtson
B
, et al
Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes
.
Proc Natl Acad Sci U S A
2016
;
113
:
E968
77
.
24.
Zhao
Y
,
Weber
SR
,
Lease
J
,
Russo
M
,
Siedlecki
CA
,
Xu
L-C
, et al
Liquid biopsy of vitreous reveals an abundant vesicle population consistent with the size and morphology of exosomes
.
Transl Vis Sci Technol
2018
;
7
:
6
.
25.
Rikkert
LG
,
Nieuwland
R
,
Terstappen
LWMM
,
Coumans
FAW
. 
Quality of extracellular vesicle images by transmission electron microscopy is operator and protocol dependent
.
J Extracell Vesicles.
2019
;
8
:
1555419
.
26.
Keklikoglou
I
,
Cianciaruso
C
,
Güç
E
,
Squadrito
ML
,
Spring
LM
,
Tazzyman
S
, et al
Chemotherapy elicits pro-metastatic extracellular vesicles in breast cancer models
.
Nat Cell Biol
2019
;
21
:
190
202
.
27.
Christianson
HC
,
Svensson
KJ
,
Kuppevelt
TH van
,
Li
J-P
,
Belting
M
. 
Cancer cell exosomes depend on cell-surface heparan sulfate proteoglycans for their internalization and functional activity
.
Proc Natl Acad Sci U S A
2013
;
110
:
17380
5
.
28.
Atai
NA
,
Balaj
L
,
van Veen
H
,
Breakefield
XO
,
Jarzyna
PA
,
Van Noorden
CJF
, et al
Heparin blocks transfer of extracellular vesicles between donor and recipient cells
.
J Neurooncol
2013
;
115
:
343
51
.
29.
Liu
C-L
,
Chen
M-J
,
Lin
J-C
,
Lin
C-H
,
Huang
W-C
,
Cheng
S-P
, et al
Doxorubicin promotes migration and invasion of breast cancer cells through the upregulation of the RhoA/MLC pathway
.
J Breast Cancer
2019
;
22
:
185
95
.
30.
Deng
Z
,
Rong
Y
,
Teng
Y
,
Zhuang
X
,
Samykutty
A
,
Mu
J
, et al
Exosomes miR-126a released from MDSC induced by DOX treatment promotes lung metastasis
.
Oncogene
2017
;
36
:
639
51
.
31.
Sweeney
R
,
Richards
KE
,
Hill
R
. 
Gemcitabine-induced exosome hypersecretion increases the chemoresistance and migration of pancreatic cancer cells
.
FASEB J
2017
;
31
:
775.20
.
32.
Aubertin
K
,
Silva
AKA
,
Luciani
N
,
Espinosa
A
,
Djemat
A
,
Charue
D
, et al
Massive release of extracellular vesicles from cancer cells after photodynamic treatment or chemotherapy
.
Sci Rep
2016
;
6
:
1
11
.
33.
Hoshino
A
,
Costa-Silva
B
,
Shen
T-L
,
Rodrigues
G
,
Hashimoto
A
,
Tesic Mark
M
, et al
Tumour exosome integrins determine organotropic metastasis
.
Nature
2015
;
527
:
329
35
.
34.
Bandari
SK
,
Purushothaman
A
,
Ramani
VC
,
Brinkley
GJ
,
Chandrashekar
DS
,
Varambally
S
, et al
Chemotherapy induces secretion of exosomes loaded with heparanase that degrades extracellular matrix and impacts tumor and host cell behavior
.
Matrix Biol
2018
;
65
:
104
18
.
35.
Giacomini
A
,
Ghedini
GC
,
Presta
M
,
Ronca
R
. 
Long pentraxin 3: a novel multifaceted player in cancer
.
Biochim Biophys Acta Rev Cancer
2018
;
1869
:
53
63
.
36.
Chi
J-Y
,
Hsiao
Y-W
,
Li
C-F
,
Lo
Y-C
,
Lin
Z-Y
,
Hong
J-Y
, et al
Targeting chemotherapy-induced PTX3 in tumor stroma to prevent the progression of drug-resistant cancers
.
Oncotarget
2015
;
6
:
23987
4001
.
37.
Doni
A
,
Stravalaci
M
,
Inforzato
A
,
Magrini
E
,
Mantovani
A
,
Garlanda
C
, et al
The long pentraxin PTX3 as a link between innate immunity, tissue remodeling, and cancer
.
Front Immunol
2019
;
10
:
712
.
38.
Bonavita
E
,
Gentile
S
,
Rubino
M
,
Maina
V
,
Papait
R
,
Kunderfranco
P
, et al
PTX3 is an extrinsic oncosuppressor regulating complement-dependent inflammation in cancer
.
Cell
2015
;
160
:
700
14
.
39.
Hamad
RR
,
Eriksson
MJ
,
Berg
E
,
Larsson
A
,
Bremme
K
. 
Impaired endothelial function and elevated levels of pentraxin 3 in early-onset preeclampsia
.
Acta Obstet Gynecol Scand
2012
;
91
:
50
6
.
40.
Suliman
ME
,
Yilmaz
MI
,
Carrero
JJ
,
Qureshi
AR
,
Saglam
M
,
Ipcioglu
OM
, et al
Novel links between the long Pentraxin 3, endothelial dysfunction, and albuminuria in early and advanced chronic kidney disease
.
CJASN
2008
;
3
:
976
85
.
41.
Carrizzo
Albino
,
Lenzi
Paola
,
Procaccini
Claudio
,
Damato
Antonio
,
Biagioni
Francesca
,
Ambrosio
Mariateresa
, et al
Pentraxin 3 induces vascular endothelial dysfunction through a P-selectin/Matrix metalloproteinase-1 pathway
.
Circulation
2015
;
131
:
1495
505
.
42.
Zlibut
A
,
Bocsan
IC
,
Agoston-Coldea
L
. 
Pentraxin-3 and endothelial dysfunction
.
Adv Clin Chem
2019
;
91
:
163
79
.
43.
Alyahya
R
,
Sudha
T
,
Racz
M
,
Stain
SC
,
Mousa
SA
. 
Anti-metastasis efficacy and safety of non-anticoagulant heparin derivative versus low molecular weight heparin in surgical pancreatic cancer models
.
Int J Oncol
2015
;
46
:
1225
31
.
44.
Harvey
JR
,
Mellor
P
,
Kirby
JA
,
Lennard
T
,
Ali
S
. 
Preventing breast cancer metastasis - heparins as inhibitors of chemokine function
.
Cancer Res
2005
;
65
:
1229
30
.
45.
Seeholzer
N
,
Thürlimann
B
,
Köberle
D
,
Hess
D
,
Korte
W
. 
Combining chemotherapy and low-molecular-weight heparin for the treatment of advanced breast cancer: results on clinical response, transforming growth factor-beta 1 and fibrin monomer in a phase II study
.
Blood Coagul Fibrinolysis
2007
;
18
:
415
23
.