Skip to main content
Top
Published in: Breast Cancer Research 1/2019

Open Access 01-12-2019 | Metastasis | Research article

Investigating circulating tumor cells and distant metastases in patient-derived orthotopic xenograft models of triple-negative breast cancer

Published in: Breast Cancer Research | Issue 1/2019

Login to get access

Abstract

Background

Circulating tumor cells (CTCs) represent a temporal “snapshot” of a patient’s cancer and changes that occur during disease evolution. There is an extensive literature studying CTCs in breast cancer patients, and particularly in those with metastatic disease. In parallel, there is an increasing use of patient-derived models in preclinical investigations of human cancers. Yet studies are still limited demonstrating CTC shedding and metastasis formation in patient-derived models of breast cancer.

Methods

We used seven patient-derived orthotopic xenograft (PDOX) models generated from triple-negative breast cancer (TNBC) patients to study CTCs and distant metastases. Tumor fragments from PDOX tissue from each of the seven models were implanted into 57 NOD scid gamma (NSG) mice, and tumor growth and volume were monitored. Human CTC capture from mouse blood was first optimized on the marker-agnostic Vortex CTC isolation platform, and whole blood was processed from 37 PDOX tumor-bearing mice.

Results

Staining and imaging revealed the presence of CTCs in 32/37 (86%). The total number of CTCs varied between different PDOX tumor models and between individual mice bearing the same PDOX tumors. CTCs were heterogeneous and showed cytokeratin (CK) positive, vimentin (VIM) positive, and mixed CK/VIM phenotypes. Metastases were detected in the lung (20/57, 35%), liver (7/57, 12%), and brain (1/57, less than 2%). The seven different PDOX tumor models displayed varying degrees of metastatic potential, including one TNBC PDOX tumor model that failed to generate any detectable metastases (0/8 mice) despite having CTCs present in the blood of 5/5 tested, suggesting that CTCs from this particular PDOX tumor model may typify metastatic inefficiency.

Conclusion

PDOX tumor models that shed CTCs and develop distant metastases represent an important tool for investigating TNBC.
Appendix
Available only for authorised users
Literature
2.
go back to reference Deng G, et al. Single cell mutational analysis of PIK3CA in circulating tumor cells and metastases in breast cancer reveals heterogeneity, discordance, and mutation persistence in cultured disseminated tumor cells from bone marrow. BMC Cancer. 2014;14:456.PubMedPubMedCentralCrossRef Deng G, et al. Single cell mutational analysis of PIK3CA in circulating tumor cells and metastases in breast cancer reveals heterogeneity, discordance, and mutation persistence in cultured disseminated tumor cells from bone marrow. BMC Cancer. 2014;14:456.PubMedPubMedCentralCrossRef
3.
go back to reference Zardavas D, Irrthum A, Swanton C, Piccart M. Clinical management of breast cancer heterogeneity. Nat Rev Clin Oncol. 2015;12:381–94.PubMedCrossRef Zardavas D, Irrthum A, Swanton C, Piccart M. Clinical management of breast cancer heterogeneity. Nat Rev Clin Oncol. 2015;12:381–94.PubMedCrossRef
4.
5.
go back to reference Roulot A, et al. Tumoral heterogeneity of breast cancer. Ann Biol Clin (Paris). 2016;74:653–60. Roulot A, et al. Tumoral heterogeneity of breast cancer. Ann Biol Clin (Paris). 2016;74:653–60.
6.
go back to reference Savas P, et al. The subclonal architecture of metastatic breast cancer: results from a prospective community-based rapid autopsy program “CASCADE”. PLoS Med. 2016;13:e1002204.PubMedPubMedCentralCrossRef Savas P, et al. The subclonal architecture of metastatic breast cancer: results from a prospective community-based rapid autopsy program “CASCADE”. PLoS Med. 2016;13:e1002204.PubMedPubMedCentralCrossRef
7.
go back to reference Avigdor BE, et al. Mutational profiles of breast cancer metastases from a rapid autopsy series reveal multiple evolutionary trajectories. JCI Insight. 2017;2:24.CrossRef Avigdor BE, et al. Mutational profiles of breast cancer metastases from a rapid autopsy series reveal multiple evolutionary trajectories. JCI Insight. 2017;2:24.CrossRef
8.
go back to reference Ellsworth RE, Blackburn HL, Shriver CD, Soon-Shiong P, Ellsworth DL. Molecular heterogeneity in breast cancer: state of the science and implications for patient care. Semin Cell Dev Biol. 2017;64:65–72.PubMedCrossRef Ellsworth RE, Blackburn HL, Shriver CD, Soon-Shiong P, Ellsworth DL. Molecular heterogeneity in breast cancer: state of the science and implications for patient care. Semin Cell Dev Biol. 2017;64:65–72.PubMedCrossRef
11.
go back to reference De Marchi T, Foekens JA, Umar A, Martens JW. Endocrine therapy resistance in estrogen receptor (ER)-positive breast cancer. Drug Discov Today. 2016;21:1181–8.PubMedCrossRef De Marchi T, Foekens JA, Umar A, Martens JW. Endocrine therapy resistance in estrogen receptor (ER)-positive breast cancer. Drug Discov Today. 2016;21:1181–8.PubMedCrossRef
12.
go back to reference Luque-Cabal M, García-Teijido P, Fernández-Pérez Y, Sánchez-Lorenzo L, Palacio-Vázquez I. Mechanisms behind the resistance to trastuzumab in HER2-amplified breast cancer and strategies to overcome it. Clin Med Insights Oncol. 2016;10(Suppl 1):21–30.PubMedPubMedCentral Luque-Cabal M, García-Teijido P, Fernández-Pérez Y, Sánchez-Lorenzo L, Palacio-Vázquez I. Mechanisms behind the resistance to trastuzumab in HER2-amplified breast cancer and strategies to overcome it. Clin Med Insights Oncol. 2016;10(Suppl 1):21–30.PubMedPubMedCentral
13.
go back to reference Gingras I, Gebhart G, de Azambuja E, Piccart-Gebhart M. HER2-positive breast cancer is lost in translation: time for patient-centered research. Nat Rev Clin Oncol. 2017;14:669–81.PubMedCrossRef Gingras I, Gebhart G, de Azambuja E, Piccart-Gebhart M. HER2-positive breast cancer is lost in translation: time for patient-centered research. Nat Rev Clin Oncol. 2017;14:669–81.PubMedCrossRef
14.
go back to reference Parakh S, et al. Evolution of anti-HER2 therapies for cancer treatment. Cancer Treat Rev. 2017;59:1–21.PubMedCrossRef Parakh S, et al. Evolution of anti-HER2 therapies for cancer treatment. Cancer Treat Rev. 2017;59:1–21.PubMedCrossRef
15.
go back to reference Reinhardt F, Franken A, Fehm T, Neubauer H. Navigation through inter- and intratumoral heterogeneity of endocrine resistance mechanisms in breast cancer: a potential role for liquid biopsies? Tumour Biol. 2017;39:1010428317731511.PubMedCrossRef Reinhardt F, Franken A, Fehm T, Neubauer H. Navigation through inter- and intratumoral heterogeneity of endocrine resistance mechanisms in breast cancer: a potential role for liquid biopsies? Tumour Biol. 2017;39:1010428317731511.PubMedCrossRef
16.
go back to reference Kalimutho M, et al. Targeted therapies for triple-negative breast cancer: combating a stubborn disease. Trends Pharmacol Sci. 2015;36:822–46.PubMedCrossRef Kalimutho M, et al. Targeted therapies for triple-negative breast cancer: combating a stubborn disease. Trends Pharmacol Sci. 2015;36:822–46.PubMedCrossRef
17.
go back to reference Székely B, Silber AL, Pusztai L. New therapeutic strategies for triple-negative breast cancer. Oncology (Williston Park). 2017;31:130–7. Székely B, Silber AL, Pusztai L. New therapeutic strategies for triple-negative breast cancer. Oncology (Williston Park). 2017;31:130–7.
18.
go back to reference Malorni L, et al. Clinical and biologic features of triple-negative breast cancers in a large cohort of patients with long-term follow-up. Breast Cancer Res Treat. 2012;136:795–804.PubMedPubMedCentralCrossRef Malorni L, et al. Clinical and biologic features of triple-negative breast cancers in a large cohort of patients with long-term follow-up. Breast Cancer Res Treat. 2012;136:795–804.PubMedPubMedCentralCrossRef
19.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
22.
go back to reference Zhang J, et al. Chemotherapy of metastatic triple negative breast cancer: experience of using platinum-based chemotherapy. Oncotarget. 2015;6:43135–43.PubMedPubMedCentral Zhang J, et al. Chemotherapy of metastatic triple negative breast cancer: experience of using platinum-based chemotherapy. Oncotarget. 2015;6:43135–43.PubMedPubMedCentral
23.
go back to reference Gobbini E, et al. Time trends of overall survival among metastatic breast cancer patients in the real-life ESME cohort. Eur J Cancer. 2018;96:17–24.PubMedCrossRef Gobbini E, et al. Time trends of overall survival among metastatic breast cancer patients in the real-life ESME cohort. Eur J Cancer. 2018;96:17–24.PubMedCrossRef
24.
go back to reference Geenen JJJ, Linn SC, Beijnen JH, Schellens JHM. PARP inhibitors in the treatment of triple-negative breast cancer. Clin Pharmacokinet. 2018;57(4):427–37.PubMedCrossRef Geenen JJJ, Linn SC, Beijnen JH, Schellens JHM. PARP inhibitors in the treatment of triple-negative breast cancer. Clin Pharmacokinet. 2018;57(4):427–37.PubMedCrossRef
25.
go back to reference Costa RLB, Han HS, Gradishar WJ. Targeting the PI3K/AKT/mTOR pathway in triple-negative breast cancer: a review. Breast Cancer Res Treat. 2018;169(3):397–406.PubMedCrossRef Costa RLB, Han HS, Gradishar WJ. Targeting the PI3K/AKT/mTOR pathway in triple-negative breast cancer: a review. Breast Cancer Res Treat. 2018;169(3):397–406.PubMedCrossRef
26.
go back to reference Prasanna T, et al. Optimizing poly (ADP-ribose) polymerase inhibition through combined epigenetic and immunotherapy. Cancer Sci. 2018;109(11):3383–92.PubMedPubMedCentralCrossRef Prasanna T, et al. Optimizing poly (ADP-ribose) polymerase inhibition through combined epigenetic and immunotherapy. Cancer Sci. 2018;109(11):3383–92.PubMedPubMedCentralCrossRef
27.
go back to reference Soundararajan R, et al. Targeting the interplay between epithelial-to-mesenchymal-transition and the immune system for effective immunotherapy. Cancers (Basel). 2019;11:5.CrossRef Soundararajan R, et al. Targeting the interplay between epithelial-to-mesenchymal-transition and the immune system for effective immunotherapy. Cancers (Basel). 2019;11:5.CrossRef
28.
go back to reference Voorwerk L, et al. Immune induction strategies in metastatic triple-negative breast cancer to enhance the sensitivity to PD-1 blockade: the TONIC trial. Nat Med. 2019;25(6):920–8.PubMedCrossRef Voorwerk L, et al. Immune induction strategies in metastatic triple-negative breast cancer to enhance the sensitivity to PD-1 blockade: the TONIC trial. Nat Med. 2019;25(6):920–8.PubMedCrossRef
30.
go back to reference McCann KE, Hurvitz SA, McAndrew N. Advances in targeted therapies for triple-negative breast cancer. Drugs. 2019;79(11):1217–30.PubMedCrossRef McCann KE, Hurvitz SA, McAndrew N. Advances in targeted therapies for triple-negative breast cancer. Drugs. 2019;79(11):1217–30.PubMedCrossRef
32.
go back to reference Guy CT, Cardiff RD, Muller WJ. Induction of mammary tumors by expression of polyomavirus middle T oncogene: a transgenic mouse model for metastatic disease. Mol Cell Biol. 1992;12:954–61.PubMedPubMedCentralCrossRef Guy CT, Cardiff RD, Muller WJ. Induction of mammary tumors by expression of polyomavirus middle T oncogene: a transgenic mouse model for metastatic disease. Mol Cell Biol. 1992;12:954–61.PubMedPubMedCentralCrossRef
33.
go back to reference Lin EY, et al. Progression to malignancy in the polyoma middle T oncoprotein mouse breast cancer model provides a reliable model for human diseases. Am J Pathol. 2003;163:2113–26.PubMedPubMedCentralCrossRef Lin EY, et al. Progression to malignancy in the polyoma middle T oncoprotein mouse breast cancer model provides a reliable model for human diseases. Am J Pathol. 2003;163:2113–26.PubMedPubMedCentralCrossRef
34.
go back to reference Herschkowitz JI, et al. Identification of conserved gene expression features between murine mammary carcinoma models and human breast tumors. Genome Biol. 2007;8:R76.PubMedPubMedCentralCrossRef Herschkowitz JI, et al. Identification of conserved gene expression features between murine mammary carcinoma models and human breast tumors. Genome Biol. 2007;8:R76.PubMedPubMedCentralCrossRef
35.
36.
go back to reference Manning HC, Buck JR, Cook RS. Mouse models of breast cancer: platforms for discovering precision imaging diagnostics and future cancer medicine. J Nucl Med. 2016;57(Suppl 1):60S–8S.PubMedCrossRef Manning HC, Buck JR, Cook RS. Mouse models of breast cancer: platforms for discovering precision imaging diagnostics and future cancer medicine. J Nucl Med. 2016;57(Suppl 1):60S–8S.PubMedCrossRef
37.
38.
go back to reference Gengenbacher N, Singhal M, Augustin HG. Preclinical mouse solid tumour models: status quo, challenges and perspectives. Nat Rev Cancer. 2017;17:751–65.PubMedCrossRef Gengenbacher N, Singhal M, Augustin HG. Preclinical mouse solid tumour models: status quo, challenges and perspectives. Nat Rev Cancer. 2017;17:751–65.PubMedCrossRef
40.
41.
go back to reference Izumchenko E, et al. Patient-derived xenografts effectively capture responses to oncology therapy in a heterogeneous cohort of patients with solid tumors. Ann Oncol. 2017;28:2595–605.PubMedPubMedCentralCrossRef Izumchenko E, et al. Patient-derived xenografts effectively capture responses to oncology therapy in a heterogeneous cohort of patients with solid tumors. Ann Oncol. 2017;28:2595–605.PubMedPubMedCentralCrossRef
42.
go back to reference Sulaiman A, Wang L. Bridging the divide: preclinical research discrepancies between triple-negative breast cancer cell lines and patient tumors. Oncotarget. 2017;8:113269–81.PubMedPubMedCentralCrossRef Sulaiman A, Wang L. Bridging the divide: preclinical research discrepancies between triple-negative breast cancer cell lines and patient tumors. Oncotarget. 2017;8:113269–81.PubMedPubMedCentralCrossRef
43.
go back to reference Petrillo LA, et al. Xenografts faithfully recapitulate breast cancer-specific gene expression patterns of parent primary breast tumors. Breast Cancer Res Treat. 2012;135:913–22.PubMedCrossRef Petrillo LA, et al. Xenografts faithfully recapitulate breast cancer-specific gene expression patterns of parent primary breast tumors. Breast Cancer Res Treat. 2012;135:913–22.PubMedCrossRef
44.
go back to reference Zhang H, et al. Patient-derived xenografts of triple-negative breast cancer reproduce molecular features of patient tumors and respond to mTOR inhibition. Breast Cancer Res. 2014;16:R36.PubMedPubMedCentralCrossRef Zhang H, et al. Patient-derived xenografts of triple-negative breast cancer reproduce molecular features of patient tumors and respond to mTOR inhibition. Breast Cancer Res. 2014;16:R36.PubMedPubMedCentralCrossRef
45.
go back to reference Zhang X, et al. A renewable tissue resource of phenotypically stable, biologically and ethnically diverse, patient-derived human breast cancer xenograft models. Cancer Res. 2013;73:4885–97.PubMedPubMedCentralCrossRef Zhang X, et al. A renewable tissue resource of phenotypically stable, biologically and ethnically diverse, patient-derived human breast cancer xenograft models. Cancer Res. 2013;73:4885–97.PubMedPubMedCentralCrossRef
46.
go back to reference Hoffman RM. Patient-derived orthotopic xenografts: better mimic of metastasis than subcutaneous xenografts. Nat Rev Cancer. 2015;15:451–2.PubMedCrossRef Hoffman RM. Patient-derived orthotopic xenografts: better mimic of metastasis than subcutaneous xenografts. Nat Rev Cancer. 2015;15:451–2.PubMedCrossRef
47.
go back to reference Byrne AT, et al. Interrogating open issues in cancer precision medicine with patient-derived xenografts. Nat Rev Cancer. 2017;17:254–68.PubMedCrossRef Byrne AT, et al. Interrogating open issues in cancer precision medicine with patient-derived xenografts. Nat Rev Cancer. 2017;17:254–68.PubMedCrossRef
48.
go back to reference Moon HG, et al. Prognostic and functional importance of the engraftment-associated genes in the patient-derived xenograft models of triple-negative breast cancers. Breast Cancer Res Treat. 2015;154:13–22.PubMedCrossRef Moon HG, et al. Prognostic and functional importance of the engraftment-associated genes in the patient-derived xenograft models of triple-negative breast cancers. Breast Cancer Res Treat. 2015;154:13–22.PubMedCrossRef
51.
52.
go back to reference Giuliano M, et al. Circulating and disseminated tumor cells from breast cancer patient-derived xenograft-bearing mice as a novel model to study metastasis. Breast Cancer Res. 2015;17:3.PubMedPubMedCentralCrossRef Giuliano M, et al. Circulating and disseminated tumor cells from breast cancer patient-derived xenograft-bearing mice as a novel model to study metastasis. Breast Cancer Res. 2015;17:3.PubMedPubMedCentralCrossRef
54.
go back to reference Powell E, et al. p53 deficiency linked to B cell translocation gene 2 (BTG2) loss enhances metastatic potential by promoting tumor growth in primary and metastatic sites in patient-derived xenograft (PDX) models of triple-negative breast cancer. Breast Cancer Res. 2016;18:13.PubMedPubMedCentralCrossRef Powell E, et al. p53 deficiency linked to B cell translocation gene 2 (BTG2) loss enhances metastatic potential by promoting tumor growth in primary and metastatic sites in patient-derived xenograft (PDX) models of triple-negative breast cancer. Breast Cancer Res. 2016;18:13.PubMedPubMedCentralCrossRef
55.
go back to reference Pillai SG, et al. Identifying biomarkers of breast cancer micrometastatic disease in bone marrow using a patient-derived xenograft mouse model. Breast Cancer Res. 2018;20:2.PubMedPubMedCentralCrossRef Pillai SG, et al. Identifying biomarkers of breast cancer micrometastatic disease in bone marrow using a patient-derived xenograft mouse model. Breast Cancer Res. 2018;20:2.PubMedPubMedCentralCrossRef
56.
go back to reference Ramirez AB, et al. Circulating tumor cell investigation in breast cancer patient-derived xenograft models by automated immunofluorescence staining, image acquisition, and single cell retrieval and analysis. BMC Cancer. 2019;19(1):220.PubMedPubMedCentralCrossRef Ramirez AB, et al. Circulating tumor cell investigation in breast cancer patient-derived xenograft models by automated immunofluorescence staining, image acquisition, and single cell retrieval and analysis. BMC Cancer. 2019;19(1):220.PubMedPubMedCentralCrossRef
57.
go back to reference Tachtsidis A, et al. Human-specific RNA analysis shows uncoupled epithelial-mesenchymal plasticity in circulating and disseminated tumour cells from human breast cancer xenografts. Clin Exp Metastasis. 2019;36(4):393–409.PubMedCrossRef Tachtsidis A, et al. Human-specific RNA analysis shows uncoupled epithelial-mesenchymal plasticity in circulating and disseminated tumour cells from human breast cancer xenografts. Clin Exp Metastasis. 2019;36(4):393–409.PubMedCrossRef
59.
go back to reference Ignatiadis M, Lee M, Jeffrey SS. Circulating tumor cells and circulating tumor DNA: challenges and opportunities on the path to clinical utility. Clin Cancer Res. 2015;21:4786–800.PubMedCrossRef Ignatiadis M, Lee M, Jeffrey SS. Circulating tumor cells and circulating tumor DNA: challenges and opportunities on the path to clinical utility. Clin Cancer Res. 2015;21:4786–800.PubMedCrossRef
61.
go back to reference Gabriel MT, Calleja LR, Chalopin A, Ory B, Heymann D. Circulating tumor cells: a review of non-EpCAM-based approaches for cell enrichment and isolation. Clin Chem. 2016;62:571–81.PubMedCrossRef Gabriel MT, Calleja LR, Chalopin A, Ory B, Heymann D. Circulating tumor cells: a review of non-EpCAM-based approaches for cell enrichment and isolation. Clin Chem. 2016;62:571–81.PubMedCrossRef
62.
go back to reference Sollier E, et al. Size-selective collection of circulating tumor cells using Vortex technology. Lab Chip. 2014;14:63–77.PubMedCrossRef Sollier E, et al. Size-selective collection of circulating tumor cells using Vortex technology. Lab Chip. 2014;14:63–77.PubMedCrossRef
63.
go back to reference Dhar M, et al. Label-free enumeration, collection and downstream cytological and cytogenetic analysis of circulating tumor cells. Sci Rep. 2016;6:35474.PubMedPubMedCentralCrossRef Dhar M, et al. Label-free enumeration, collection and downstream cytological and cytogenetic analysis of circulating tumor cells. Sci Rep. 2016;6:35474.PubMedPubMedCentralCrossRef
64.
go back to reference Kidess-Sigal E, et al. Enumeration and targeted analysis of KRAS, BRAF and PIK3CA mutations in CTCs captured by a label-free platform: comparison to ctDNA and tissue in metastatic colorectal cancer. Oncotarget. 2016;7:85349–64.PubMedPubMedCentralCrossRef Kidess-Sigal E, et al. Enumeration and targeted analysis of KRAS, BRAF and PIK3CA mutations in CTCs captured by a label-free platform: comparison to ctDNA and tissue in metastatic colorectal cancer. Oncotarget. 2016;7:85349–64.PubMedPubMedCentralCrossRef
66.
67.
68.
go back to reference Che J, et al. Classification of large circulating tumor cells isolated with ultra-high throughput microfluidic Vortex technology. Oncotarget. 2016;7:12748–60.PubMedPubMedCentralCrossRef Che J, et al. Classification of large circulating tumor cells isolated with ultra-high throughput microfluidic Vortex technology. Oncotarget. 2016;7:12748–60.PubMedPubMedCentralCrossRef
69.
go back to reference Paulmurugan R, et al. Folate receptor-targeted polymeric micellar nanocarriers for delivery of orlistat as a repurposed drug against triple-negative breast cancer. Mol Cancer Ther. 2016;15:221–31.PubMedCrossRef Paulmurugan R, et al. Folate receptor-targeted polymeric micellar nanocarriers for delivery of orlistat as a repurposed drug against triple-negative breast cancer. Mol Cancer Ther. 2016;15:221–31.PubMedCrossRef
70.
go back to reference Windberger U, Bartholovitsch A, Plasenzotti R, Korak KJ, Heinze G. Whole blood viscosity, plasma viscosity and erythrocyte aggregation in nine mammalian species: reference values and comparison of data. Exp Physiol. 2003;88:431–40.PubMedCrossRef Windberger U, Bartholovitsch A, Plasenzotti R, Korak KJ, Heinze G. Whole blood viscosity, plasma viscosity and erythrocyte aggregation in nine mammalian species: reference values and comparison of data. Exp Physiol. 2003;88:431–40.PubMedCrossRef
71.
go back to reference Cabel L, et al. Circulating tumor cells: clinical validity and utility. Int J Clin Oncol. 2017;22:421–30.PubMedCrossRef Cabel L, et al. Circulating tumor cells: clinical validity and utility. Int J Clin Oncol. 2017;22:421–30.PubMedCrossRef
72.
go back to reference Paez-Ribes M, Man S, Xu P, Kerbel RS. Development of patient derived xenograft models of overt spontaneous breast cancer metastasis: a cautionary note. PLoS One. 2016;11:e0158034.PubMedPubMedCentralCrossRef Paez-Ribes M, Man S, Xu P, Kerbel RS. Development of patient derived xenograft models of overt spontaneous breast cancer metastasis: a cautionary note. PLoS One. 2016;11:e0158034.PubMedPubMedCentralCrossRef
73.
74.
77.
go back to reference Luzzi KJ, et al. Multistep nature of metastatic inefficiency: dormancy of solitary cells after successful extravasation and limited survival of early micrometastases. Am J Pathol. 1998;153:865–73.PubMedPubMedCentralCrossRef Luzzi KJ, et al. Multistep nature of metastatic inefficiency: dormancy of solitary cells after successful extravasation and limited survival of early micrometastases. Am J Pathol. 1998;153:865–73.PubMedPubMedCentralCrossRef
78.
go back to reference Chambers AF, Naumov GN, Vantyghem SA, Tuck AB. Molecular biology of breast cancer metastasis. Clinical implications of experimental studies on metastatic inefficiency. Breast Cancer Res. 2000;2:400–7.PubMedPubMedCentralCrossRef Chambers AF, Naumov GN, Vantyghem SA, Tuck AB. Molecular biology of breast cancer metastasis. Clinical implications of experimental studies on metastatic inefficiency. Breast Cancer Res. 2000;2:400–7.PubMedPubMedCentralCrossRef
79.
go back to reference Azevedo AS, Follain G, Patthabhiraman S, Harlepp S, Goetz JG. Metastasis of circulating tumor cells: favorable soil or suitable biomechanics, or both? Cell Adhes Migr. 2015;9:345–56.CrossRef Azevedo AS, Follain G, Patthabhiraman S, Harlepp S, Goetz JG. Metastasis of circulating tumor cells: favorable soil or suitable biomechanics, or both? Cell Adhes Migr. 2015;9:345–56.CrossRef
81.
go back to reference Strilic B, Offermanns S. Intravascular survival and extravasation of tumor cells. Cancer Cell. 2017;32:282–93.PubMedCrossRef Strilic B, Offermanns S. Intravascular survival and extravasation of tumor cells. Cancer Cell. 2017;32:282–93.PubMedCrossRef
82.
go back to reference Labelle M, Hynes RO. The initial hours of metastasis: the importance of cooperative host-tumor cell interactions during hematogenous dissemination. Cancer Discov. 2012;2:1091–9.PubMedPubMedCentralCrossRef Labelle M, Hynes RO. The initial hours of metastasis: the importance of cooperative host-tumor cell interactions during hematogenous dissemination. Cancer Discov. 2012;2:1091–9.PubMedPubMedCentralCrossRef
85.
87.
go back to reference Cheung KJ, et al. Polyclonal breast cancer metastases arise from collective dissemination of keratin 14-expressing tumor cell clusters. Proc Natl Acad Sci U S A. 2016;113:E854–63.PubMedPubMedCentralCrossRef Cheung KJ, et al. Polyclonal breast cancer metastases arise from collective dissemination of keratin 14-expressing tumor cell clusters. Proc Natl Acad Sci U S A. 2016;113:E854–63.PubMedPubMedCentralCrossRef
89.
go back to reference Suo Y, et al. Proportion of circulating tumor cell clusters increases during cancer metastasis. Cytometry A. 2017;91:250–3.PubMedCrossRef Suo Y, et al. Proportion of circulating tumor cell clusters increases during cancer metastasis. Cytometry A. 2017;91:250–3.PubMedCrossRef
90.
go back to reference Giuliano M, et al. Perspective on circulating tumor cell clusters: why it takes a village to metastasize. Cancer Res. 2018;78:845–52.PubMedCrossRef Giuliano M, et al. Perspective on circulating tumor cell clusters: why it takes a village to metastasize. Cancer Res. 2018;78:845–52.PubMedCrossRef
91.
go back to reference Mu Z, et al. Prospective assessment of the prognostic value of circulating tumor cells and their clusters in patients with advanced-stage breast cancer. Breast Cancer Res Treat. 2015;154:563–71.PubMedCrossRef Mu Z, et al. Prospective assessment of the prognostic value of circulating tumor cells and their clusters in patients with advanced-stage breast cancer. Breast Cancer Res Treat. 2015;154:563–71.PubMedCrossRef
92.
go back to reference Wang C, et al. Longitudinally collected CTCs and CTC-clusters and clinical outcomes of metastatic breast cancer. Breast Cancer Res Treat. 2017;161:83–94.PubMedCrossRef Wang C, et al. Longitudinally collected CTCs and CTC-clusters and clinical outcomes of metastatic breast cancer. Breast Cancer Res Treat. 2017;161:83–94.PubMedCrossRef
93.
go back to reference Larsson AM, et al. Longitudinal enumeration and cluster evaluation of circulating tumor cells improve prognostication for patients with newly diagnosed metastatic breast cancer in a prospective observational trial. Breast Cancer Res. 2018;20:48.PubMedPubMedCentralCrossRef Larsson AM, et al. Longitudinal enumeration and cluster evaluation of circulating tumor cells improve prognostication for patients with newly diagnosed metastatic breast cancer in a prospective observational trial. Breast Cancer Res. 2018;20:48.PubMedPubMedCentralCrossRef
94.
go back to reference Hou JM, et al. Clinical significance and molecular characteristics of circulating tumor cells and circulating tumor microemboli in patients with small-cell lung cancer. J Clin Oncol. 2012;30:525–32.PubMedCrossRef Hou JM, et al. Clinical significance and molecular characteristics of circulating tumor cells and circulating tumor microemboli in patients with small-cell lung cancer. J Clin Oncol. 2012;30:525–32.PubMedCrossRef
96.
go back to reference Fabisiewicz A, Grzybowska E. CTC clusters in cancer progression and metastasis. Med Oncol. 2017;34:12.PubMedCrossRef Fabisiewicz A, Grzybowska E. CTC clusters in cancer progression and metastasis. Med Oncol. 2017;34:12.PubMedCrossRef
97.
99.
go back to reference Cheng SB, et al. Three-dimensional scaffold chip with thermosensitive coating for capture and reversible release of individual and cluster of circulating tumor cells. Anal Chem. 2017;89:7924–32.CrossRefPubMed Cheng SB, et al. Three-dimensional scaffold chip with thermosensitive coating for capture and reversible release of individual and cluster of circulating tumor cells. Anal Chem. 2017;89:7924–32.CrossRefPubMed
101.
go back to reference Powell AA, et al. Single cell profiling of circulating tumor cells: transcriptional heterogeneity and diversity from breast cancer cell lines. PLoS One. 2012;7:e33788.PubMedPubMedCentralCrossRef Powell AA, et al. Single cell profiling of circulating tumor cells: transcriptional heterogeneity and diversity from breast cancer cell lines. PLoS One. 2012;7:e33788.PubMedPubMedCentralCrossRef
102.
go back to reference Kallergi G, et al. Epithelial to mesenchymal transition markers expressed in circulating tumour cells of early and metastatic breast cancer patients. Breast Cancer Res. 2011;13:R59.PubMedPubMedCentralCrossRef Kallergi G, et al. Epithelial to mesenchymal transition markers expressed in circulating tumour cells of early and metastatic breast cancer patients. Breast Cancer Res. 2011;13:R59.PubMedPubMedCentralCrossRef
103.
104.
go back to reference Alix-Panabières C, Mader S, Pantel K. Epithelial-mesenchymal plasticity in circulating tumor cells. J Mol Med (Berl). 2017;95:133–42.CrossRef Alix-Panabières C, Mader S, Pantel K. Epithelial-mesenchymal plasticity in circulating tumor cells. J Mol Med (Berl). 2017;95:133–42.CrossRef
105.
go back to reference Huang RY, et al. An EMT spectrum defines an anoikis-resistant and spheroidogenic intermediate mesenchymal state that is sensitive to e-cadherin restoration by a src-kinase inhibitor, saracatinib (AZD0530). Cell Death Dis. 2013;4:e915.PubMedPubMedCentralCrossRef Huang RY, et al. An EMT spectrum defines an anoikis-resistant and spheroidogenic intermediate mesenchymal state that is sensitive to e-cadherin restoration by a src-kinase inhibitor, saracatinib (AZD0530). Cell Death Dis. 2013;4:e915.PubMedPubMedCentralCrossRef
106.
go back to reference Liao TT, Yang MH. Revisiting epithelial-mesenchymal transition in cancer metastasis: the connection between epithelial plasticity and stemness. Mol Oncol. 2017;11:792–804.PubMedPubMedCentralCrossRef Liao TT, Yang MH. Revisiting epithelial-mesenchymal transition in cancer metastasis: the connection between epithelial plasticity and stemness. Mol Oncol. 2017;11:792–804.PubMedPubMedCentralCrossRef
107.
go back to reference Jolly MK, Mani SA, Levine H. Hybrid epithelial/mesenchymal phenotype(s): the ‘fittest’ for metastasis? Biochim Biophys Acta Rev Cancer. 2018;1870:151–7.PubMedCrossRef Jolly MK, Mani SA, Levine H. Hybrid epithelial/mesenchymal phenotype(s): the ‘fittest’ for metastasis? Biochim Biophys Acta Rev Cancer. 2018;1870:151–7.PubMedCrossRef
108.
go back to reference Beerling E, et al. Plasticity between epithelial and mesenchymal states unlinks EMT from metastasis-enhancing stem cell capacity. Cell Rep. 2016;14:2281–8.PubMedPubMedCentralCrossRef Beerling E, et al. Plasticity between epithelial and mesenchymal states unlinks EMT from metastasis-enhancing stem cell capacity. Cell Rep. 2016;14:2281–8.PubMedPubMedCentralCrossRef
110.
go back to reference Chaffer CL, San Juan BP, Lim E, Weinberg RA. EMT, cell plasticity and metastasis. Cancer Metastasis Rev. 2016;35:645–54.PubMedCrossRef Chaffer CL, San Juan BP, Lim E, Weinberg RA. EMT, cell plasticity and metastasis. Cancer Metastasis Rev. 2016;35:645–54.PubMedCrossRef
112.
113.
114.
go back to reference Chen Y, et al. Dual reporter genetic mouse models of pancreatic cancer identify an epithelial-to-mesenchymal transition-independent metastasis program. EMBO Mol Med. 2018;10:10.CrossRef Chen Y, et al. Dual reporter genetic mouse models of pancreatic cancer identify an epithelial-to-mesenchymal transition-independent metastasis program. EMBO Mol Med. 2018;10:10.CrossRef
116.
go back to reference Hensler M, et al. Gene expression profiling of circulating tumor cells and peripheral blood mononuclear cells from breast cancer patients. Oncoimmunology. 2015;5:e1102827.PubMedPubMedCentralCrossRef Hensler M, et al. Gene expression profiling of circulating tumor cells and peripheral blood mononuclear cells from breast cancer patients. Oncoimmunology. 2015;5:e1102827.PubMedPubMedCentralCrossRef
117.
go back to reference Onstenk W, et al. Gene expression profiles of circulating tumor cells versus primary tumors in metastatic breast cancer. Cancer Lett. 2015;362:36–44.PubMedCrossRef Onstenk W, et al. Gene expression profiles of circulating tumor cells versus primary tumors in metastatic breast cancer. Cancer Lett. 2015;362:36–44.PubMedCrossRef
118.
go back to reference Magbanua MJM, et al. Expanded genomic profiling of circulating tumor cells in metastatic breast cancer patients to assess biomarker status and biology over time (CALGB 40502 and CALGB 40503, Alliance). Clin Cancer Res. 2018;24:1486–99.PubMedPubMedCentralCrossRef Magbanua MJM, et al. Expanded genomic profiling of circulating tumor cells in metastatic breast cancer patients to assess biomarker status and biology over time (CALGB 40502 and CALGB 40503, Alliance). Clin Cancer Res. 2018;24:1486–99.PubMedPubMedCentralCrossRef
120.
122.
go back to reference Kwan TT, et al. A digital RNA signature of circulating tumor cells predicting early therapeutic response in localized and metastatic breast cancer. Cancer Discov. 2018;8(10):1286–99.PubMedCrossRefPubMedCentral Kwan TT, et al. A digital RNA signature of circulating tumor cells predicting early therapeutic response in localized and metastatic breast cancer. Cancer Discov. 2018;8(10):1286–99.PubMedCrossRefPubMedCentral
123.
go back to reference Yeung C, et al. Estrogen, progesterone, and HER2/neu receptor discordance between primary and metastatic breast tumours-a review. Cancer Metastasis Rev. 2016;35:427–37.PubMedCrossRef Yeung C, et al. Estrogen, progesterone, and HER2/neu receptor discordance between primary and metastatic breast tumours-a review. Cancer Metastasis Rev. 2016;35:427–37.PubMedCrossRef
Metadata
Title
Investigating circulating tumor cells and distant metastases in patient-derived orthotopic xenograft models of triple-negative breast cancer
Publication date
01-12-2019
Published in
Breast Cancer Research / Issue 1/2019
Electronic ISSN: 1465-542X
DOI
https://doi.org/10.1186/s13058-019-1182-4

Other articles of this Issue 1/2019

Breast Cancer Research 1/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine