386
Views
12
CrossRef citations to date
0
Altmetric
Review

Vascular and extracellular matrix remodeling by physical approaches to improve drug delivery at the tumor site

, ORCID Icon, ORCID Icon & ORCID Icon
Pages 1703-1726 | Received 08 Jul 2020, Accepted 21 Aug 2020, Published online: 04 Sep 2020

References

  • Li Y, Wang J, Wientjes MG, et al. Delivery of nanomedicines to extracellular and intracellular compartments of a solid tumor. Adv Drug Deliv Rev. 2012;64:29–39.
  • Marcucci F, Corti A. How to improve exposure of tumor cells to drugs — promoter drugs increase tumor uptake and penetration of effector drugs. Adv Drug Deliv Rev. 2012;64:53–68.
  • Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. nature. 2000;407:249.
  • Matsumura Y, Maeda H, New A. Concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. Cancer Res. 1986;46:6387–6392.
  • Nichols JW, Bae YHEPR. Evidence and fallacy. J Control Release. 2014;190:451–464.
  • Heldin C-H, Rubin K, Pietras K, et al. High interstitial fluid pressure - an obstacle in cancer therapy. Nat Rev Cancer. 2004;4:806–813.
  • Ariffin AB, Forde PF, Jahangeer S, et al. Releasing pressure in tumors: what do we know so far and where do we go from here? A review. Cancer Res. 2014;74:2655–2662.
  • Ojha T, Pathak V, Shi Y, et al. Pharmacological and physical vessel modulation strategies to improve EPR-mediated drug targeting to tumors. Adv Drug Deliv Rev. 2017;119:44–60.
  • Dhaliwal A, Zheng G. Improving accessibility of EPR-insensitive tumor phenotypes using EPR-adaptive strategies: designing a new perspective in nanomedicine delivery. Theranostics. 2019;9:8091.
  • Yang S, Gao H. Nanoparticles for modulating tumor microenvironment to improve drug delivery and tumor therapy. Pharmacol Res. 2017;126:97–108.
  • Zhou Y, Chen X, Cao J, et al. Overcoming the biological barriers in the tumor microenvironment for improving drug delivery and efficacy. J Mater Chem B. 2020;8:6765–6781.
  • Henke E, Nandigama R, Ergün S. Extracellular Matrix in the tumor microenvironment and its impact on cancer therapy. Front Mol Biosci. 2020;6:160.
  • Paszek MJ, Zahir N, Johnson KR, et al. Tensional homeostasis and the malignant phenotype. Cancer Cell. 2005;8:241–254.
  • Kauppila S, Stenbäck F, Risteli J, et al. Aberrant type I and type III collagen gene expression in human breast cancer in vivo. J Pathol J Pathol Soc G B Irel. 1998;186:262–268.
  • Theocharis AD, Skandalis SS, Gialeli C, et al. Extracellular matrix structure. Adv Drug Deliv Rev. 2016;97:4–27.
  • Lu P, Weaver VM, Werb Z. The extracellular matrix: A dynamic niche in cancer progression. J Cell Biol. 2012;196:395–406.
  • Netti PA, Berk DA, Swartz MA, et al. Role of extracellular matrix assembly in interstitial transport in solid tumors. Cancer Res. 2000;60:2497–2503.
  • Jain RK. Normalizing Tumor microenvironment to treat cancer: bench to bedside to biomarkers. J Clin Oncol. 2013;31:2205–2218.
  • Burgstaller G, Oehrle B, Gerckens M, et al. The instructive extracellular matrix of the lung: basic composition and alterations in chronic lung disease. Eur Respir J. 2017;50:1601805.
  • Minchinton AI, Tannock IF. Drug penetration in solid tumours. Nat Rev Cancer. 2006;6:583–592.
  • Brown E, McKee T, di.Tomaso E, et al. Dynamic imaging of collagen and its modulation in tumors in vivo using second-harmonic generation. Nat Med. 2003;9:796–800.
  • Dolor A, Szoka FC. Digesting a path forward: the utility of collagenase tumor treatment for improved drug delivery. Mol Pharm. 2018;15:2069–2083.
  • Chauhan VP, Martin JD, Liu H, et al. Angiotensin inhibition enhances drug delivery and potentiates chemotherapy by decompressing tumour blood vessels. Nat Commun. 2013;4:1–11.
  • Barry-Hamilton V, Spangler R, Marshall D, et al. Allosteric inhibition of lysyl oxidase–like-2 impedes the development of a pathologic microenvironment. Nat Med. 2010;16:1009.
  • Coussens LM, Fingleton B, Matrisian LM. Matrix metalloproteinase inhibitors and cancer—trials and tribulations. Science. 2002;295:2387–2392.
  • Diop-Frimpong B, Chauhan VP, Krane S, et al. Losartan inhibits collagen I synthesis and improves the distribution and efficacy of nanotherapeutics in tumors. Proc Natl Acad Sci. 2011;108:2909–2914.
  • Khawar IA, Kim JH, Kuh H-J. Improving drug delivery to solid tumors: priming the tumor microenvironment. J Control Release. 2015;201:78–89.
  • Nakamura Y, Mochida A, Choyke PL, et al. Nanodrug delivery: is the enhanced permeability and retention effect sufficient for curing cancer? Bioconjug Chem. 2016;27:2225–2238.
  • Wang Y, Gan G, Wang B, et al. Cancer-associated Fibroblasts promote irradiated cancer cell recovery through autophagy. EBioMedicine. 2017;17:45–56.
  • Hanahan D, Weinberg RA. Hallmarks of Cancer: the next generation. Cell. 2011;144:646–674.
  • Fuks Z, Kolesnick R. Engaging the vascular component of the tumor response. Cancer Cell. 2005;8:89–91.
  • Potiron VA, Abderrahmani R, Clément-Colmou K, et al. Improved functionality of the vasculature during conventionally fractionated radiation therapy of prostate cancer. PloS One. 2013;8:e84076.
  • Garcia-Barros M, Paris F, Cordon-Cardo C, et al. Tumor response to radiotherapy regulated by endothelial cell apoptosis. Science. 2003;300:1155–1159.
  • DuRoss AN, Neufeld MJ, Rana S, et al. Integrating nanomedicine into clinical radiotherapy regimens. Adv Drug Deliv Rev. 2019;144:35–56.
  • Schwickert HC, Stiskal M, Roberts TP, et al. Contrast-enhanced MR imaging assessment of tumor capillary permeability: effect of irradiation on delivery of chemotherapy. Radiology. 1996;198:893–898.
  • Moding EJ, Kastan MB, Kirsch DG. Strategies for optimizing the response of cancer and normal tissues to radiation. Nat Rev Drug Discov. 2013;12:526–542.
  • Stapleton S, Jaffray D, Milosevic M. Radiation effects on the tumor microenvironment: implications for nanomedicine delivery. Adv Drug Deliv Rev. 2017;109:119–130.
  • Shi J, Kantoff PW, Wooster R, et al. Cancer nanomedicine: progress, challenges and opportunities. Nat Rev Cancer. 2017;17:20.
  • Roh HD, Boucher Y, Kalnicki S, et al. Interstitial hypertension in carcinoma of uterine cervix in patients: possible correlation with tumor oxygenation and radiation response. Cancer Res. 1991;51:6695–6698.
  • Znati CA, Rosenstein M, Boucher Y, et al. Effect of radiation on interstitial fluid pressure and oxygenation in a human tumor xenograft. Cancer Res. 1996;56:964–968.
  • Giustini AJ, Petryk AA, Hoopes PJ. Ionizing radiation increases systemic nanoparticle tumor accumulation. Nanomed Nanotechnol Biol Med. 2012;8:818–821.
  • Moeller BJ, Cao Y, Li CY, et al. Radiation activates HIF-1 to regulate vascular radiosensitivity in tumors: role of reoxygenation, free radicals, and stress granules. Cancer Cell. 2004;5:429–441.
  • Dewhirst MW, Cao Y, Moeller B. Cycling hypoxia and free radicals regulate angiogenesis and radiotherapy response. Nat Rev Cancer. 2008;8:425–437.
  • Lee C-G, Heijn M, Di Tomaso E, et al. Anti-vascular endothelial growth factor treatment augments tumor radiation response under normoxic or hypoxic conditions. Cancer Res. 2000;60:5565–5570.
  • Tachiiri S, Katagiri T, Tsunoda T, et al. Analysis of gene-expression profiles after gamma irradiation of normal human fibroblasts. Int J Radiat Oncol Biol Phys. 2006;64:272–279.
  • Qian L-W, Mizumoto K, Urashima T, et al. Radiation-induced increase in invasive potential of human pancreatic cancer cells and its blockade by a matrix metalloproteinase inhibitor, CGS27023. Clin Cancer Res. 2002;8:1223–1227.
  • Kumar A, Collins HM, Scholefield JH, et al. Increased type-IV collagenase (MMP-2 and MMP-9) activity following preoperative radiotherapy in rectal cancer. Br J Cancer. 2000;82:960–965.
  • Angenete E, Öresland T, Falk P, et al. Preoperative radiotherapy and extracellular matrix remodeling in rectal mucosa and tumour matrix metalloproteinases and plasminogen components. Acta Oncol. 2009;48:1144–1151.
  • Chetty C, Bhoopathi P, Rao JS, et al. Inhibition of matrix metalloproteinase‐2 enhances radiosensitivity by abrogating radiation‐induced FoxM1‐mediated G2/M arrest in A549 lung cancer cells. Int J Cancer. 2009;124:2468–2477.
  • Paquette B, Baptiste C, Therriault H, et al. In vitro irradiation of basement membrane enhances the invasiveness of breast cancer cells. Br J Cancer. 2007;97:1505–1512.
  • Speake WJ, Dean RA, Kumar A, et al. Radiation induced MMP expression from rectal cancer is short lived but contributes to in vitro invasion. Eur J Surg Oncol EJSO. 2005;31:869–874.
  • Shankar A, Kumar S, Iskander A, et al. Subcurative radiation significantly increases cell proliferation, invasion, and migration of primary glioblastoma multiforme in vivo. Chin J Cancer. 2014;33:148.
  • Ager EI, Kozin SV, Kirkpatrick ND, et al. Blockade of MMP14 activity in murine breast carcinomas: implications for macrophages, vessels, and radiotherapy. JNCI J Natl Cancer Inst. 2015;4:107.
  • Strup-Perrot C, M-C V-B, Vandamme M, et al. Expression and activation of MMP −2, −3, −9, −14 are induced in rat colon after abdominal X-irradiation. Scand J Gastroenterol. 2006;41:60–70.
  • Angenete E, Langenskiöld M, Falk P, et al. Matrix metalloproteinases in rectal mucosa, tumour and plasma: response after preoperative irradiation. Int J Colorectal Dis. 2007;22:667–674.
  • Angenete E, Langenskiöld M, Palmgren I, et al. uPA and PAI-1 in rectal cancer—relationship to radiotherapy and clinical outcome. J Surg Res. 2009;153:46–53.
  • Kagan HM, Li W. Lysyl oxidase: properties, specificity, and biological roles inside and outside of the cell. J Cell Biochem. 2003;88:660–672.
  • Erler JT, Giaccia AJ. Lysyl oxidase mediates hypoxic control of metastasis. Cancer Res. 2006;66:10238–10241.
  • Egeblad M, Rasch MG, Weaver VM. Dynamic interplay between the collagen scaffold and tumor evolution. Curr Opin Cell Biol. 2010;22:697–706.
  • Shen CJ, Sharma A, Vuong D-V, et al. Ionizing radiation induces tumor cell lysyl oxidase secretion. BMC Cancer. 2014;14:532.
  • Xie P, Yu H, Wang F, et al. Inhibition of LOXL2 enhances the radiosensitivity of castration-resistant prostate cancer cells associated with the reversal of the emt process. Catalá A, editor. BioMed Res Int. 2019;2019:4012590.
  • Chen HH, Kuo MT. Improving radiotherapy in cancer treatment: promises and challenges. Oncotarget. 2017;8:62742.
  • Li J, Shang W, Li Y, et al. Advanced nanomaterials targeting hypoxia to enhance radiotherapy. Int J Nanomedicine. 2018;13:5925.
  • Lammers T, Peschke P, Kühnlein R, et al. Effect of radiotherapy and hyperthermia on the tumor accumulation of HPMA copolymer-based drug delivery systems. J Control Release. 2007;117:333–341.
  • Lammers T, Subr V, Peschke P, et al. Image-guided and passively tumour-targeted polymeric nanomedicines for radiochemotherapy. Br J Cancer. 2008;99:900–910.
  • Davies CDL, Lundstrøm LM, Frengen J, et al. Radiation improves the distribution and uptake of liposomal doxorubicin (caelyx) in human osteosarcoma xenografts. Cancer Res. 2004;64:547–553.
  • Stapleton S, Dunne M, Milosevic M, et al. Radiation and heat improve the delivery and efficacy of Nanotherapeutics by modulating Intratumoral fluid dynamics. ACS Nano. 2018;12:7583–7600.
  • Miller MA, Chandra R, Cuccarese MF, et al. Radiation therapy primes tumors for nanotherapeutic delivery via macrophage-mediated vascular bursts. Sci Transl Med. 2017;9:392.
  • Koukourakis MI, Koukouraki S, Giatromanolaki A, et al. Liposomal doxorubicin and conventionally fractionated radiotherapy in the treatment of locally advanced non-small-cell lung cancer and head and neck cancer. J Clin Oncol. 1999;17:3512–3521.
  • Rosenthal DI, Yom SS, Liu L, et al. A phase I study of SPI-077 (Stealth® liposomal cisplatin) concurrent with radiation therapy for locally advanced head and neck cancer. Invest New Drugs. 2002;20:343–349.
  • Dipetrillo T, Milas L, Evans D, et al. Paclitaxel poliglumex (PPX-Xyotax) and concurrent radiation for esophageal and gastric cancer: a phase I study. Am J Clin Oncol. 2006;29:376–379.
  • Zee J, van D, Vujaskovic Z, et al. The Kadota Fund International Forum 2004–Clinical group consensus*. Int J Hyperthermia. 2008;24:111–122.
  • Koning GA, Eggermont AMM, Lindner LH, et al. Hyperthermia and thermosensitive liposomes for improved delivery of chemotherapeutic drugs to solid tumors. Pharm Res. 2010;27:1750–1754.
  • Hu Q, Huang Z, Duan Y, et al. Reprogramming tumor microenvironment with photothermal therapy. Bioconjug Chem. 2020;31:1268–1278.
  • Datta NR, Kok HP, Crezee H, et al. Integrating Loco-regional hyperthermia into the current oncology practice: SWOT and TOWS analyses. Front Oncol. 2020;10:819.
  • Vaupel PW, Kelleher DK. Pathophysiological and vascular characteristics of tumours and their importance for hyperthermia: heterogeneity is the key issue. Int J Hyperthermia. 2010;26:211–223.
  • Kirui DK, Koay EJ, Guo X, et al. Tumor vascular permeabilization using localized mild hyperthermia to improve macromolecule transport. Nanomed Nanotechnol Biol Med. 2014;10:1487–1496.
  • Melancon MP, Elliott AM, Shetty A, et al. Near-infrared light modulated photothermal effect increases vascular perfusion and enhances polymeric drug delivery. J Control Release. 2011;156:265–272.
  • Lapin NA, Krzykawska-Serda M, Dilliard S, et al. The effects of non-invasive radiofrequency electric field hyperthermia on biotransport and biodistribution of fluorescent [60]fullerene derivative in a murine orthotopic model of breast adenocarcinoma. J Control Release. 2017;260:92–99.
  • Li L, TLM TH, Bolkestein M, et al. Improved intratumoral nanoparticle extravasation and penetration by mild hyperthermia. J Control Release. 2013;167:130–137.
  • Gao H, Bi Y, Chen J, et al. Near-infrared light-triggered switchable nanoparticles for targeted chemo/photothermal cancer therapy. ACS Appl Mater Interfaces. 2016;8:15103–15112.
  • Baronzio GF, Galante F, Gramaglia A, et al. Tumor microcirculation and its significance in therapy: possible role of omega-3 fatty acids as rheological modifiers. Med Hypotheses. 1998;50:175–182.
  • Fite BZ, Kheirolomoom A, Foiret JL, et al. Dynamic contrast enhanced MRI detects changes in vascular transport rate constants following treatment with thermally-sensitive liposomal doxorubicin. J Control Release. 2017;256:203–213.
  • Song CW, Park HJ, Lee CK, et al. Implications of increased tumor blood flow and oxygenation caused by mild temperature hyperthermia in tumor treatment. Int J Hyperthermia. 2005;21:761–767.
  • Corr SJ, Shamsudeen S, Vergara LA, et al. A new imaging platform for visualizing biological effects of non-invasive radiofrequency electric-field cancer hyperthermia. Muñoz-Barrutia A, editor. Plos One. 2015;10:e0136382.
  • Marangon I, Silva AAK, Guilbert T, et al. Tumor Stiffening, a Key Determinant of Tumor Progression, is Reversed by Nanomaterial-Induced Photothermal Therapy. Theranostics. 2017;7:329–343.
  • Kolosnjaj-Tabi J, Marangon I, Nicolas-Boluda A, et al. Nanoparticle-based hyperthermia, a local treatment modulating the tumor extracellular matrix. Pharmacol Res. 2017;126:123–137.
  • Yudina A, Moonen C. Ultrasound-induced cell permeabilisation and hyperthermia: strategies for local delivery of compounds with intracellular mode of action. Int J Hyperthermia. 2012;28:311–319.
  • Ryu JS, Raucher D. Elastin-like polypeptides: the influence of its molecular weight on local hyperthermia-induced tumor accumulation. Eur J Pharm Biopharm. 2014;88:382–389.
  • Kolosnjaj-Tabi J, Di Corato R, Lartigue L, et al. Heat-generating iron oxide nanocubes: subtle “destructurators” of the tumoral microenvironment. ACS Nano. 2014;8:4268–4283.
  • Thakkar S, Sharma D, Kalia K, et al. Tumor microenvironment targeted nanotherapeutics for cancer therapy and diagnosis: a review. Acta Biomater. 2020;101:43–68.
  • Lokerse WJM, Bolkestein M, Dalm SU, et al. Comparing the therapeutic potential of thermosensitive liposomes and hyperthermia in two distinct subtypes of breast cancer. J Control Release. 2017;258:34–42.
  • Raeesi V, Chan WCW. Improving nanoparticle diffusion through tumor collagen matrix by photo-thermal gold nanorods. Nanoscale. 2016;8:12524–12530.
  • Piehler S, Wucherpfennig L, Tansi FL, et al. Hyperthermia affects collagen fiber architecture and induces apoptosis in pancreatic and fibroblast tumor hetero-spheroids in vitro. Nanomed Nanotechnol Biol Med. 2020;28:102183.
  • Kolosnjaj-Tabi J, Kralj S, Griseti E, et al. Magnetic silica-coated iron oxide nanochains as photothermal agents, disrupting the extracellular matrix, and eradicating cancer cells. Cancers (Basel). 2019;11:2040.
  • Ta T, Bartolak-Suki E, Park E-J, et al. Localized delivery of doxorubicin in vivo from polymer-modified thermosensitive liposomes with MR-guided focused ultrasound-mediated heating. J Control Release. 2014;194:71–81.
  • Beola L, Asín L, Fratila RM, et al. Dual role of magnetic nanoparticles as intracellular hotspots and extracellular matrix disruptors triggered by magnetic hyperthermia in 3D cell culture models. ACS Appl Mater Interfaces. 2018;10:44301–44313.
  • Li J, Xie C, Huang J, et al. Semiconducting polymer nanoenzymes with photothermic activity for enhanced cancer therapy. Angew Chem Int Ed. 2018;57:3995–3998.
  • Ramos-Docampo MA, Fernández-Medina M, Taipaleenmäki E, et al. Microswimmers with heat delivery capacity for 3D Cell spheroid penetration. ACS Nano. 2019;13:12192–12205.
  • Sebeke L, Deenen DA, Maljaars E, et al. Model predictive control for MR-HIFU-mediated, uniform hyperthermia. Int J Hyperthermia. 2019;36:1039–1049.
  • Hancock HA, Smith LH, Cuesta J, et al. Investigations into Pulsed high-intensity focused ultrasound–enhanced delivery: preliminary evidence for a novel mechanism. Ultrasound Med Biol. 2009;35:1722–1736.
  • Lee S, Han H, Koo H, et al. Extracellular matrix remodeling in vivo for enhancing tumor-targeting efficiency of nanoparticle drug carriers using the pulsed high intensity focused ultrasound. J Control Release. 2017;263:68–78.
  • Jiang T, Zhang B, Shen S, et al. Tumor microenvironment modulation by cyclopamine improved photothermal therapy of biomimetic gold nanorods for pancreatic ductal adenocarcinomas. ACS Appl Mater Interfaces. 2017;9:31497–31508.
  • Tan T, Hu H, Wang H, et al. Bioinspired lipoproteins-mediated photothermia remodels tumor stroma to improve cancer cell accessibility of second nanoparticles. Nat Commun. 2019;10:3322.
  • Sorrin AJ, Ruhi MK, Ferlic NA, et al. Photodynamic therapy and the biophysics of the tumor microenvironment. Photochem Photobiol. 2020;96:232–259.
  • Krammer B. Vascular effects of photodynamic therapy. Anticancer Res. 2001;21:4271–4277.
  • Chen B, Pogue BW, Hoopes PJ, et al. Vascular and cellular targeting for photodynamic therapy. Crit Rev Eukaryot Gene Expr. 2006;16:279–305.
  • Fingar VH, Wieman TJ, Wiehle SA, et al. The role of microvascular damage in photodynamic therapy: the effect of treatment on vessel constriction, permeability, and leukocyte adhesion. Cancer Res. 1992;52:4914–4921.
  • Fingar VH, Wieman TJ, Doak KW. Changes in tumor interstitial pressure induced by photodynamic therapy. Photochem Photobiol. 1991;53:763–768.
  • Malik Z, Faraggi A, Savion N. Ultrastructural damage in photosensitized endothelial cells: dependence on hematoporphyrin delivery pathways. J Photochem Photobiol B. 1992;14:359–368.
  • Leunig A, Staub F, Plesnila N, et al. Effect of photodynamic treatment of human endothelial cells on cell volume and cell viability. Int J Oncol. 1996;8:1217–1221.
  • Sporn LA, Foster TH. Photofrin and Light induces microtubule depolymerization in cultured human endothelial cells. Cancer Res. 1992;52:3443–3448.
  • Gil M, Bieniasz M, Seshadri M, et al. Photodynamic therapy augments the efficacy of oncolytic vaccinia virus against primary and metastatic tumours in mice. Br J Cancer. 2011;105:1512–1521.
  • Snyder JW, Greco WR, Bellnier DA, et al. Photodynamic therapy: a means to enhanced drug delivery to tumors. Cancer Res. 2003;63:8126–8131.
  • Cheng C, Debefve E, Haouala A, et al. Photodynamic therapy selectively enhances liposomal doxorubicin uptake in sarcoma tumors to rodent lungs. Lasers Surg Med. 2010;42:391–399.
  • Perentes JY, Wang Y, Wang X, et al. Low-dose vascular photodynamic therapy decreases tumor interstitial fluid pressure, which promotes liposomal doxorubicin distribution in a murine sarcoma metastasis model. Transl Oncol. 2014;7:393–399.
  • Sano K, Nakajima T, Choyke PL, et al. The effect of photoimmunotherapy followed by liposomal daunorubicin in a mixed tumor model: a demonstration of the super-enhanced permeability and retention effect after photoimmunotherapy. Mol Cancer Ther. 2014;13:426–432.
  • Luo D, Carter KA, Miranda D, et al. Chemophototherapy: an emerging treatment option for solid tumors. Adv Sci. 2017;4:1600106.
  • Lv T, Huang Z-F, Wang H-W, et al. Evaluation of collagen alteration after topical photodynamic therapy (PDT) using second harmonic generation (SHG) microscopy – in vivo study in a mouse model. Photodiagnosis Photodyn Ther. 2012;9:164–169.
  • Mills SJ, Farrar MD, Ashcroft GS, et al. Topical photodynamic therapy following excisional wounding of human skin increases production of transforming growth factor-β3 and matrix metalloproteinases 1 and 9, with associated improvement in dermal matrix organization. Br J Dermatol. 2014;171:55–62.
  • Ryu A-R, Lee M-Y. Chlorin e6-mediated photodynamic therapy promotes collagen production and suppresses MMPs expression via modulating AP-1 signaling in P. acnes-stimulated HaCaT cells. Photodiagnosis Photodyn Ther. 2017;20:71–77.
  • Yova DM, Hovhannisyan VA, Theodossiou T. Photochemical effects and hypericin photosensitized processes in collagen. J Biomed Opt. 2001;6:52–58.
  • Hovhannisyan V, Guo HW, Hovhannisyan A, et al. Photo-induced processes in collagen-hypericin system revealed by fluorescence spectroscopy and multiphoton microscopy. Biomed Opt Express. 2014;5:1355–1362.
  • Hovhannisyan V, Hovhannisyan A, Ghukasyan V, et al. Hypericin-mediated selective photomodification of connective tissues. Appl Phys Lett. 2014;105:263701.
  • Ferrario A, Chantrain CF, von Tiehl K, et al. The matrix metalloproteinase inhibitor prinomastat enhances photodynamic therapy responsiveness in a mouse tumor model. Cancer Res. 2004;64:2328–2332.
  • Karrer S, Bosserhoff AK, Weiderer P, et al. Influence of 5-aminolevulinic acid and red light on collagen metabolism of human dermal fibroblasts. J Invest Dermatol. 2003;120:325–331.
  • Zhang C, Wang J, Chou A, et al. Photodynamic therapy induces antifibrotic alterations in primary human vocal fold fibroblasts. Laryngoscope. 2018;128:E323–E331.
  • Biel MA. Photodynamic therapy treatment of early oral and laryngeal cancers. Photochem Photobiol. 2007;83:1063–1068.
  • Ihsanullah KM, Kumar BN, Zhao Y, et al. Stepwise-activatable hypoxia triggered nanocarrier-based photodynamic therapy for effective synergistic bioreductive chemotherapy. Biomaterials. 2020;245:119982.
  • Tachibana K, Uchida T, Ogawa K, et al. Induction of cell-membrane porosity by ultrasound. Lancet. 1999;353:1409.
  • Frenkel V. Ultrasound mediated delivery of drugs and genes to solid tumors. Adv Drug Deliv Rev. 2008;60:1193–1208.
  • Ferrara K, Pollard R, Borden M. Ultrasound microbubble contrast agents: fundamentals and application to gene and drug delivery. Annu Rev Biomed Eng. 2007;9:415–447.
  • Hernot S, Klibanov AL. Microbubbles in ultrasound-triggered drug and gene delivery. Adv Drug Deliv Rev. 2008;60:1153–1166.
  • Lentacker I, De Cock I, Deckers R, et al. Understanding ultrasound induced sonoporation: definitions and underlying mechanisms. Adv Drug Deliv Rev. 2014;72:49–64.
  • Tharkar P, Varanasi R, Wong WSF, et al. Nano-enhanced drug delivery and therapeutic ultrasound for cancer treatment and beyond. Front Bioeng Biotechnol. 2019;7:324.
  • Skyba DM, Price RJ, Linka AZ, et al. Direct in vivo visualization of intravascular destruction of microbubbles by ultrasound and its local effects on tissue. Circulation. 1998;98:290–293.
  • Chen H, Kreider W, Brayman AA, et al. Blood vessel deformations on microsecond time scales by ultrasonic cavitation. Phys Rev Lett. 2011;106:034301.
  • Chen H, Brayman AA, Evan AP, et al. Preliminary observations on the spatial correlation between short-burst microbubble oscillations and vascular bioeffects. Ultrasound Med Biol. 2012;38:2151–2162.
  • Wamel AV, Kooiman K, Emmer M, et al. Ultrasound microbubble induced endothelial cell permeability. J Control Release. 2006;116:e100–e102.
  • Patil AV, Rychak JJ, Klibanov AL, et al. Real-time technique for improving molecular imaging and guiding drug delivery in large blood vessels: in vitro and ex vivo results. Mol Imaging. 2011;10:7290. 2011.00002
  • Meijering BDM, Henning RH, Gilst WHV, et al. Optimization of ultrasound and microbubbles targeted gene delivery to cultured primary endothelial cells. J Drug Target. 2007;15:664–671.
  • Juffermans LJM, Meijering BDM, Henning RH, et al. Ultrasound and microbubble-targeted delivery of small interfering RNA into primary endothelial cells is more effective than delivery of plasmid DNA. Ultrasound Med Biol. 2014;40:532–540.
  • Meijering BDM, Juffermans LJM, van Wamel A, et al. Ultrasound and microbubble-targeted delivery of macromolecules is regulated by induction of endocytosis and pore formation. Circ Res. 2009;104:679–687.
  • Juffermans LJM, van Dijk A, Jongenelen CAM, et al. Ultrasound and microbubble-induced intra- and intercellular bioeffects in primary endothelial cells. Ultrasound Med Biol. 2009;35:1917–1927.
  • Bekeredjian R, Kroll RD, Fein E, et al. Ultrasound targeted microbubble destruction increases capillary permeability in hepatomas. Ultrasound Med Biol. 2007;33:1592–1598.
  • Mukherjee D, Wong J, Griffin B, et al. Ten-fold augmentation of endothelial uptake of vascular endothelial growth factor with ultrasound after systemic administration. J Am Coll Cardiol. 2000;35:1678–1686.
  • Cochran MC, Eisenbrey JR, Soulen MC, et al. Disposition of ultrasound sensitive polymeric drug carrier in a rat hepatocellular carcinoma model. Acad Radiol. 2011;18:1341–1348.
  • Yan F, Li L, Deng Z, et al. Paclitaxel-liposome–microbubble complexes as ultrasound-triggered therapeutic drug delivery carriers. J Control Release. 2013;166:246–255.
  • Qin J, Wang T-Y, Willmann JK. Sonoporation: applications for cancer therapy. Ther Ultrasound. 2016;880:263–291. Springer.
  • Chowdhury SM, Wang T-Y, Bachawal S, et al. Ultrasound-guided therapeutic modulation of hepatocellular carcinoma using complementary microRNAs. J Control Release. 2016;238:272–280.
  • Devulapally R, Sekar NM, Sekar TV, et al. Polymer nanoparticles mediated codelivery of antimiR-10b and antimiR-21 for achieving triple negative breast cancer therapy. ACS Nano. 2015;9:2290–2302.
  • Han H, Kim D, Jang Y, et al. Focused ultrasound-triggered chemo-gene therapy with multifunctional nanocomplex for enhancing therapeutic efficacy. J Control Release. 2020;322:346–356.
  • Kolosnjaj-Tabi J, Gibot L, Fourquaux I, et al. Electric field-responsive nanoparticles and electric fields: physical, chemical, biological mechanisms and therapeutic prospects. Adv Drug Deliv Rev. 2019;138:56–67.
  • Yarmush ML, Golberg A, Serša G, et al. Electroporation-based technologies for medicine: principles, applications, and challenges. Annu Rev Biomed Eng. 2014;16:295–320.
  • Gehl J, Sersa G, Matthiessen LW, et al. Updated standard operating procedures for electrochemotherapy of cutaneous tumours and skin metastases. Acta Oncol. 2018;57:874–882.
  • Ramirez L, Orlowski S, An D, et al. Electrochemotherapy on liver tumours in rabbits. Br J Cancer. 1998;77:2104–2111.
  • Sersa G, Beravs K, Cemazar M, et al. Contrast enhanced MRI assessment of tumor blood volume after application of electric pulses. Electro- Magnetobiology. 1998;17:299–306.
  • Gehl J, Skovsgaard T, Mir LM. Vascular reactions to in vivo electroporation: characterization and consequences for drug and gene delivery. Biochim Biophys Acta BBA - Gen Subj. 2002;1569:51–58.
  • Sersa G, Jarm T, Kotnik T, et al. Vascular disrupting action of electroporation and electrochemotherapy with bleomycin in murine sarcoma. Br J Cancer. 2008;98:388–398.
  • Gehl J, Geertsen PF. Efficient palliation of haemorrhaging malignant melanoma skin metastases by electrochemotherapy. Melanoma Res. 2000;10:585–589.
  • Markelc B, Bellard E, Sersa G, et al. In vivo molecular imaging and histological analysis of changes induced by electric pulses used for plasmid DNA electrotransfer to the skin: a study in a dorsal window chamber in mice. J Membr Biol. 2012;245:545–554.
  • Markelc B, Bellard E, Sersa G, et al. Increased permeability of blood vessels after reversible electroporation is facilitated by alterations in endothelial cell-to-cell junctions. J Control Release. 2018;276:30–41.
  • Sersa G, Cemazar M, Miklavcic D, et al. Tumor blood flow modifying effect of electrochemotherapy with bleomycin. Anticancer Res. 1999;19:4017–4022.
  • Sersa G, Cemazar M, Parkins CS, et al. Tumour blood flow changes induced by application of electric pulses. Eur J Cancer. 1999;35:672–677.
  • Sersa G, Krzic M, Sentjurc M, et al. Reduced blood flow and oxygenation in SA-1 tumours after electrochemotherapy with cisplatin. Br J Cancer. 2002;87:1047–1054.
  • Kanthou C, Kranjc S, Sersa G, et al. The endothelial cytoskeleton as a target of electroporation-based therapies. Mol Cancer Ther. 2006;5:3145–3152.
  • Meulenberg CJW, Todorovic V, Cemazar M. Differential cellular effects of electroporation and electrochemotherapy in monolayers of human microvascular endothelial cells. PloS One. 2012;7:e52713.
  • Kodama H, Shamay Y, Kimura Y, et al. Electroporation-induced changes in tumor vasculature and microenvironment can promote the delivery and increase the efficacy of sorafenib nanoparticles. Bioelectrochemistry Amst Neth. 2019;130:107328.
  • Kis E, Baltás E, Á K, et al. Successful treatment of multiple basaliomas with bleomycin-based electrochemotherapy: a case series of three patients with Gorlin-Goltz Syndrome. Acta Derm Venereol. 2012;92:648–651.
  • Campana LG, Mocellin S, Basso M, et al. Bleomycin-based electrochemotherapy: clinical outcome from a single institution’s experience with 52 patients. Ann Surg Oncol. 2009;16:191–199.
  • Maor E, Ivorra A, Leor J, et al. The effect of irreversible electroporation on blood vessels. Technol Cancer Res Treat. 2007;6:307–312.
  • Golberg A, Villiger M, Broelsch GF, et al. Skin regeneration with all accessory organs following ablation with irreversible electroporation. J Tissue Eng Regen Med. 2018;12:98–113.
  • Weltmann KD, Von Woedtke T. Plasma medicine—current state of research and medical application. Plasma Phys Control Fusion. 2016;59:014031.
  • Privat-Maldonado A, Bengtson C, Razzokov J, et al. Modifying the tumour microenvironment: challenges and future perspectives for anticancer plasma treatments. Cancers (Basel). 2019;11:1920.
  • Arndt S, Unger P, Berneburg M, et al. Cold atmospheric plasma (CAP) activates angiogenesis-related molecules in skin keratinocytes, fibroblasts and endothelial cells and improves wound angiogenesis in an autocrine and paracrine mode. J Dermatol Sci. 2018;89:181–190.
  • Duchesne C, Banzet S, Lataillade -J-J, et al. Cold atmospheric plasma modulates endothelial nitric oxide synthase signalling and enhances burn wound neovascularisation. J Pathol. 2019;249:368–380.
  • Nikitovic D, Corsini E, Kouretas D, et al. ROS-major mediators of extracellular matrix remodeling during tumor progression. Food Chem Toxicol. 2013;61:178–186.
  • Keyvani A, Atyabi SM, Sardari S, et al. Effects of cold atmospheric plasma jet on collagen structure in different treatment times. Basic Res J Med Clin Sci. 2017;6:84–90.
  • Dong X, Chen M, Wang Y, et al. A mechanistic study of plasma treatment effects on demineralized dentin surfaces for improved adhesive/dentin interface bonding. Clin Plasma Med. 2014;2:11–16.
  • Samouillan V, Merbahi N, Yousfi M, et al. Effect of low-temperature plasma jet on thermal stability and physical structure of type I Collagen. IEEE Trans Plasma Sci. 2012;40:1688–1695.
  • Wiegand C, Fink S, Beier O, et al. Dose- and time-dependent cellular effects of cold atmospheric pressure plasma evaluated in 3D skin models. Skin Pharmacol Physiol. 2016;29:257–265.
  • Arndt S, Unger P, Wacker E, et al. Cold atmospheric plasma (CAP) changes gene expression of key molecules of the wound healing machinery and improves wound healing in vitro and in vivo. PloS One. 2013;8:e79325–e79325.
  • Shi X, Cai J, Xu G, et al. Effect of cold plasma on cell viability and collagen synthesis in cultured murine fibroblasts. Plasma Sci Technol. 2016;18:353–359.
  • Vijayarangan V, Delalande A, Dozias S, et al. Cold atmospheric plasma parameters investigation for efficient drug delivery in hela cells. IEEE Trans Radiat Plasma Med Sci. 2018;2:109–115.
  • Jinno M, Tachibana K, Motomura H, et al. Improvement of efficiency and viability in plasma gene transfection by plasma minimization and optimization electrode configuration. Jpn J Appl Phys. 2016;55:07LG09.
  • Zhu W, Lee S-J, Castro NJ, et al. Synergistic effect of cold atmospheric plasma and drug loaded core-shell nanoparticles on inhibiting breast cancer cell growth. Sci Rep. 2016;6:21974.
  • Kim GC, Kim GJ, Park SR, et al. Air plasma coupled with antibody-conjugated nanoparticles: a new weapon against cancer. J Phys Appl Phys. 2008;42:032005.
  • Wang M, Geilich BM, Keidar M, et al. Killing malignant melanoma cells with protoporphyrin IX-loaded polymersome-mediated photodynamic therapy and cold atmospheric plasma. Int J Nanomedicine. 2017;12:4117–4127.
  • Raavé R, van Kuppevelt TH, Daamen WF. Chemotherapeutic drug delivery by tumoral extracellular matrix targeting. J Control Release. 2018;274:1–8.
  • Guo J, Zeng H, Chen Y. Emerging Nano drug delivery systems targeting cancer-associated fibroblasts for improved antitumor effect and tumor drug penetration. Mol Pharm. 2020;17:1028–1048.
  • Van der Steen SC, Raavé R, Langerak S, et al. Targeting the extracellular matrix of ovarian cancer using functionalized, drug loaded lyophilisomes. Eur J Pharm Biopharm. 2017;113:229–239.
  • Li L, Zhou S, Lv N, et al. Photosensitizer-encapsulated ferritins mediate photodynamic therapy against cancer-associated fibroblasts and improve tumor accumulation of nanoparticles. Mol Pharm. 2018;15:3595–3599.
  • Vail DM, Amantea MA, Colbern GT, et al. Pegylated liposomal doxorubicin: proof of principle using preclinical animal models and pharmacokinetic studies. Semin Oncol. 2004;31(6 Suppl 13):16–35. Elsevier.
  • Dreher MR, Liu W, Michelich CR, et al. Tumor vascular permeability, accumulation, and penetration of macromolecular drug carriers. J Natl Cancer Inst. 2006;98:335–344.
  • Wang H, Lin Y, Nienhaus K, et al. The protein corona on nanoparticles as viewed from a nanoparticle-sizing perspective. WIREs Nanomedicine Nanobiotechnology. 2018;10:e1500.
  • Cai R, Chen C. The Crown and the Scepter: roles of the protein corona in nanomedicine. Adv Mater Deerfield Beach Fla. 2019;31:e1805740.
  • Appelboom G, Detappe A, LoPresti M, et al. Stereotactic modulation of blood-brain barrier permeability to enhance drug delivery. Neuro-Oncol. 2016;18:1601–1609.
  • Hynynen K. Focused ultrasound for blood–brain disruption and delivery of therapeutic molecules into the brain. Expert Opin Drug Deliv. 2007;4:27–35.
  • Lorenzo MF, Thomas SC, Kani Y, et al. Temporal characterization of blood-brain barrier disruption with high-frequency electroporation. Cancers (Basel). 2019;11:1850.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.