Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2890 2024-02-17 06:33:41 |
2 format correct Meta information modification 2890 2024-02-17 06:35:18 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Mavrogenis, A.F.; Altsitzioglou, P.; Tsukamoto, S.; Errani, C. Biopsy Techniques for Musculoskeletal Tumors. Encyclopedia. Available online: https://encyclopedia.pub/entry/55107 (accessed on 03 May 2024).
Mavrogenis AF, Altsitzioglou P, Tsukamoto S, Errani C. Biopsy Techniques for Musculoskeletal Tumors. Encyclopedia. Available at: https://encyclopedia.pub/entry/55107. Accessed May 03, 2024.
Mavrogenis, Andreas F., Pavlos Altsitzioglou, Shinji Tsukamoto, Costantino Errani. "Biopsy Techniques for Musculoskeletal Tumors" Encyclopedia, https://encyclopedia.pub/entry/55107 (accessed May 03, 2024).
Mavrogenis, A.F., Altsitzioglou, P., Tsukamoto, S., & Errani, C. (2024, February 17). Biopsy Techniques for Musculoskeletal Tumors. In Encyclopedia. https://encyclopedia.pub/entry/55107
Mavrogenis, Andreas F., et al. "Biopsy Techniques for Musculoskeletal Tumors." Encyclopedia. Web. 17 February, 2024.
Biopsy Techniques for Musculoskeletal Tumors
Edit

Biopsy is a pivotal component in the diagnostic process of bone and soft tissue tumors. The objective is to obtain adequate tissue without compromising local tumor dissemination and the patient’s survival.

biopsy percutaneous incisional liquid

1. Introduction

Biopsy is a compromise between representative tissue sampling and avoidance of tissue contamination. Various biopsy techniques, including closed/percutaneous (imaging-guided or free hand; fine-needle aspiration, core needle) and open (incisional or excisional) biopsies, aim to provide representative tissue sample while minimizing complications [1]. The incidence of complications for closed biopsies ranges from 0 to 10%, compared to up to 16% for open biopsies [2]. The primary complications associated with the biopsy approach are hemorrhage, nerve apraxia, and infection [2].

2. Liquid Biopsy

Liquid biopsy acquired from bodily fluids near malignant cells provides valuable information without invasive tissue removal [3][4]. Especially for tumors that are difficult to approach, even a least invasive percutaneous biopsy may be uncomfortable and increase the risk of complications. Additionally, conventional biopsies may only cover a limited portion of the tumor, which may not accurately reflect it [5]. Blood is the most suitable bodily fluid for liquid biopsy, while urine, cerebrospinal fluid, and saliva may also be beneficial depending on the primary tumor [4]. Liquid biopsies from blood can be used for multiplexed cancer profiling analyses including circulating biomarkers (such as cross-linked type 1 collagen, bone sialoprotein, TRAcP5B, osteoprotegerin) and metabolites (such as pyridinoline and deoxypyridino). Three basic categories of actionable biological components may be acquired from a blood liquid biopsy, including CTCs, cell-free ctDNA, and EVs, or exosomes [6].

2.1. Circulating Tumor Cells

CTCs are tumor cells that enter the bloodstream. Ashworth initially recognized them almost 150 years ago, and they are now widely employed in therapeutic practice [7]. Isolating CTCs has been challenging due to their low number in the general circulation. However, new microfluidic platforms like the U.S. Food and Drug Administration (FDA)-approved Cell Search platform and next-generation sequencing allow for deep phenotyping of every isolated tumor cell [8]. Recent improvements enable liquid biopsies to reveal the genomic mosaicism, mutational landscape, epigenetics, and gene and protein expression of the original tumor.

2.2. Circulating Tumor DNA

The majority of ctDNA found in the general circulation is 180–200 bp in length, indicating that it originates from apoptotic and necrotic primary tumor cells [6]. Tumor DNA may reveal mutations and copy number variation, evaluating the need for specific-target treatment. For instance, the V600E mutation in BRAF is seen in several cancers such as metastatic colorectal cancer, melanoma, and papillary thyroid carcinoma [9][10][11][12]. Targeting the V600E form of BRAF with medicines such vemurafenib, dabrafenib, and trametinib may influence treatment choices [13].

2.3. Extracellular Vesicles (EVs)

EVs are lipid bilayer particles with a diameter of 30 to 1000 nm that may be divided into three types: apoptotic bodies, large EVs (microvesicles), and tiny EVs (exosomes). The biogenesis and biological function vary [14][15]. Cell-secreted EVs can include other biological components, such as DNA, RNA, and proteins, which can be used in determining a diagnosis and prognosis [14][16]. More EVs are produced in cancer cells than normal cells, and play an important role in bone metastases, osteosarcoma, and Ewing’s sarcoma [17][18]. In addition, normal and cancer cells show increased EV production due to hypoxia, increased intracellular calcium or pH, oxidative stress, ionizing radiation, and ultrasound [19]. Further, while RNA can degrade in circulation, encapsulating the RNA in EVs makes them more stable and amenable to transcriptional analyses [20]. Using various methods to gather biological data from patients’ blood may aid in accurate diagnosis, prognosis, and monitoring drug-resistant clones for informed treatment decisions [21]. Due to cost, scalability, repeatability, and separation procedures, bone liquid biopsy is not widely used in clinical trials, despite preclinical breakthroughs. However, it has shown substantial advances and preclinical and clinical uses in bone tumors including secondary and primary malignancies.

3. Liquid Biopsy in Bone Metastases

3.1. Circulating Tumor DNA

Circulating tumor DNA analysis may reveal the mutational landscape of metastatic disease and predict recurrence or response to therapy [22]. A retrospective study of primary breast cancer patients detected metastatic disease, including bone metastases, by measuring tumor-specific chromosomal rearrangements in ctDNA using droplet-based digital PCR technologies from plasma samples, nearly 1 year before clinical recurrence detection. The amount of ctDNA was directly proportional to disease progression. This suggests that ctDNA detection may be a useful technique for early metastasis detection. Liquid biopsy may also reveal minimum residual disease (MRD), indicating treatment and prognosis. Plasma tumor-associated ctDNA detection and analysis are effective indicators for identifying and monitoring MRD in breast cancer patients at high risk of recurrence [23][24]. Levels of ctDNA at baseline are linked to increased bone metastases and poor prognosis in non-small cell lung cancer (NSCLC) patients [25]. Late-stage NSCLC patients show increased ctDNA levels in patients with bone metastases [26], while higher ctDNA levels are detected in prostate cancer patients with visceral metastases than in those with bone metastases [27]. Detecting MRD using liquid biopsy is still in its infancy, and larger-scale longitudinal investigations are needed to investigate false positives and negative cases.

3.2. Circulating Tumor Cells

Circulating tumor cells have been shown to be beneficial for the diagnosis, prognosis, and monitoring of patients with metastatic breast cancer; in these patients, CTC detection and characterization has been linked to bone and hepatic metastases [28]. Multiple metastatic locations are associated with greater CTC counts, but bone-only metastatic breast cancer patients have lower CTC counts and improved prognoses [29][30]. Patients with one or two bone metastases show significantly fewer CTCs than those with more bone metastases [30].
CTCs exhibit a subset of metastasis-initiating cells expressing CD44, CD47, and c-MET. When transferred from a patient to immunocompromised animals, these cells cause metastases in the lungs, liver, and bones [31]. The discovery and quantification of CTCs may predict lung cancer prognosis, with high CTC counts as a predictor of bone metastases in advanced lung cancer patients [32][33] and in monitoring bone metastases in castration-resistant prostate cancer patients. Other studies indicate that CTC counts > 5 per 7.5 mL of blood predict bone metastases and worse overall survival [34][35][36]. Another study recommended a liquid biopsy method that detects and quantifies both CTCs and ctDNA simultaneously. Peripheral blood samples collected before and after treatment in a homogenous cohort of HER2-negative breast cancer patients [37] were examined prospectively in the COMET trial (NCT01745757). Compared to non-metastatic patients, greater CTCs and ctDNA mutations in tumor protein 53, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha, and estrogen receptor 1 (ESR1) genes were observed in bone, liver, and brain metastasis patients [37]. Similarly, results of other investigations have indicated that the CTCs-ctDNA signature is effective for the diagnosis and prognosis of metastatic breast cancer [38][39][40][41].

3.3. Extracellular Vesicles Cargo and miRNAs

Bhadresha et al. [42] identified 15 genes consistently upregulated in bone metastasis patients. Using serum-derived EVs, five genes (HSP90AA1, osteopontin, IL-3, VEGFA, and protein tyrosine kinase 2) were upregulated in breast and lung cancer patients with bone metastasis. The results indicated that EV-derived mRNA may be used to detect early bone metastases in cases of breast or lung cancer [42]. Plasma-derived EV miRNAs (hsa-miR-574-5p, 328-3p and 423-3p) from NSCLC patients were examined retrospectively as early biomarkers of bone metastases [43]. In that study, Yang et al. observed that EV-derived miR-181a-5p- is increased in prostate cancer patients with bone metastases. Bryant et al. obtained similar results for miR-141 and miR-375 [44]. Prostate microparticle-specific EVs have been reported to be more prevalent in metastatic prostate cancer than in no-metastatic disease and outperformed the FDA-approved CellSearch system in predicting CTCs [45]. ExoDX, a recently evaluated urine exosomal gene expression platform, outperformed the gold standard in predicting high-grade prostate cancer in patients with uncertain PSA scores (ROC AUC 0.7 vs. 0.62) and identifying benign prostate hyperplasia, reducing unnecessary biopsies [46].

4. Liquid Biopsy in Bone Sarcomas

4.1. Circulating Tumor DNA

Few studies have examined plasma-derived ctDNAs from osteosarcoma patients [47][48]. One study examined somatic mutations with tumor burden and prognosis using targeted next-generation sequencing (NGS) to detect tumor-specific somatic alterations in plasma samples at various stages of treatment, allowing for disease burden monitoring [47]. In the research of Shulman et al. [48], NGS hybrid capture assay ctDNA levels in peripheral blood samples of newly diagnosed localized osteosarcoma and Ewing sarcoma patients were linked to tumor burden, recurrence, and poor clinical outcomes; interestingly, ctDNA analysis revealed unexpected genetic characteristics of osteosarcoma, such as chromosomal arm 8q copy number increases [48]. Genetic mutations, including STAG2 and TP53 loss-of-function mutations, translocation events and fusion genes, have been identified in Ewing sarcoma patients, allowing for ctDNA monitoring of the bone malignancy [49]. In the study of Shulman et al. [48], ctDNA detection in plasma samples was linked to a poor clinical outcome in newly diagnosed Ewing sarcoma patients and revealed genomic information like EWSR1 fusion and STAG2 loss-of-function mutations [48]. Hayashi et al. observed that plasma EWSR1-FLI1 fusion gene levels are associated to tumor burden and therapeutic response in Ewing sarcoma patients, suggesting another potential use for liquid biopsy. In addition, EWS-FLI1 levels in the blood fall following chemotherapy or surgery and subsequently increase after tumor recurrence [50].

4.2. Circulating Tumor Cells

Osteosarcoma metastasis may be predicted by CTCs [51]. Li et al. [52] found additional CTCs in peripheral blood of metastatic osteosarcoma patients compared to those with localized disease in a prospective analysis. Additionally, CTC count has been shown to be negatively linked with the patient’s response following neoadjuvant chemotherapy [52]. Preclinical studies indicate that CTC count variations after treatment or surgery can indicate tumor sensitivity and metastasis [53][54]. An increased percentage of mesenchymal CTCs in peripheral blood of osteosarcoma patients after chemotherapy treatment has been linked to lower disease-free survival. This highlights the importance of monitoring changes in CTCs to assess treatment efficacy and detect disease recurrence or metastasis. In Ewing sarcoma patients, CTC characterization using tumor-specific markers (i.e., CD99 expression) and chromosomal translocations (e.g., EWSR1-FLI1 transcript fusion gene amplification) has been described [55][56]. In those patients, CTCs detected at diagnosis correlates with worse clinical outcomes and increased recurrence disease and metastasis [56][57].

4.3. Extracellular Vesicles Cargo and miRNAs

A liquid biopsy has been used to study EVs as diagnostic or prognostic serum indicators in osteosarcoma. RNA analysis of circulating EVs in metastatic osteosarcoma samples has revealed various transcriptome changes, offering a novel therapeutically useful method for tracking metastatic osteosarcoma [58]. Osteosarcoma patients’ peripheral blood contains miRNAs that are known to partly circulate within EVs and have oncogenic or antitumor suppressive functions. Several biomarkers, including miR-148a [59], miR-574-3p, miR-214, miR-335-5p, miR-491, miR-221, miR-191, and miR-421, are becoming important diagnostic and prognostic indicators, while osteosarcoma patients have lower levels of miR-124, miR-101, and miR-195 in their blood compared to those of healthy persons [60][61][62][63][64]. These data may be used to develop a prognostic approach for osteosarcoma employing a mix of miRNAs.
Recently, Ewing sarcoma has been studied for circulating miRNAs. One example of a circulating miRNA linked to Ewing sarcoma development is miR-125b, which has been shown to be lower in patients’ blood after surgery compared to that in healthy controls [65]. Decreased expression of this gene has been linked to a poor chemotherapy response in the same study [65]. Research is now focusing on Ewing sarcoma-derived EVs cargo as a predictive biomarker source, notably protein content. Ewing sarcoma-derived tiny EVs may be biomarked by CD99, HINT1, and NGFR membrane proteins according to Samuel et al. They used these EV surface proteins to immuno-enrich Ewing sarcoma-associated tiny EVs and identify EWS-FLI1 and EWS-ERG fusion transcripts in plasma from localized and metastatic patients [66].

5. Clinical Implication of Liquid Biopsy in Monitoring Drug Resistance

5.1. Liquid Biopsy in Chemoresistant Primary and Secondary Bone Tumors

Recent research indicates that the tumor secretome, including DNA fragments from drug-resistant cells with mutations, is abundant in plasma, making blood-based liquid biopsy crucial [67]. Recent studies on plasma samples of small patient cohorts have identified resistance mutations during treatment. While the data is clinically informative about the therapy response, it has not yet been fully validated in clinical practice. Quantification and analysis of ctDNA are effective tools for analyzing various tumor types [68]. The potential of ctDNA for monitoring treatment efficacy can be demonstrated in the finding that breast cancer patients with metastases, treated with aromatase inhibitors, and carrying ESR1 mutations in ctDNA are likely to show resistance to endocrine therapy and experience shorter progression-free survival [69].
Liquid biopsy can also identify biomarkers linked to CDK inhibitor resistance and predict metastatic disease in advanced breast cancer patients with hormone receptor-positive/HER2-. Patients receiving CDK inhibitor and endocrine therapy show specific ctDNA mutations, such as retinoblastoma, ESR1, fibroblast growth factor receptor 1, or phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha alterations [70][71][72], potentially influencing disease outcomes and therapeutic decisions. Additionally, detecting and quantifying CTC acquired resistance may serve as a predictive marker for treatment outcomes [73]. In castration-resistant prostate cancer patients treated with docetaxel, CTC count in blood is a reliable indication of therapy sensitivity and survival [74].
Osteosarcoma treatment difficulty stems from genetic instability and the emergence of chemotherapy resistance after selection pressure; low levels of miR-375 in osteosarcoma patients have been associated with poor response to preoperative chemotherapy [75]. Recent research has linked tumor-associated miRNAs to osteosarcoma chemoresistance, including miR-491, which is decreased in serum from patients compared to that in healthy controls. This decrease is linked to increased metastasis, poor chemoresponse, and lower survival rates [76]. Serum miR-21 levels are considerably greater in osteosarcoma patients compared to those in controls and are associated with advanced Enneking stage and chemotherapy resistance [77]. Reduced miR-125b levels in Ewing sarcoma patients are linked to poor treatment response and chemoresistance development [65].

5.2. Implication of Extracellular Vesicles in Chemoresistance

EVs play a crucial role in drug resistance transmission [78], making them valuable for monitoring its emergence during therapy. miRNAs may represent indicators for chemoresistance. High quantities of membrane transporter pump P-glycoprotein in EVs from doxorubicin-resistant osteosarcoma MG63 cells facilitate horizontal transmission of resistance to susceptible cancer cells [79]. Additionally, miR-25-3p overexpression in osteosarcoma patients’ blood has been linked to tumor development and medication resistance [80]. The levels of miR-222-3p in EVs derived from NSCLC patients’ blood may predict sensitivity to gemcitabine and identify individuals with advanced and resistant illness [81]. In addition, platinum-resistant NSCLC patients have increased EV-derived miR-425-3p in their blood compared to that of platinum-sensitive individuals [82]. Research indicates that miR-222 from doxorubicin-resistant breast cancer cells is transported by EVs to M2 macrophages, inducing polarization. In contrast, miR-222 overexpression suppresses the expression of the tensin homolog gene and phosphatase activity, leading to Akt phosphorylation and activation, which promotes the proliferation of cancer cells, as well as their migration and invasion through positive feedback. EVs from the plasma of chemoresistant breast cancer patients show increased levels of miRNA-222 [83], while paclitaxel-treated cells from the human osteotropic breast cancer cell line MDA-MB-231 release EVs enriched in Survivin [84]. Unexpectedly, EVs can directly inhibit anti-neoplastic drugs; EVs from HER2-positive breast cancer patients behave as decoy receptors for trastuzumab, affecting its activity [85].
EVs generated by cancer cells include HER2 on their surface that is bound by trastuzumab systemically, reducing the quantity of antibodies available for cell binding. Yang et al. identified increased GSTP1 mRNA levels in EVs from non-responding breast cancer patients treated with neoadjuvant chemotherapy compared to those of responders. GSTP1-containing EVs have been shown to transmit drug resistance horizontally, suggesting their potential as negative predictors of chemoresistance and clinical outcomes in breast cancer patients receiving anthracycline/taxane treatment [86].
Transient receptor potential channel 5 mRNA in EVs isolated from the blood of metastatic breast cancer patients may predict chemoresistance [87]. Kharaziha et al. [88] identified MDR-1, MDR-3, endophilin-A2, and poly(A) binding protein 4 as enriched proteins in EVs from both prostate cancer cells resistant to docetaxel and castration-resistant prostate cancer patient serum, suggesting potential as biomarkers for therapeutic response or resistance [88]. The field is promising in cancer research, although larger longitudinal studies are necessary to confirm the effect of biomarkers.

5.3. Factors Hindering the Clinical Applications of Liquid Biopsies

Despite the potential of liquid biopsies, difficulties must be solved before broad clinical use. Due to the sensitivity of the approaches, even little variations in sample collection or processing may significantly impact the final output. The use of serum instead of plasma may increase cell-free DNA from other sources, lowering the diagnostic power of NGS-based tests, particularly for uncommon variations [89]. Lifestyle variables may impact cell-free DNA release in the bloodstream, creating a complex set of confounding factors that are challenging to detect and define [90]. CTCs are uncommon and difficult to acquire, and although the CellSearch technique offers a uniform approach, it is limited in its viability. They are only suitable for DNA and FACS/Immunofluorescence investigations, not RNA-based or functional experiments such as patient-derived xenografts or in vitro drug sensitivity testing [91][92]. CTC analysis also has limitations similar to those of conventional biopsies, since it may not reflect the complete tumor, but rather a subset of cells that survived in the circulation. An experiment is underway to partly address this problem by selecting various blood collection locations. Current research suggests that arterial blood and blood from near the main tumor may provide more CTCs [93][94].
Despite efforts to develop guidelines for sample treatment, liquid biopsy requires training, specialized facilities, and expertise in the interpretation of results [95]. EVs have unique preanalytical obstacles, in addition to the broader issues mentioned above. They represent a new source of biological information; however, further research is needed to develop EVs as a liquid biopsy. EV isolation is a prime example, as several methods have been investigated for isolating EVs [96]. Unfortunately, there is no perfect strategy for EV separation, and findings may vary based on the investigator’s approach [96]. Additionally, lifestyle variables may also promote EV release, making tumor-specific exosome identification difficult [19].

References

  1. Pohlig, F.; Kirchhoff, C.; Lenze, U.; Schauwecker, J.; Burgkart, R.; Rechl, H.; von Eisenhart-Rothe, R. Percutaneous Core Needle Biopsy versus Open Biopsy in Diagnostics of Bone and Soft Tissue Sarcoma: A Retrospective Study. Eur. J. Med. Res. 2012, 17, 29.
  2. Le, H.B.Q.; Lee, S.T.; Munk, P.L. Image-Guided Musculoskeletal Biopsies. Semin. Intervent. Radiol. 2010, 27, 191–198.
  3. Eslami-S, Z.; Cortés-Hernández, L.E.; Alix-Panabières, C. The Metastatic Cascade as the Basis for Liquid Biopsy Development. Front. Oncol. 2020, 10, 1055.
  4. Palmirotta, R.; Lovero, D.; Cafforio, P.; Felici, C.; Mannavola, F.; Pellè, E.; Quaresmini, D.; Tucci, M.; Silvestris, F. Liquid Biopsy of Cancer: A Multimodal Diagnostic Tool in Clinical Oncology. Ther. Adv. Med. Oncol. 2018, 10, 1758835918794630.
  5. Li, X.; Seebacher, N.A.; Hornicek, F.J.; Xiao, T.; Duan, Z. Application of Liquid Biopsy in Bone and Soft Tissue Sarcomas: Present and Future. Cancer Lett. 2018, 439, 66–77.
  6. Perakis, S.; Speicher, M.R. Emerging Concepts in Liquid Biopsies. BMC Med. 2017, 15, 75.
  7. de Wit, S.; van Dalum, G.; Terstappen, L.W.M.M. Detection of Circulating Tumor Cells. Scientifica 2014, 2014, 819362.
  8. Yang, C.; Xia, B.-R.; Jin, W.-L.; Lou, G. Circulating Tumor Cells in Precision Oncology: Clinical Applications in Liquid Biopsy and 3D Organoid Model. Cancer Cell Int. 2019, 19, 341.
  9. Mauri, G.; Bonazzina, E.; Amatu, A.; Tosi, F.; Bencardino, K.; Gori, V.; Massihnia, D.; Cipani, T.; Spina, F.; Ghezzi, S.; et al. The Evolutionary Landscape of Treatment for BRAFV600E Mutant Metastatic Colorectal Cancer. Cancers 2021, 13, 137.
  10. Ascierto, P.A.; Kirkwood, J.M.; Grob, J.-J.; Simeone, E.; Grimaldi, A.M.; Maio, M.; Palmieri, G.; Testori, A.; Marincola, F.M.; Mozzillo, N. The Role of BRAF V600 Mutation in Melanoma. J. Transl. Med. 2012, 10, 85.
  11. Pellegrini, C.; Di Nardo, L.; Cipolloni, G.; Martorelli, C.; De Padova, M.; Antonini, A.; Maturo, M.G.; Del Regno, L.; Strafella, S.; Micantonio, T.; et al. Heterogeneity of BRAF, NRAS, and TERT Promoter Mutational Status in Multiple Melanomas and Association with MC1R Genotype: Findings from Molecular and Immunohistochemical Analysis. J. Mol. Diagn. 2018, 20, 110–122.
  12. Rashid, F.A.; Bhat, G.H.; Khan, M.S.; Tabassum, S.; Bhat, M.H. Variations in MAP Kinase Gladiators and Risk of Differentiated Thyroid Carcinoma. Mol. Clin. Oncol. 2022, 16, 45.
  13. Crowley, E.; Di Nicolantonio, F.; Loupakis, F.; Bardelli, A. Liquid Biopsy: Monitoring Cancer-Genetics in the Blood. Nat. Rev. Clin. Oncol. 2013, 10, 472–484.
  14. Colombo, M.; Raposo, G.; Théry, C. Biogenesis, Secretion, and Intercellular Interactions of Exosomes and Other Extracellular Vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289.
  15. Doyle, L.M.; Wang, M.Z. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727.
  16. Stevic, I.; Buescher, G.; Ricklefs, F.L. Monitoring Therapy Efficiency in Cancer through Extracellular Vesicles. Cells 2020, 9, 130.
  17. Green, T.M.; Alpaugh, M.L.; Barsky, S.H.; Rappa, G.; Lorico, A. Breast Cancer-Derived Extracellular Vesicles: Characterization and Contribution to the Metastatic Phenotype. Biomed. Res. Int. 2015, 2015, 634865.
  18. Taverna, S.; Giusti, I.; D’Ascenzo, S.; Pizzorno, L.; Dolo, V. Breast Cancer Derived Extracellular Vesicles in Bone Metastasis Induction and Their Clinical Implications as Biomarkers. Int. J. Mol. Sci. 2020, 21, 3573.
  19. Ludwig, N.; Whiteside, T.L.; Reichert, T.E. Challenges in Exosome Isolation and Analysis in Health and Disease. Int. J. Mol. Sci. 2019, 20, 4684.
  20. Kim, K.M.; Abdelmohsen, K.; Mustapic, M.; Kapogiannis, D.; Gorospe, M. RNA in Extracellular Vesicles. Wiley Interdiscip. Rev. RNA 2017, 8, e1413.
  21. Siravegna, G.; Marsoni, S.; Siena, S.; Bardelli, A. Integrating Liquid Biopsies into the Management of Cancer. Nat. Rev. Clin. Oncol. 2017, 14, 531–548.
  22. Incorvaia, L.; Castiglia, M.; Perez, A.; Massihnia, D.; Caruso, S.; Altintas, S.; Calò, V.; Russo, A. Liquid Biopsy in Breast Cancer. In Liquid Biopsy in Cancer Patients: The Hand Lens for Tumor Evolution; Russo, A., Giordano, A., Rolfo, C., Eds.; Current Clinical Pathology; Springer International Publishing: Cham, Switzerland, 2017; pp. 77–84.
  23. Garcia-Murillas, I.; Schiavon, G.; Weigelt, B.; Ng, C.; Hrebien, S.; Cutts, R.J.; Cheang, M.; Osin, P.; Nerurkar, A.; Kozarewa, I.; et al. Mutation Tracking in Circulating Tumor DNA Predicts Relapse in Early Breast Cancer. Sci. Transl. Med. 2015, 7, 302ra133.
  24. Beaver, J.A.; Jelovac, D.; Balukrishna, S.; Cochran, R.; Croessmann, S.; Zabransky, D.J.; Wong, H.Y.; Toro, P.V.; Cidado, J.; Blair, B.G.; et al. Detection of Cancer DNA in Plasma of Patients with Early-Stage Breast Cancer. Clin. Cancer Res. 2014, 20, 2643–2650.
  25. Pécuchet, N.; Zonta, E.; Didelot, A.; Combe, P.; Thibault, C.; Gibault, L.; Lours, C.; Rozenholc, Y.; Taly, V.; Laurent-Puig, P.; et al. Base-Position Error Rate Analysis of Next-Generation Sequencing Applied to Circulating Tumor DNA in Non-Small Cell Lung Cancer: A Prospective Study. PLoS Med. 2016, 13, e1002199.
  26. Jia, J.; Huang, B.; Zhuang, Z.; Chen, S. Circulating Tumor DNA as Prognostic Markers for Late Stage NSCLC with Bone Metastasis. Int. J. Biol. Markers 2018, 33, 222–230.
  27. Vandekerkhove, G.; Struss, W.J.; Annala, M.; Kallio, H.M.L.; Khalaf, D.; Warner, E.W.; Herberts, C.; Ritch, E.; Beja, K.; Loktionova, Y.; et al. Circulating Tumor DNA Abundance and Potential Utility in De Novo Metastatic Prostate Cancer. Eur. Urol. 2019, 75, 667–675.
  28. Bidard, F.-C.; Peeters, D.J.; Fehm, T.; Nolé, F.; Gisbert-Criado, R.; Mavroudis, D.; Grisanti, S.; Generali, D.; Garcia-Saenz, J.A.; Stebbing, J.; et al. Clinical Validity of Circulating Tumour Cells in Patients with Metastatic Breast Cancer: A Pooled Analysis of Individual Patient Data. Lancet Oncol. 2014, 15, 406–414.
  29. Moussavi-Harami, S.F.; Wisinski, K.B.; Beebe, D.J. Circulating Tumor Cells in Metastatic Breast Cancer: A Prognostic and Predictive Marker. J. Patient Cent. Res. Rev. 2014, 1, 85–92.
  30. De Giorgi, U.; Valero, V.; Rohren, E.; Mego, M.; Doyle, G.V.; Miller, M.C.; Ueno, N.T.; Handy, B.C.; Reuben, J.M.; Macapinlac, H.A.; et al. Circulating Tumor Cells and Bone Metastases as Detected by FDG-PET/CT in Patients with Metastatic Breast Cancer. Ann. Oncol. 2010, 21, 33–39.
  31. Baccelli, I.; Schneeweiss, A.; Riethdorf, S.; Stenzinger, A.; Schillert, A.; Vogel, V.; Klein, C.; Saini, M.; Bäuerle, T.; Wallwiener, M.; et al. Identification of a Population of Blood Circulating Tumor Cells from Breast Cancer Patients That Initiates Metastasis in a Xenograft Assay. Nat. Biotechnol. 2013, 31, 539–544.
  32. Cheng, M.; Liu, L.; Yang, H.-S.; Liu, G.-F. Circulating Tumor Cells Are Associated with Bone Metastasis of Lung Cancer. Asian Pac. J. Cancer Prev. 2014, 15, 6369–6374.
  33. Krebs, M.G.; Sloane, R.; Priest, L.; Lancashire, L.; Hou, J.-M.; Greystoke, A.; Ward, T.H.; Ferraldeschi, R.; Hughes, A.; Clack, G.; et al. Evaluation and Prognostic Significance of Circulating Tumor Cells in Patients with Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2011, 29, 1556–1563.
  34. Shaffer, D.R.; Leversha, M.A.; Danila, D.C.; Lin, O.; Gonzalez-Espinoza, R.; Gu, B.; Anand, A.; Smith, K.; Maslak, P.; Doyle, G.V.; et al. Circulating Tumor Cell Analysis in Patients with Progressive Castration-Resistant Prostate Cancer. Clin. Cancer Res. 2007, 13, 2023–2029.
  35. de Bono, J.S.; Scher, H.I.; Montgomery, R.B.; Parker, C.; Miller, M.C.; Tissing, H.; Doyle, G.V.; Terstappen, L.W.W.M.; Pienta, K.J.; Raghavan, D. Circulating Tumor Cells Predict Survival Benefit from Treatment in Metastatic Castration-Resistant Prostate Cancer. Clin. Cancer Res. 2008, 14, 6302–6309.
  36. Helo, P.; Cronin, A.M.; Danila, D.C.; Wenske, S.; Gonzalez-Espinoza, R.; Anand, A.; Koscuiszka, M.; Väänänen, R.-M.; Pettersson, K.; Chun, F.K.-H.; et al. Circulating Prostate Tumor Cells Detected by Reverse Transcription-PCR in Men with Localized or Castration-Refractory Prostate Cancer: Concordance with CellSearch Assay and Association with Bone Metastases and with Survival. Clin. Chem. 2009, 55, 765–773.
  37. Bortolini Silveira, A.; Bidard, F.-C.; Tanguy, M.-L.; Girard, E.; Trédan, O.; Dubot, C.; Jacot, W.; Goncalves, A.; Debled, M.; Levy, C.; et al. Multimodal Liquid Biopsy for Early Monitoring and Outcome Prediction of Chemotherapy in Metastatic Breast Cancer. NPJ Breast Cancer 2021, 7, 115.
  38. Kodahl, A.R.; Ehmsen, S.; Pallisgaard, N.; Jylling, A.M.B.; Jensen, J.D.; Laenkholm, A.-V.; Knoop, A.S.; Ditzel, H.J. Correlation between Circulating Cell-Free PIK3CA Tumor DNA Levels and Treatment Response in Patients with PIK3CA-Mutated Metastatic Breast Cancer. Mol. Oncol. 2018, 12, 925–935.
  39. Dawson, S.-J.; Tsui, D.W.Y.; Murtaza, M.; Biggs, H.; Rueda, O.M.; Chin, S.-F.; Dunning, M.J.; Gale, D.; Forshew, T.; Mahler-Araujo, B.; et al. Analysis of Circulating Tumor DNA to Monitor Metastatic Breast Cancer. N. Engl. J. Med. 2013, 368, 1199–1209.
  40. Wang, P.; Bahreini, A.; Gyanchandani, R.; Lucas, P.C.; Hartmaier, R.J.; Watters, R.J.; Jonnalagadda, A.R.; Trejo Bittar, H.E.; Berg, A.; Hamilton, R.L.; et al. Sensitive Detection of Mono-and Polyclonal ESR1 Mutations in Primary Tumors, Metastatic Lesions, and Cell-Free DNA of Breast Cancer Patients. Clin. Cancer Res. 2016, 22, 1130–1137.
  41. Rossi, G.; Mu, Z.; Rademaker, A.W.; Austin, L.K.; Strickland, K.S.; Costa, R.L.B.; Nagy, R.J.; Zagonel, V.; Taxter, T.J.; Behdad, A.; et al. Cell-Free DNA and Circulating Tumor Cells: Comprehensive Liquid Biopsy Analysis in Advanced Breast Cancer. Clin. Cancer Res. 2018, 24, 560–568.
  42. Bhadresha, K.P.; Patel, M.; Jain, N.K.; Rawal, R.M. A Predictive Biomarker Panel for Bone Metastases: Liquid Biopsy Approach. J. Bone Oncol. 2021, 29, 100374.
  43. Yang, X.-R.; Pi, C.; Yu, R.; Fan, X.-J.; Peng, X.-X.; Zhang, X.-C.; Chen, Z.-H.; Wu, X.; Shao, Y.; Wu, Y.-L.; et al. Correlation of Exosomal microRNA Clusters with Bone Metastasis in Non-Small Cell Lung Cancer. Clin. Exp. Metastasis 2021, 38, 109–117.
  44. Bryant, R.J.; Pawlowski, T.; Catto, J.W.F.; Marsden, G.; Vessella, R.L.; Rhees, B.; Kuslich, C.; Visakorpi, T.; Hamdy, F.C. Changes in Circulating microRNA Levels Associated with Prostate Cancer. Br. J. Cancer 2012, 106, 768–774.
  45. Biggs, C.N.; Siddiqui, K.M.; Al-Zahrani, A.A.; Pardhan, S.; Brett, S.I.; Guo, Q.Q.; Yang, J.; Wolf, P.; Power, N.E.; Durfee, P.N.; et al. Prostate Extracellular Vesicles in Patient Plasma as a Liquid Biopsy Platform for Prostate Cancer Using Nanoscale Flow Cytometry. Oncotarget 2016, 7, 8839–8849.
  46. McKiernan, J.; Donovan, M.J.; Margolis, E.; Partin, A.; Carter, B.; Brown, G.; Torkler, P.; Noerholm, M.; Skog, J.; Shore, N.; et al. A Prospective Adaptive Utility Trial to Validate Performance of a Novel Urine Exosome Gene Expression Assay to Predict High-Grade Prostate Cancer in Patients with Prostate-Specific Antigen 2-10ng/Ml at Initial Biopsy. Eur. Urol. 2018, 74, 731–738.
  47. Barris, D.M.; Weiner, S.B.; Dubin, R.A.; Fremed, M.; Zhang, X.; Piperdi, S.; Zhang, W.; Maqbool, S.; Gill, J.; Roth, M.; et al. Detection of Circulating Tumor DNA in Patients with Osteosarcoma. Oncotarget 2018, 9, 12695–12704.
  48. Shulman, D.S.; Klega, K.; Imamovic-Tuco, A.; Clapp, A.; Nag, A.; Thorner, A.R.; Van Allen, E.; Ha, G.; Lessnick, S.L.; Gorlick, R.; et al. Detection of Circulating Tumour DNA Is Associated with Inferior Outcomes in Ewing Sarcoma and Osteosarcoma: A Report from the Children’s Oncology Group. Br. J. Cancer 2018, 119, 615–621.
  49. Shukla, N.N.; Patel, J.A.; Magnan, H.; Zehir, A.; You, D.; Tang, J.; Meng, F.; Samoila, A.; Slotkin, E.K.; Ambati, S.R.; et al. Plasma DNA-Based Molecular Diagnosis, Prognostication, and Monitoring of Patients with EWSR1 Fusion-Positive Sarcomas. JCO Precis. Oncol. 2017, 1, 1–11.
  50. Hayashi, M.; Chu, D.; Meyer, C.F.; Llosa, N.J.; McCarty, G.; Morris, C.D.; Levin, A.S.; Wolinsky, J.-P.; Albert, C.M.; Steppan, D.A.; et al. Highly Personalized Detection of Minimal Ewing Sarcoma Disease Burden from Plasma Tumor DNA. Cancer 2016, 122, 3015–3023.
  51. Zhang, H.; Gao, P.; Xiao, X.; Heger, M.; Geng, L.; Fan, B.; Yuan, Y.; Huang, C.; Chen, G.; Liu, Y.; et al. A Liquid Biopsy-Based Method for the Detection and Quantification of Circulating Tumor Cells in Surgical Osteosarcoma Patients. Int. J. Oncol. 2017, 50, 1075–1086.
  52. Li, M.; Lu, Y.; Long, Z.; Li, M.; Kong, J.; Chen, G.; Wang, Z. Prognostic and Clinicopathological Significance of Circulating Tumor Cells in Osteosarcoma. J. Bone Oncol. 2019, 16, 100236.
  53. Wu, Z.-J.; Tan, J.-C.; Qin, X.; Liu, B.; Yuan, Z.-C. Significance of Circulating Tumor Cells in Osteosarcoma Patients Treated by Neoadjuvant Chemotherapy and Surgery. Cancer Manag. Res. 2018, 10, 3333–3339.
  54. Chalopin, A.; Tellez-Gabriel, M.; Brown, H.K.; Vallette, F.; Heymann, M.-F.; Gouin, F.; Heymann, D. Isolation of Circulating Tumor Cells in a Preclinical Model of Osteosarcoma: Effect of Chemotherapy. J. Bone Oncol. 2018, 12, 83–90.
  55. Benini, S.; Gamberi, G.; Cocchi, S.; Garbetta, J.; Alberti, L.; Righi, A.; Gambarotti, M.; Picci, P.; Ferrari, S. Detection of Circulating Tumor Cells in Liquid Biopsy from Ewing Sarcoma Patients. Cancer Manag. Res. 2018, 10, 49–60.
  56. Schleiermacher, G.; Peter, M.; Oberlin, O.; Philip, T.; Rubie, H.; Mechinaud, F.; Sommelet-Olive, D.; Landman-Parker, J.; Bours, D.; Michon, J.; et al. Increased Risk of Systemic Relapses Associated with Bone Marrow Micrometastasis and Circulating Tumor Cells in Localized Ewing Tumor. J. Clin. Oncol. 2003, 21, 85–91.
  57. Hayashi, M.; Zhu, P.; McCarty, G.; Meyer, C.F.; Pratilas, C.A.; Levin, A.; Morris, C.D.; Albert, C.M.; Jackson, K.W.; Tang, C.-M.; et al. Size-Based Detection of Sarcoma Circulating Tumor Cells and Cell Clusters. Oncotarget 2017, 8, 78965–78977.
  58. Bao, Q.; Gong, L.; Wang, J.; Wen, J.; Shen, Y.; Zhang, W. Extracellular Vesicle RNA Sequencing Reveals Dramatic Transcriptomic Alterations Between Metastatic and Primary Osteosarcoma in a Liquid Biopsy Approach. Ann. Surg. Oncol. 2018, 25, 2642–2651.
  59. Ma, W.; Zhang, X.; Chai, J.; Chen, P.; Ren, P.; Gong, M. Circulating miR-148a Is a Significant Diagnostic and Prognostic Biomarker for Patients with Osteosarcoma. Tumor Biol. 2014, 35, 12467–12472.
  60. Wang, T.; Ji, F.; Dai, Z.; Xie, Y.; Yuan, D. Increased Expression of microRNA-191 as a Potential Serum Biomarker for Diagnosis and Prognosis in Human Osteosarcoma. Cancer Biomark. 2015, 15, 543–550.
  61. Zhou, S.; Wang, B.; Hu, J.; Zhou, Y.; Jiang, M.; Wu, M.; Qin, L.; Yang, X. miR-421 Is a Diagnostic and Prognostic Marker in Patients with Osteosarcoma. Tumor Biol. 2016, 37, 9001–9007.
  62. Cong, C.; Wang, W.; Tian, J.; Gao, T.; Zheng, W.; Zhou, C. Identification of Serum miR-124 as a Biomarker for Diagnosis and Prognosis in Osteosarcoma. Cancer Biomark. 2018, 21, 449–454.
  63. Yao, Z.-S.; Li, C.; Liang, D.; Jiang, X.-B.; Tang, J.-J.; Ye, L.-Q.; Yuan, K.; Ren, H.; Yang, Z.-D.; Jin, D.-X.; et al. Diagnostic and Prognostic Implications of Serum miR-101 in Osteosarcoma. Cancer Biomark. 2018, 22, 127–133.
  64. Cai, H.; Zhao, H.; Tang, J.; Wu, H. Serum miR-195 Is a Diagnostic and Prognostic Marker for Osteosarcoma. J. Surg. Res. 2015, 194, 505–510.
  65. Nie, C.L.; Ren, W.H.; Ma, Y.; Xi, J.S.; Han, B. Circulating miR-125b as a Biomarker of Ewing’s Sarcoma in Chinese Children. Genet. Mol. Res. 2015, 14, 19049–19056.
  66. Samuel, G.; Crow, J.; Klein, J.B.; Merchant, M.L.; Nissen, E.; Koestler, D.C.; Laurence, K.; Liang, X.; Neville, K.; Staggs, V.; et al. Ewing Sarcoma Family of Tumors-Derived Small Extracellular Vesicle Proteomics Identify Potential Clinical Biomarkers. Oncotarget 2020, 11, 2995–3012.
  67. Russano, M.; Napolitano, A.; Ribelli, G.; Iuliani, M.; Simonetti, S.; Citarella, F.; Pantano, F.; Dell’Aquila, E.; Anesi, C.; Silvestris, N.; et al. Liquid Biopsy and Tumor Heterogeneity in Metastatic Solid Tumors: The Potentiality of Blood Samples. J. Exp. Clin. Cancer Res. 2020, 39, 95.
  68. Bettegowda, C.; Sausen, M.; Leary, R.J.; Kinde, I.; Wang, Y.; Agrawal, N.; Bartlett, B.R.; Wang, H.; Luber, B.; Alani, R.M.; et al. Detection of Circulating Tumor DNA in Early- and Late-Stage Human Malignancies. Sci. Transl. Med. 2014, 6, 224ra24.
  69. Schiavon, G.; Hrebien, S.; Garcia-Murillas, I.; Cutts, R.J.; Pearson, A.; Tarazona, N.; Fenwick, K.; Kozarewa, I.; Lopez-Knowles, E.; Ribas, R.; et al. Analysis of ESR1 Mutation in Circulating Tumor DNA Demonstrates Evolution during Therapy for Metastatic Breast Cancer. Sci. Transl. Med. 2015, 7, 313ra182.
  70. O’Leary, B.; Hrebien, S.; Morden, J.P.; Beaney, M.; Fribbens, C.; Huang, X.; Liu, Y.; Bartlett, C.H.; Koehler, M.; Cristofanilli, M.; et al. Early Circulating Tumor DNA Dynamics and Clonal Selection with Palbociclib and Fulvestrant for Breast Cancer. Nat. Commun. 2018, 9, 896.
  71. O’Leary, B.; Cutts, R.J.; Liu, Y.; Hrebien, S.; Huang, X.; Fenwick, K.; André, F.; Loibl, S.; Loi, S.; Garcia-Murillas, I.; et al. The Genetic Landscape and Clonal Evolution of Breast Cancer Resistance to Palbociclib plus Fulvestrant in the PALOMA-3 Trial. Cancer Discov. 2018, 8, 1390–1403.
  72. Fribbens, C.; O’Leary, B.; Kilburn, L.; Hrebien, S.; Garcia-Murillas, I.; Beaney, M.; Cristofanilli, M.; Andre, F.; Loi, S.; Loibl, S.; et al. Plasma ESR1 Mutations and the Treatment of Estrogen Receptor-Positive Advanced Breast Cancer. J. Clin. Oncol. 2016, 34, 2961–2968.
  73. Galardi, F.; De Luca, F.; Biagioni, C.; Migliaccio, I.; Curigliano, G.; Minisini, A.M.; Bonechi, M.; Moretti, E.; Risi, E.; McCartney, A.; et al. Circulating Tumor Cells and Palbociclib Treatment in Patients with ER-Positive, HER2-Negative Advanced Breast Cancer: Results from a Translational Sub-Study of the TREnd Trial. Breast Cancer Res. 2021, 23, 38.
  74. Okegawa, T.; Itaya, N.; Hara, H.; Tambo, M.; Nutahara, K. Circulating Tumor Cells as a Biomarker Predictive of Sensitivity to Docetaxel Chemotherapy in Patients with Castration-Resistant Prostate Cancer. Anticancer Res. 2014, 34, 6705–6710.
  75. Liu, W.; Zhao, X.; Zhang, Y.-J.; Fang, G.-W.; Xue, Y. MicroRNA-375 as a Potential Serum Biomarker for the Diagnosis, Prognosis, and Chemosensitivity Prediction of Osteosarcoma. J. Int. Med. Res. 2018, 46, 975–983.
  76. Wang, S.-N.; Luo, S.; Liu, C.; Piao, Z.; Gou, W.; Wang, Y.; Guan, W.; Li, Q.; Zou, H.; Yang, Z.-Z.; et al. miR-491 Inhibits Osteosarcoma Lung Metastasis and Chemoresistance by Targeting αB-Crystallin. Mol. Ther. 2017, 25, 2140–2149.
  77. Yuan, J.; Chen, L.; Chen, X.; Sun, W.; Zhou, X. Identification of Serum microRNA-21 as a Biomarker for Chemosensitivity and Prognosis in Human Osteosarcoma. J. Int. Med. Res. 2012, 40, 2090–2097.
  78. Xavier, C.P.R.; Caires, H.R.; Barbosa, M.A.G.; Bergantim, R.; Guimarães, J.E.; Vasconcelos, M.H. The Role of Extracellular Vesicles in the Hallmarks of Cancer and Drug Resistance. Cells 2020, 9, 1141.
  79. Torreggiani, E.; Roncuzzi, L.; Perut, F.; Zini, N.; Baldini, N. Multimodal Transfer of MDR by Exosomes in Human Osteosarcoma. Int. J. Oncol. 2016, 49, 189–196.
  80. Yoshida, A.; Fujiwara, T.; Uotani, K.; Morita, T.; Kiyono, M.; Yokoo, S.; Hasei, J.; Nakata, E.; Kunisada, T.; Ozaki, T. Clinical and Functional Significance of Intracellular and Extracellular microRNA-25-3p in Osteosarcoma. Acta Med. Okayama 2018, 72, 165–174.
  81. Wei, F.; Ma, C.; Zhou, T.; Dong, X.; Luo, Q.; Geng, L.; Ding, L.; Zhang, Y.; Zhang, L.; Li, N.; et al. Exosomes Derived from Gemcitabine-Resistant Cells Transfer Malignant Phenotypic Traits via Delivery of miRNA-222-3p. Mol. Cancer 2017, 16, 132.
  82. Yuwen, D.; Ma, Y.; Wang, D.; Gao, J.; Li, X.; Xue, W.; Fan, M.; Xu, Q.; Shen, Y.; Shu, Y. Prognostic Role of Circulating Exosomal miR-425-3p for the Response of NSCLC to Platinum-Based Chemotherapy. Cancer Epidemiol. Biomarkers Prev. 2019, 28, 163–173.
  83. Chen, W.-X.; Wang, D.-D.; Zhu, B.; Zhu, Y.-Z.; Zheng, L.; Feng, Z.-Q.; Qin, X.-H. Exosomal miR-222 from Adriamycin-Resistant MCF-7 Breast Cancer Cells Promote Macrophages M2 Polarization via PTEN/Akt to Induce Tumor Progression. Aging 2021, 13, 10415–10430.
  84. Kreger, B.T.; Johansen, E.R.; Cerione, R.A.; Antonyak, M.A. The Enrichment of Survivin in Exosomes from Breast Cancer Cells Treated with Paclitaxel Promotes Cell Survival and Chemoresistance. Cancers 2016, 8, 111.
  85. Ciravolo, V.; Huber, V.; Ghedini, G.C.; Venturelli, E.; Bianchi, F.; Campiglio, M.; Morelli, D.; Villa, A.; Della Mina, P.; Menard, S.; et al. Potential Role of HER2-Overexpressing Exosomes in Countering Trastuzumab-Based Therapy. J. Cell Physiol. 2012, 227, 658–667.
  86. Yang, S.-J.; Wang, D.-D.; Li, J.; Xu, H.-Z.; Shen, H.-Y.; Chen, X.; Zhou, S.-Y.; Zhong, S.-L.; Zhao, J.-H.; Tang, J.-H. Predictive Role of GSTP1-Containing Exosomes in Chemotherapy-Resistant Breast Cancer. Gene 2017, 623, 5–14.
  87. Wang, T.; Ning, K.; Lu, T.-X.; Sun, X.; Jin, L.; Qi, X.; Jin, J.; Hua, D. Increasing Circulating Exosomes-Carrying TRPC5 Predicts Chemoresistance in Metastatic Breast Cancer Patients. Cancer Sci. 2017, 108, 448–454.
  88. Kharaziha, P.; Chioureas, D.; Rutishauser, D.; Baltatzis, G.; Lennartsson, L.; Fonseca, P.; Azimi, A.; Hultenby, K.; Zubarev, R.; Ullén, A.; et al. Molecular Profiling of Prostate Cancer Derived Exosomes May Reveal a Predictive Signature for Response to Docetaxel. Oncotarget 2015, 6, 21740–21754.
  89. Pittella-Silva, F.; Chin, Y.M.; Chan, H.T.; Nagayama, S.; Miyauchi, E.; Low, S.-K.; Nakamura, Y. Plasma or Serum: Which Is Preferable for Mutation Detection in Liquid Biopsy? Clin. Chem. 2020, 66, 946–957.
  90. Merker, J.D.; Oxnard, G.R.; Compton, C.; Diehn, M.; Hurley, P.; Lazar, A.J.; Lindeman, N.; Lockwood, C.M.; Rai, A.J.; Schilsky, R.L.; et al. Circulating Tumor DNA Analysis in Patients with Cancer: American Society of Clinical Oncology and College of American Pathologists Joint Review. J. Clin. Oncol. 2018, 36, 1631–1641.
  91. Wong, K.H.K.; Tessier, S.N.; Miyamoto, D.T.; Miller, K.L.; Bookstaver, L.D.; Carey, T.R.; Stannard, C.J.; Thapar, V.; Tai, E.C.; Vo, K.D.; et al. Whole Blood Stabilization for the Microfluidic Isolation and Molecular Characterization of Circulating Tumor Cells. Nat. Commun. 2017, 8, 1733.
  92. Fehm, T.N.; Meier-Stiegen, F.; Driemel, C.; Jäger, B.; Reinhardt, F.; Naskou, J.; Franken, A.; Neubauer, H.; Neves, R.P.L.; van Dalum, G.; et al. Diagnostic Leukapheresis for CTC Analysis in Breast Cancer Patients: CTC Frequency, Clinical Experiences and Recommendations for Standardized Reporting. Cytom. Part A 2018, 93, 1213–1219.
  93. Terai, M.; Mu, Z.; Eschelman, D.J.; Gonsalves, C.F.; Kageyama, K.; Chervoneva, I.; Orloff, M.; Weight, R.; Mastrangelo, M.J.; Cristofanilli, M.; et al. Arterial Blood, Rather Than Venous Blood, is a Better Source for Circulating Melanoma Cells. EBioMedicine 2015, 2, 1821–1826.
  94. Buscail, E.; Chiche, L.; Laurent, C.; Vendrely, V.; Denost, Q.; Denis, J.; Thumerel, M.; Lacorte, J.-M.; Bedel, A.; Moreau-Gaudry, F.; et al. Tumor-Proximal Liquid Biopsy to Improve Diagnostic and Prognostic Performances of Circulating Tumor Cells. Mol. Oncol. 2019, 13, 1811–1826.
  95. Heidrich, I.; Ačkar, L.; Mossahebi Mohammadi, P.; Pantel, K. Liquid Biopsies: Potential and Challenges. Int. J. Cancer 2021, 148, 528–545.
  96. Brennan, K.; Martin, K.; FitzGerald, S.P.; O’Sullivan, J.; Wu, Y.; Blanco, A.; Richardson, C.; Mc Gee, M.M. A Comparison of Methods for the Isolation and Separation of Extracellular Vesicles from Protein and Lipid Particles in Human Serum. Sci. Rep. 2020, 10, 1039.
More
Information
Subjects: Orthopedics
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , ,
View Times: 163
Revisions: 2 times (View History)
Update Date: 17 Feb 2024
1000/1000