The Fibrillar Matrix: Novel Avenues for Breast Cancer Detection and Treatment

Rasha Rezk, Raquel Marín-García, Annica K.B.Gad

PDF(338 KB)
PDF(338 KB)
Engineering ›› 2021, Vol. 7 ›› Issue (10) : 1375-1380. DOI: 10.1016/j.eng.2021.04.024

The Fibrillar Matrix: Novel Avenues for Breast Cancer Detection and Treatment

Author information +
History +

Abstract

Breast cancer is marked by large increases in the protein fibers around tumor cells. These fibers increase the mechanical stiffness of the tissue, which has long been used for tumor diagnosis by manual palpation. Recent research in bioengineering has led to the development of novel biomaterials that model the mechanical and architectural properties of the tumor microenvironment and can be used to understand how these cues regulate the growth and spread of breast cancer. Herein, we provide an overview of how the mechanical properties of breast tumor tissues differ from those of normal breast tissue and non-cancerous lesions. We also describe how biomaterial models make it possible to understand how the stiffness and viscosity of the extracellular environment regulate cell migration and breast cancer metastasis. We highlight the need for biomaterial models that allow independent analysis of the individual and different mechanical properties of the tumor microenvironment and that use cells derived from different regions within tumors. These models will guide the development of novel mechano-based therapies against breast cancer metastasis.

Keywords

Breast cancer / Tissue stiffness / Cancer metastasis / Cell migration / Bioengineered scaffold / Viscosity

Cite this article

Download citation ▾
Rasha Rezk, Raquel Marín-García, Annica K.B.Gad. The Fibrillar Matrix: Novel Avenues for Breast Cancer Detection and Treatment. Engineering, 2021, 7(10): 1375‒1380 https://doi.org/10.1016/j.eng.2021.04.024

References

[[1]]
Discher DE, Janmey P, Wang YL. Tissue cells feel and respond to the stiffness of their substrate. Science 2005;310(5751):1139–43.
[[2]]
Heinrich MA, Alert R, LaChance JM, Zajdel TJ, Košmrlj A, Cohen DJ. Sizedependent patterns of cell proliferation and migration in freely-expanding epithelia. eLife 2020;9:e58945.
[[3]]
Northcott JM, Dean IS, Mouw JK, Weaver VM. Feeling stress: the mechanics of cancer progression and aggression. Front Cell Dev Biol 2018;6:17.
[[4]]
Kumar S, Weaver VM. Mechanics, malignancy, and metastasis: the force journey of a tumor cell. Cancer Metastasis Rev 2009;28(1–2):113–27.
[[5]]
Butcher DT, Alliston T, Weaver VM. A tense situation: forcing tumour progression. Nat Rev Cancer 2009;9(2):108–22.
[[6]]
Michor F, Liphardt J, Ferrari M, Widom J. What does physics have to do with cancer? Nat Rev Cancer 2011;11(9):657–70.
[[7]]
Acerbi I, Cassereau L, Dean I, Shi Q, Au A, Park C, et al. Human breast cancer invasion and aggression correlates with ECM stiffening and immune cell infiltration. Integr Biol 2015;7(10):1120–34.
[[8]]
Makki J. Diversity of breast carcinoma: histological subtypes and clinical relevance. Clin Med Insights Pathol 2015;8:23–31.
[[9]]
Weigelt B, Reis-Filho JS. Histological and molecular types of breast cancer: is there a unifying taxonomy? Nat Rev Clin Oncol 2009;6(12):718–30.
[[10]]
Tian J, Liu Q, Wang X, Xing P, Yang Z, Wu C. Application of 3D and 2D quantitative shear wave elastography (SWE) to differentiate between benign and malignant breast masses. Sci Rep 2017;7(1):41216.
[[11]]
Chang JM, Moon WK, Cho N, Yi A, Koo HR, Han W, et al. Clinical application of shear wave elastography (SWE) in the diagnosis of benign and malignant breast diseases. Breast Cancer Res Treat 2011;129(1):89–97.
[[12]]
Ansardamavandi A, Tafazzoli-Shadpour M, Omidvar R, Jahanzad I. Quantification of effects of cancer on elastic properties of breast tissue by atomic force microscopy. J Mech Behav Biomed Mater 2016;60:234–42.
[[13]]
Berg WA, Mendelson EB, Cosgrove DO, Doré CJ, Gay J, Henry JP, et al. Quantitative maximum shear-wave stiffness of breast masses as a predictor of histopathologic severity. AJR Am J Roentgenol 2015;205(2):448–55.
[[14]]
Suvannarerg V, Chitchumnong P, Apiwat W, Lertdamrongdej L, Tretipwanit N, Pisarnturakit P, et al. Diagnostic performance of qualitative and quantitative shear wave elastography in differentiating malignant from benign breast masses, and association with the histological prognostic factors. Quant Imaging Med Surg 2019;9(3):386–98.
[[15]]
Rabin C, Benech N. Quantitative breast elastography from B-mode images. Med Phys 2019;46(7):3001–12.
[[16]]
Barcus CE, Keely PJ, Eliceiri KW, Schuler LA. Stiff collagen matrices increase tumorigenic prolactin signaling in breast cancer cells. J Biol Chem 2013;288 (18):12722–32.
[[17]]
Perepelyuk M, Terajima M, Wang AY, Georges PC, Janmey PA, Yamauchi M, et al. Hepatic stellate cells and portal fibroblasts are the major cellular sources of collagens and lysyl oxidases in normal liver and early after injury. Am J Physiol Gastrointest Liver Physiol 2013;304(6):G605–14.
[[18]]
Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell 2009;139(5):891–906.
[[19]]
Provenzano PP, Inman DR, Eliceiri KW, Knittel JG, Yan L, Rueden CT, et al. Collagen density promotes mammary tumor initiation and progression. BMC Med 2008;6(1):11.
[[20]]
Conklin MW, Eickhoff JC, Riching KM, Pehlke CA, Eliceiri KW, Provenzano PP, et al. Aligned collagen is a prognostic signature for survival in human breast carcinoma. Am J Pathol 2011;178(3):1221–32.
[[21]]
Gierach GL, Ichikawa L, Kerlikowske K, Brinton LA, Farhat GN, Vacek PM, et al. Relationship between mammographic density and breast cancer death in the Breast Cancer Surveillance Consortium. J Natl Cancer Inst 2012;104 (16):1218–27.
[[22]]
Fenner J, Stacer AC, Winterroth F, Johnson TD, Luker KE, Luker GD. Macroscopic stiffness of breast tumors predicts metastasis. Sci Rep 2015;4(1):5512.
[[23]]
Chaudhuri O, Koshy ST, Branco da Cunha C, Shin JW, Verbeke CS, Allison KH, et al. Extracellular matrix stiffness and composition jointly regulate the induction of malignant phenotypes in mammary epithelium. Nat Mater 2014;13(10):970–8.
[[24]]
Stowers RS, Shcherbina A, Israeli J, Gruber JJ, Chang J, Nam S, et al. Matrix stiffness induces a tumorigenic phenotype in mammary epithelium through changes in chromatin accessibility. Nat Biomed Eng 2019;3(12):1009–19.
[[25]]
Berger AJ, Linsmeier KM, Kreeger PK, Masters KS. Decoupling the effects of stiffness and fiber density on cellular behaviors via an interpenetrating network of gelatin-methacrylate and collagen. Biomaterials 2017;141:125–35.
[[26]]
Bangasser B, Rosenfeld S, Odde D. Determinants of maximal force transmission in a motor-clutch model of cell traction in a compliant microenvironment. Biophys J 2013;105(3):581–92.
[[27]]
Wang WY, Davidson CD, Lin D, Baker BM. Actomyosin contractility-dependent matrix stretch and recoil induces rapid cell migration. Nat Commun 2019;10 (1):1186.
[[28]]
Baker BM, Trappmann B, Wang WY, Sakar MS, Kim IL, Shenoy VB, et al. Cellmediated fibre recruitment drives extracellular matrix mechanosensing in engineered fibrillar microenvironments. Nat Mater 2015;14(12):1262–8.
[[29]]
Matera DL, DiLillo KM, Smith MR, Davidson CD, Parikh R, Said M, et al. Microengineered 3D pulmonary interstitial mimetics highlight a critical role for matrix degradation in myofibroblast differentiation. Sci Adv 2020;6(37): eabb5069.
[[30]]
Sinkus R, Siegmann K, Xydeas T, Tanter M, Claussen C, Fink M. MR elastography of breast lesions: understanding the solid/liquid duality can improve the specificity of contrast-enhanced MR mammography. Magn Reson Med 2007;58(6):1135–44.
[[31]]
Wu W, Chen L, Wang Y, Jin J, Xie X, Zhang J. Hyaluronic acid predicts poor prognosis in breast cancer patients. Medicine 2020;99(22):e20438.
[[32]]
Streitberger KJ, Sack I, Krefting D, Pfüller C, Braun J, Paul F, et al. Brain viscoelasticity alteration in chronic-progressive multiple sclerosis. PLoS ONE 2012;7(1):e29888.
[[33]]
Wang Y, Xu C, Jiang N, Zheng L, Zeng J, Qiu C, et al. Quantitative analysis of the cell-surface roughness and viscoelasticity for breast cancer cells discrimination using atomic force microscopy. Scanning 2016;38(6):558–63.
[[34]]
Nematbakhsh Y, Pang KT, Lim CT. Correlating the viscoelasticity of breast cancer cells with their malignancy. Converg Sci Phys Oncol 2017;3(3):034003.
[[35]]
Harada T, Swift J, Irianto J, Shin JW, Spinler KR, Athirasala A, et al. Nuclear lamin stiffness is a barrier to 3D migration, but softness can limit survival. J Cell Biol 2014;204(5):669–82.
[[36]]
Swift J, Discher DE. The nuclear lamina is mechano-responsive to ECM elasticity in mature tissue. J Cell Sci 2014;127(14):3005–15.
[[37]]
Gong Z, Szczesny SE, Caliari SR, Charrier EE, Chaudhuri O, Cao X, et al. Matching material and cellular timescales maximizes cell spreading on viscoelastic substrates. Proc Natl Acad Sci USA 2018;115(12):E2686–95.
[[38]]
Wolf K, Lindert MT, Krause M, Alexander S, Riet JT, Willis AL, et al. Physical limits of cell migration: control by ECM space and nuclear deformation and tuning by proteolysis and traction force. J Cell Biol 2013;201(7):1069–84.
[[39]]
Wisdom KM, Adebowale K, Chang J, Lee JY, Nam S, Desai R, et al. Matrix mechanical plasticity regulates cancer cell migration through confining microenvironments. Nat Commun 2018;9(1):4144.
[[40]]
Chaudhuri O, Cooper-White J, Janmey PA, Mooney DJ, Shenoy VB. Effects of extracellular matrix viscoelasticity on cellular behaviour. Nature 2020;584 (7822):535–46.
[[41]]
Chaudhuri O. Viscoelastic hydrogels for 3D cell culture. Biomater Sci 2017;5 (8):1480–90.
[[42]]
Chaudhuri O, Gu L, Klumpers D, Darnell M, Bencherif SA, Weaver JC, et al. Hydrogels with tunable stress relaxation regulate stem cell fate and activity. Nat Mater 2016;15(3):326–34.
[[43]]
Cameron AR, Frith JE, Cooper-White JJ. The influence of substrate creep on mesenchymal stem cell behaviour and phenotype. Biomaterials 2011;32 (26):5979–93.
[[44]]
Charrier EE, Pogoda K, Wells RG, Janmey PA. Control of cell morphology and differentiation by substrates with independently tunable elasticity and viscous dissipation. Nat Commun 2018;9(1):449.
[[45]]
Chaudhuri O, Gu L, Darnell M, Klumpers D, Bencherif SA, Weaver JC, et al. Substrate stress relaxation regulates cell spreading. Nat Commun 2015;6: 6365.
[[46]]
Chopra A, Murray ME, Byfield FJ, Mendez MG, Halleluyan R, Restle DJ, et al. Augmentation of integrin-mediated mechanotransduction by hyaluronic acid. Biomaterials 2014;35(1):71–82.
[[47]]
Ranga A, Lutolf MP, Hilborn J, Ossipov DA. Hyaluronic acid hydrogels formed in situ by transglutaminase-catalyzed reaction. Biomacromolecules 2016;17 (5):1553–60.
[[48]]
Kleine-Brüggeney H, van Vliet LD, Mulas C, Gielen F, Agley CC, Silva JCR, et al. Longterm perfusion culture of monoclonal embryonic stem cells in 3D hydrogel beads for continuous optical analysis of differentiation. Small 2019;15(5):1804576.
[[49]]
Pavel M, Renna M, Park SJ, Menzies FM, Ricketts T, Füllgrabe J, et al. Contact inhibition controls cell survival and proliferation via YAP/TAZ-autophagy axis. Nat Commun 2018;9(1):2961.
[[50]]
Rezk R, Jia BZ, Wendler A, Dimov I, Watts C, Markaki AE, et al. Spatial heterogeneity of cell-matrix adhesive forces predicts human glioblastoma migration. Neurooncol Adv 2020;2(1):vdaa081.
[[51]]
Grundy TJ, De Leon E, Griffin KR, Stringer BW, Day BW, Fabry B, et al. Differential response of patient-derived primary glioblastoma cells to environmental stiffness. Sci Rep 2016;6(1):23353.
[[52]]
Plodinec M, Loparic M, Monnier CA, Obermann EC, Zanetti-Dallenbach R, Oertle P, et al. The nanomechanical signature of breast cancer. Nat Nanotechnol 2012;7(11):757–65.
[[53]]
Swaminathan V, Mythreye K, O’Brien ET, Berchuck A, Blobe GC, Superfine R. Mechanical stiffness grades metastatic potential in patient tumor cells and in cancer cell lines. Cancer Res 2011;71(15):5075–80.
[[54]]
Kenny PA, Bissell MJ. Tumor reversion: correction of malignant behavior by microenvironmental cues. Int J Cancer 2003;107(5):688–95.
[[55]]
Berger AJ, Renner CM, Hale I, Yang X, Ponik SM, Weisman PS, et al. Scaffold stiffness influences breast cancer cell invasion via EGFR-linked Mena upregulation and matrix remodeling. Matrix Biol 2020;85–86:80–93.
PDF(338 KB)

Accesses

Citations

Detail

Sections
Recommended

/