Reversing activity of cancer associated fibroblast for staged glycolipid micelles against internal breast tumor cells

Yun Zhu, Fangying Yu, Yanan Tan, Yun Hong, Tingting Meng, Yupeng Liu, Suhuan Dai, Guoxi Qiu, Hong Yuan, Fuqiang Hu, Yun Zhu, Fangying Yu, Yanan Tan, Yun Hong, Tingting Meng, Yupeng Liu, Suhuan Dai, Guoxi Qiu, Hong Yuan, Fuqiang Hu

Abstract

Rationale: Nano-carrier based combinational therapies for tumor cells hold great potential to improve the outcomes of patients. However, cancer associated fibroblasts (CAFs) in desmoplastic tumors and the derived pathological tumor stroma severely impede the access and sensitibity of tumor cells to antitumor therapies. Methods: Glycolipid-based polymeric micelles (GLPM) were developed to encapsulate an angiotensin II receptor I inhibitor (telmisartan, Tel) and a cytotoxic drug (doxorubicin, DOX) respectively, which could exert combinational antitumor efficacy by reprogramming tumor microenvironment to expose the vulnerability of internal tumor cells. Results: As demonstrated, α-SMA positive CAFs significantly decreased after the pre-administration of GLPM/Tel in vitro, which accordingly inhibited the secretion of the CAFs derived stroma. The tumor vessels were further decompressed as a result of the alleviated solid stress inside the tumor masses, which promoted more intratumoral drug delivery and penetration. Ultimately, staged administration of the combined GLPM/Tel and GLPM/DOX at the screened molar ratio not only inhibited the stroma continuously, but also achieved a synergistic antitumor effect through the apoptosis-related peroxisome proliferator-activated receptor-gamma (PPAR-γ) pathway. Conclusion: In summary, the strategy of suppressing tumor stroma for subsequent combinational therapies against internal breast tumor cells could provide avenues for management of intractable desmoplastic tumors.

Keywords: cancer associated fibroblasts; doxorubicin; glycolipid micelles; stroma; telmisartan..

Conflict of interest statement

Competing Interests: The authors have declared that no competing interest exists.

© The author(s).

Figures

Figure 1
Figure 1
Schematic illustration of the mechanism of remodeling the tumor microenvironment for enhanced synergistic antitumor therapy against breast tumor cells. Pre-administered GLPM/Tel primarily decreased the tumor stroma by reversing the activity of CAFs to alleviate solid stress and facilitate drug penetration in desmoplastic breast tumors. The subsequent administration of combined GLPM/Tel and GLPM/DOX enhanced antitumor efficacy collaboratively through the PPAR-γ signal pathway.
Figure 2
Figure 2
Pharmacological action evaluation of GLPM/Tel in activated NIH/3T3 cells. A) Images of GLPM/Tel and GLPM/DOX visualized by TEM. B) Immunofluorescence staining analysis of α-SMA expression after different drugs treatment for 48h. C) Western blot analysis of the α-SMA and CTGF proteins after PBS, Tel and GLPM/Tel incubation for 48h. D) The intracellular uptake of Tel and GLPM/Tel on normal NIH/3T3 and activated NIH/3T3 cell lines. *p

Figure 3

The evaluation of synergistic antitumor…

Figure 3

The evaluation of synergistic antitumor efficiency between GLPM/Tel and GLPM/DOX in MCF-7 cells…

Figure 3
The evaluation of synergistic antitumor efficiency between GLPM/Tel and GLPM/DOX in MCF-7 cells in vitro. A) Intracellular uptake of Tel and GLPM/Tel on MCF-7 cells. B) Western blot analysis of apoptosis related PPAR-γ and cleaved-caspase 3 proteins after drugs treatment for 48h. The combination index (CI) vs Fa plots of C) combinational Tel and DOX, D) GLPM/Tel and GLPM/DOX was calculated according to the MTT assays. E) Evaluation of antitumor efficacy by Annexin V-FITC/PI staining after different drugs treatments. **p < 0.01, ***p < 0.001 as determined by two-tailed student's t-test.

Figure 4

Enhanced drugs penetration by pre-administrated…

Figure 4

Enhanced drugs penetration by pre-administrated GLPM/Tel in vitro and in vivo . A)…

Figure 4
Enhanced drugs penetration by pre-administrated GLPM/Tel in vitro and in vivo. A) Z-stack images of penetration of GLPM/DOX in MCTSs were visualized by confocal microscopy after PBS, Tel and GLPM/Tel treatment for 2 days. Scale bar: 200μm. B) Curves of fluorescence intensity vs different depth in MCTSs calculated by ImageJ software. C) Fluorescent images of ICG labelled GLPM distribution on breast xenografts at determined time intervals after saline, Tel and GLPM/Tel treatments for three times. The tumor position is circled by the red lines. D) Semi-quantitative analysis of fluorescence intensity from the excised tumor masses. E) Distribution patterns of ICG labelled GLPM in vivo. Bar: 50μm. F) Graphic illustration of changed distribution patterns in breast tumors after drug treatments. T: tumor cells area, S: stromal area, red line: blood vessel, blue dots: accumulated drugs.

Figure 5

The remodeled tumor microenvironment at…

Figure 5

The remodeled tumor microenvironment at pre-administration of GLPM/Tel. A) Changes in the drug…

Figure 5
The remodeled tumor microenvironment at pre-administration of GLPM/Tel. A) Changes in the drug penetration capacity of breast tumors with saline, Tel and GLPM/Tel post-injection. Green fluorescence indicates α-SMA positive CAFs, and purple fluorescence indicates CD31 positive vessels, and red fluorescence indicates subsequently administered GLPM/DiI. Bar: 100μm. Semi-quantitative analysis of B) α-SMA positive CAFs, C) diameter of tumor vessels and D) penetrated DiI. E) Images of immunohistochemical staining and semi-quantitative analysis for F) TGF-β and G) CCL2 (n=5). *p < 0.05, **p < 0.01, ***p < 0.001 as determined by two-tailed Student's t-test.

Figure 6

Investigation of alleviated solid stress…

Figure 6

Investigation of alleviated solid stress at post-administration of GLPM/Tel. A) Estimation of solid…

Figure 6
Investigation of alleviated solid stress at post-administration of GLPM/Tel. A) Estimation of solid stress in breast tumors surgically excised from mice after saline, Tel and GLPM/Tel treatment. B) Images and semi-quantitative analysis of HIF-α stained by immunofluorescent staining (n=5). Bar: 100μm. *p

Figure 7

Antitumor efficacy of combination administration…

Figure 7

Antitumor efficacy of combination administration strategy. a) Scheme of the strategy for combinational…

Figure 7
Antitumor efficacy of combination administration strategy. a) Scheme of the strategy for combinational therapies in vivo. b) In vivo tumor inhibition evaluation of different drug treatments. The results are shown as the means ± SD. c) The weights of excised tumor masses and d) the weight of mice was monitored during drug treatment (n=5). *p < 0.05, **p < 0.01, ***p < 0.001 as determined by two-tailed Student's t-test. e) HE staining and TUNEL staining for tumor mass were adopted after different drugs treatments to study the efficacy of remodelingthe tumor microenvironment and subsequent synergistic antitumor efficacy. Scale bar: 100μm. I: saline, II: Tel, III: DOX, IV: Tel+combination (Tel+DOX); I': GLPM/Tel, II': GLPM/DOX, III': GLPM/Tel+GLPM/DOX, IV': GLPM/Tel+combined NPs (GLPM/Tel+GLPM/DOX).
All figures (7)
Similar articles
Cited by
References
    1. Peng H, Chen B, Huang W, Tang Y, Jiang Y, Zhang W. et al. Reprogramming Tumor-Associated Macrophages To Reverse EGFR(T790M) Resistance by Dual-Targeting Codelivery of Gefitinib/Vorinostat. Nano Lett. 2017;17:7684–90. - PubMed
    1. Greco F, Vicent MJ. Combination therapy: opportunities and challenges for polymer-drug conjugates as anticancer nanomedicines. Adv Drug Deliv Rev. 2009;61:1203–13. - PubMed
    1. Mura S, Nicolas J, Couvreur P. Stimuli-responsive nanocarriers for drug delivery. Nature Materials. 2013;12:991–1003. - PubMed
    1. Valkenburg KC, de Groot AE, Pienta KJ. Targeting the tumour stroma to improve cancer therapy. Nat Rev Clin Oncol. 2018;15:366–81. - PMC - PubMed
    1. Zhang B, Jin K, Jiang T, Wang L, Shen S, Luo Z. et al. Celecoxib normalizes the tumor microenvironment and enhances small nanotherapeutics delivery to A549 tumors in nude mice. Sci Rep. 2017;7:10071. - PMC - PubMed
Show all 62 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib .nbib
Format: AMA APA MLA NLM
Figure 3
Figure 3
The evaluation of synergistic antitumor efficiency between GLPM/Tel and GLPM/DOX in MCF-7 cells in vitro. A) Intracellular uptake of Tel and GLPM/Tel on MCF-7 cells. B) Western blot analysis of apoptosis related PPAR-γ and cleaved-caspase 3 proteins after drugs treatment for 48h. The combination index (CI) vs Fa plots of C) combinational Tel and DOX, D) GLPM/Tel and GLPM/DOX was calculated according to the MTT assays. E) Evaluation of antitumor efficacy by Annexin V-FITC/PI staining after different drugs treatments. **p < 0.01, ***p < 0.001 as determined by two-tailed student's t-test.
Figure 4
Figure 4
Enhanced drugs penetration by pre-administrated GLPM/Tel in vitro and in vivo. A) Z-stack images of penetration of GLPM/DOX in MCTSs were visualized by confocal microscopy after PBS, Tel and GLPM/Tel treatment for 2 days. Scale bar: 200μm. B) Curves of fluorescence intensity vs different depth in MCTSs calculated by ImageJ software. C) Fluorescent images of ICG labelled GLPM distribution on breast xenografts at determined time intervals after saline, Tel and GLPM/Tel treatments for three times. The tumor position is circled by the red lines. D) Semi-quantitative analysis of fluorescence intensity from the excised tumor masses. E) Distribution patterns of ICG labelled GLPM in vivo. Bar: 50μm. F) Graphic illustration of changed distribution patterns in breast tumors after drug treatments. T: tumor cells area, S: stromal area, red line: blood vessel, blue dots: accumulated drugs.
Figure 5
Figure 5
The remodeled tumor microenvironment at pre-administration of GLPM/Tel. A) Changes in the drug penetration capacity of breast tumors with saline, Tel and GLPM/Tel post-injection. Green fluorescence indicates α-SMA positive CAFs, and purple fluorescence indicates CD31 positive vessels, and red fluorescence indicates subsequently administered GLPM/DiI. Bar: 100μm. Semi-quantitative analysis of B) α-SMA positive CAFs, C) diameter of tumor vessels and D) penetrated DiI. E) Images of immunohistochemical staining and semi-quantitative analysis for F) TGF-β and G) CCL2 (n=5). *p < 0.05, **p < 0.01, ***p < 0.001 as determined by two-tailed Student's t-test.
Figure 6
Figure 6
Investigation of alleviated solid stress at post-administration of GLPM/Tel. A) Estimation of solid stress in breast tumors surgically excised from mice after saline, Tel and GLPM/Tel treatment. B) Images and semi-quantitative analysis of HIF-α stained by immunofluorescent staining (n=5). Bar: 100μm. *p

Figure 7

Antitumor efficacy of combination administration…

Figure 7

Antitumor efficacy of combination administration strategy. a) Scheme of the strategy for combinational…

Figure 7
Antitumor efficacy of combination administration strategy. a) Scheme of the strategy for combinational therapies in vivo. b) In vivo tumor inhibition evaluation of different drug treatments. The results are shown as the means ± SD. c) The weights of excised tumor masses and d) the weight of mice was monitored during drug treatment (n=5). *p < 0.05, **p < 0.01, ***p < 0.001 as determined by two-tailed Student's t-test. e) HE staining and TUNEL staining for tumor mass were adopted after different drugs treatments to study the efficacy of remodelingthe tumor microenvironment and subsequent synergistic antitumor efficacy. Scale bar: 100μm. I: saline, II: Tel, III: DOX, IV: Tel+combination (Tel+DOX); I': GLPM/Tel, II': GLPM/DOX, III': GLPM/Tel+GLPM/DOX, IV': GLPM/Tel+combined NPs (GLPM/Tel+GLPM/DOX).
All figures (7)
Figure 7
Figure 7
Antitumor efficacy of combination administration strategy. a) Scheme of the strategy for combinational therapies in vivo. b) In vivo tumor inhibition evaluation of different drug treatments. The results are shown as the means ± SD. c) The weights of excised tumor masses and d) the weight of mice was monitored during drug treatment (n=5). *p < 0.05, **p < 0.01, ***p < 0.001 as determined by two-tailed Student's t-test. e) HE staining and TUNEL staining for tumor mass were adopted after different drugs treatments to study the efficacy of remodelingthe tumor microenvironment and subsequent synergistic antitumor efficacy. Scale bar: 100μm. I: saline, II: Tel, III: DOX, IV: Tel+combination (Tel+DOX); I': GLPM/Tel, II': GLPM/DOX, III': GLPM/Tel+GLPM/DOX, IV': GLPM/Tel+combined NPs (GLPM/Tel+GLPM/DOX).

References

    1. Peng H, Chen B, Huang W, Tang Y, Jiang Y, Zhang W. et al. Reprogramming Tumor-Associated Macrophages To Reverse EGFR(T790M) Resistance by Dual-Targeting Codelivery of Gefitinib/Vorinostat. Nano Lett. 2017;17:7684–90.
    1. Greco F, Vicent MJ. Combination therapy: opportunities and challenges for polymer-drug conjugates as anticancer nanomedicines. Adv Drug Deliv Rev. 2009;61:1203–13.
    1. Mura S, Nicolas J, Couvreur P. Stimuli-responsive nanocarriers for drug delivery. Nature Materials. 2013;12:991–1003.
    1. Valkenburg KC, de Groot AE, Pienta KJ. Targeting the tumour stroma to improve cancer therapy. Nat Rev Clin Oncol. 2018;15:366–81.
    1. Zhang B, Jin K, Jiang T, Wang L, Shen S, Luo Z. et al. Celecoxib normalizes the tumor microenvironment and enhances small nanotherapeutics delivery to A549 tumors in nude mice. Sci Rep. 2017;7:10071.
    1. Moffitt RA, Marayati R, Flate EL, Volmar KE, Loeza SG, Hoadley KA. et al. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat Genet. 2015;47:1168–78.
    1. McMillin DW, Negri JM, Mitsiades CS. The role of tumour-stromal interactions in modifying drug response: challenges and opportunities. Nat Rev Drug Discov. 2013;12:217–28.
    1. Lv Y, Xu C, Zhao X, Lin C, Yang X, Xin X. et al. Nanoplatform Assembled from a CD44-Targeted Prodrug and Smart Liposomes for Dual Targeting of Tumor Microenvironment and Cancer Cells. ACS Nano. 2018;12:1519–36.
    1. Olive KP, Jacobetz MA, Davidson CJ, Gopinathan A, McIntyre D, Honess D. et al. Inhibition of Hedgehog Signaling Enhances Delivery of Chemotherapy in a Mouse Model of Pancreatic Cancer. Science. 2009;324:1457–61.
    1. Diop-Frimpong B, Chauhan VP, Krane S, Boucher Y, Jain RK. Losartan inhibits collagen I synthesis and improves the distribution and efficacy of nanotherapeutics in tumors. Proc Natl Acad Sci U S A. 2011;108:2909–14.
    1. Rk ST, Jain. Combining two strategies to improve perfusion and drug delivery in solid tumors. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:18632–7.
    1. McMillin DW, Negri JM, Mitsiades CS. The role of tumour-stromal interactions in modifying drug response: challenges and opportunities. Nature Reviews Drug Discovery. 2013;12:217–28.
    1. Zhu Y, Wen L, Shao S, Tan Y, Meng T, Yang X, Inhibition of tumor-promoting stroma to enforce subsequently targeting AT1R on tumor cells by pathological inspired micelles. Biomaterials; 2018. pp. 33–46.
    1. Yazdani S, Bansal R, Prakash J. Drug targeting to myofibroblasts: Implications for fibrosis and cancer. Adv Drug Deliv Rev. 2017;121:101–16.
    1. Whatcott CJ, Han H, Von Hoff DD. Orchestrating the Tumor Microenvironment to Improve Survival for Patients With Pancreatic Cancer: Normalization, Not Destruction. Cancer J. 2015;21:299–306.
    1. Graf D, Reinehr R, Kurz AK, Fischer R, Haussinger D. Inhibition of taurolithocholate 3-sulfate-induced apoptosis by cyclic AMP in rat hepatocytes involves protein kinase A-dependent and -independent mechanisms. Arch Biochem Biophys. 2003;415:34–42.
    1. Habashi JP, Judge DP, Holm TM, Cohn RD, Loeys BL, Cooper TK. et al. Losartan, an AT1 antagonist, prevents aortic aneurysm in a mouse model of Marfan syndrome. Science. 2006;312:117–21.
    1. Liu J, Liao S, Diop-Frimpong B, Chen W, Goel S, Naxerova K. et al. TGF-β blockade improves the distribution and efficacy of therapeutics in breast carcinoma by normalizing the tumor stroma. Proceedings of the National Academy of Sciences. 2012;109:16618.
    1. Duncan MR, Frazier KS, Abramson S, Williams S, Klapper H, Huang X. et al. Connective tissue growth factor mediates transforming growth factor beta-induced collagen synthesis: down-regulation by cAMP. FASEB journal: official publication of the Federation of American Societies for Experimental Biology. 1999;13:1774–86.
    1. Rodríguezvita J, Sánchezlópez E, Esteban V, Rupérez M, Egido J, Ruizortega M. Angiotensin II activates the Smad pathway in vascular smooth muscle cells by a transforming growth factor-beta-independent mechanism. Circulation. 2005;111:2509–17.
    1. Benson SC, Pershadsingh HA, Ho CI, Chittiboyina A, Desai P, Pravenec M. et al. Identification of telmisartan as a unique angiotensin II receptor antagonist with selective PPARgamma-modulating activity. Hypertension. 2004;43:993–1002.
    1. Li J, Chen L, Yu P, Liu B, Zhu J, Yang Y. Telmisartan exerts anti-tumor effects by activating peroxisome proliferator-activated receptor-γ in human lung adenocarcinoma A549 cells. Molecules. 2014;19:2862–76.
    1. Noguchi R, Yoshiji H, Ikenaka Y, Namisaki T, Kitade M, Kaji K. et al. Synergistic inhibitory effect of gemcitabine and angiotensin type-1 receptor blocker, losartan, on murine pancreatic tumor growth via anti-angiogenic activities. Oncology Reports. 2009;22:355–60.
    1. Koyama N, Nishida Y, Ishii T, Yoshida T, Furukawa Y, Narahara H. Telmisartan induces growth inhibition, DNA double-strand breaks and apoptosis in human endometrial cancer cells. PLoS One. 2014;9:e93050.
    1. Hu FQ, Zhao MD, Yuan H, You J, Du YZ, Zeng S. A novel chitosan oligosaccharide-stearic acid micelles for gene delivery: properties and in vitro transfection studies. International Journal of Pharmaceutics. 2006;315:158–66.
    1. Hu FQ, Liu LN, Du YZ, Yuan H. Synthesis and antitumor activity of doxorubicin conjugated stearic acid- g -chitosan oligosaccharide polymeric micelles. Biomaterials. 2009;30:6955–63.
    1. Liu X, Cheng B, Meng T, You J, Zhu Y, Lu B. et al. Synthesis and biological application of BKT-140 peptide modified polymer micelles for treating tumor metastasis with an enhanced cell internalization efficiency. Polymer Chemistry. 2015;7:1375–86.
    1. Liu N, Tan Y, Hu Y, Meng T, Wen L, Liu J. et al. A54 Peptide Modified and Redox-Responsive Glucolipid Conjugate Micelles for Intracellular Delivery of Doxorubicin in Hepatocarcinoma Therapy. ACS Appl Mater Interfaces. 2016;8:33148–56.
    1. Wan X, Min Y, Bludau H, Keith A, Sheiko SS, Jordan R. et al. Drug Combination Synergy in Worm-like Polymeric Micelles Improves Treatment Outcome for Small Cell and Non-Small Cell Lung Cancer. ACS Nano. 2018;12:2426–39.
    1. Ying M, Shen Q, Zhan C, Wei X, Gao J, Xie C. et al. A stabilized peptide ligand for multifunctional glioma targeted drug delivery. Journal of Controlled Release. 2016;243:86–98.
    1. Benson SC, Pershadsingh HA, Ho CI, Chittiboyina A, Desai P, Pravenec M. et al. Identification of Telmisartan as a Unique Angiotensin II Receptor Antagonist With Selective PPARγ-Modulating Activity. Hypertension. 2004;43:993–1002.
    1. Chou TC, Talalay P. Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzyme Regul. 1984;22:27–55.
    1. Han Y, He Z, Schulz A, Bronich TK, Jordan R, Luxenhofer R. et al. Synergistic Combinations of Multiple Chemotherapeutic Agents in High Capacity Poly(2-oxazoline) Micelles. Mol Pharm. 2015;9:2302–13.
    1. Stylianopoulos T, Martin JD, Chauhan VP, Jain SR, Diop-Frimpong B, Bardeesy N. et al. Causes, consequences, and remedies for growth-induced solid stress in murine and human tumors. Proceedings of the National Academy of Sciences of the United States of America. 2012;109:15101–8.
    1. Troncoso AB, Acosta E. The van der Waals interactions in sphere-shell and cone-shell configurations. J Phys Chem B. 2012;116:14051–61.
    1. Chauhan VP, Martin JD, Liu H, Lacorre DA, Jain SR, Kozin SV. et al. Angiotensin inhibition enhances drug delivery and potentiates chemotherapy by decompressing tumour blood vessels. Nature Communications. 2013;4:2516.
    1. Chauhan VP, Martin JD, Liu H, Lacorre DA, Jain SR, Kozin SV. et al. Angiotensin inhibition enhances drug delivery and potentiates chemotherapy by decompressing tumour blood vessels. Nat Commun. 2013;4:2516.
    1. Jain RK. Determinants of tumor blood flow: a review. Cancer Res. 1988;48:2641–58.
    1. Donahue ND, Acar H, Wilhelm S. Concepts of nanoparticle cellular uptake, intracellular trafficking, and kinetics in nanomedicine. Adv Drug Deliv Rev; 2019.
    1. Miao L, Newby JM, Lin CM, Zhang L, Xu F, Kim WY. et al. The Binding Site Barrier Elicited by Tumor-Associated Fibroblasts Interferes Disposition of Nanoparticles in Stroma-Vessel Type Tumors. ACS Nano. 2016;10:9243–58.
    1. Smith NR, Baker D, Farren M, Pommier A, Swann R, Wang X. et al. Tumor Stromal Architecture Can Define the Intrinsic Tumor Response to VEGF-Targeted Therapy. Clin Cancer Res. 2013;19:6943–56.
    1. Timmins NE, Nielsen LK. Generation of multicellular tumor spheroids by the hanging-drop method. Methods Mol Med. 2007;140:141–51.
    1. Friedrich J, Seidel C, Ebner R, Kunz-Schughart LA. Spheroid-based drug screen: considerations and practical approach. Nat Protoc. 2009;4:309–24.
    1. Cabral H, Matsumoto Y, Mizuno K, Chen Q, Murakami M, Kimura M. et al. Accumulation of sub-100 nm polymeric micelles in poorly permeable tumours depends on size. Nature Nanotechnology. 2011;6:815–23.
    1. Jain RK. Normalization of Tumor Vasculature: An Emerging Concept in Antiangiogenic Therapy. Science. 2005;307:58.
    1. Jain RK. Normalizing tumor vasculature with anti-angiogenic therapy: A new paradigm for combination therapy. Nature Medicine. 2001;7:987.
    1. Jain RK. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science. 2005;307:58–62.
    1. Jain RK. Normalizing tumor vasculature with anti-angiogenic therapy: a new paradigm for combination therapy. Nat Med. 2001;7:987–9.
    1. Huang S, Holzel M, Knijnenburg T, Schlicker A, Roepman P, McDermott U. et al. MED12 controls the response to multiple cancer drugs through regulation of TGF-beta receptor signaling. Cell. 2012;151:937–50.
    1. Nieskoski MD, Marra K, Gunn JR, Hoopes PJ, Doyley MM, Hasan T. et al. Collagen Complexity Spatially Defines Microregions of Total Tissue Pressure in Pancreatic Cancer. Sci Rep. 2017;7:10093.
    1. Padera TP, Stoll BR, Tooredman JB, Capen D, di Tomaso E, Jain RK. Pathology: cancer cells compress intratumour vessels. Nature. 2004;427:695.
    1. Stylianopoulos T, Jain RK. Combining two strategies to improve perfusion and drug delivery in solid tumors. Proc Natl Acad Sci U S A. 2013;110:18632–7.
    1. DuFort CC, DelGiorno KE, Carlson MA, Osgood RJ, Zhao C, Huang Z. et al. Interstitial Pressure in Pancreatic Ductal Adenocarcinoma Is Dominated by a Gel-Fluid Phase. Biophys J. 2016;110:2106–19.
    1. Cheng B, Lu B, Xuan L, Meng T, Tan Y, Yun Z, A pH-responsive glycolipid-like nanocarrier for optimising the time-dependent distribution of free chemical drugs in focal cells. Int J Pharm; 2017. p. 522.
    1. Fang RH, Zhang L. Combinatorial nanotherapeutics: rewiring, then killing, cancer cells. Sci Signal. 2014;7:pe13.
    1. Cheng B, Lu B, Liu X, Meng T, Tan Y, Zhu Y. et al. A pH-responsive glycolipid-like nanocarrier for optimising the time-dependent distribution of free chemical drugs in focal cells. Int J Pharm. 2017;522:210–21.
    1. Hu K, Miao L, Goodwin TJ, Li J, Liu Q, Huang L. Quercetin Remodels the Tumor Microenvironment To Improve the Permeation, Retention, and Antitumor Effects of Nanoparticles. ACS Nano. 2017;11:4916–25.
    1. Chen B, Dai W, Mei D, Liu T, Li S, He B. et al. Comprehensively priming the tumor microenvironment by cancer-associated fibroblast-targeted liposomes for combined therapy with cancer cell-targeted chemotherapeutic drug delivery system. J Control Release. 2016;241:68–80.
    1. Li M, Li M, Yin T, Shi H, Wen Y, Zhang B. et al. Targeting of cancerassociated fibroblasts enhances the efficacy of cancer chemotherapy by regulating the tumor microenvironment. Mol Med Rep. 2016;13:2476–84.
    1. Wang LY, Liu XR, Zhou Q, Sui MH, Lu ZP, Zhou ZX. et al. Terminating the criminal collaboration in pancreatic cancer: Nanoparticle-based synergistic therapy for overcoming fibroblast induced drug resistance. Biomaterials. 2017;144:105–18.
    1. Chen Q, Liu G, Liu S, Su H, Wang Y, Li J. et al. Remodeling the Tumor Microenvironment with Emerging Nanotherapeutics. Trends Pharmacol Sci. 2018;39:59–74.
    1. Truffi M, Mazzucchelli S, Bonizzi A, Sorrentino L, Allevi R, Vanna R, Nano-Strategies to Target Breast Cancer-Associated Fibroblasts: Rearranging the Tumor Microenvironment to Achieve Antitumor Efficacy. Int J Mol Sci; 2019. p. 20.

Source: PubMed

3
Se inscrever