Tumor microenvironment

The tumor microenvironment is the cellular environment in which the tumor exists, including surrounding blood vessels, immune cells, fibroblasts, other cells, signaling molecules, and the extracellular matrix (ECM).[1] The tumor and the surrounding microenvironment are closely related and interact constantly. Tumors can influence the microenvironment by releasing extracellular signals, promoting tumor angiogenesis and inducing peripheral immune tolerance, while the immune cells in the microenvironment can affect the growth and evolution of cancerous cells, such as in immuno-editing. The tumor microenvironment has been shown to contribute to tumour heterogeneity. In one of its earliest forms, this concept of interplay between the tumor and its microenvironment can be seen in Stephen Paget's "seed and soil" theory where he postulated that metastases of a particular type of cancer ("the seed") often metastasizes to certain sites ("the soil") based on the similarity of the environments of the original and secondary tumor sites.[2] Later, experiments by Halachmi and Witz in mice showed that for the same cancer cell line, inoculation in mice (where the tumor microenvironment could affect the cancer) created greater tumorigenicity than the same strain inoculated in in vitro culture.[3][4]

Vasculature in the tumor microenvironment

80-90% of cancer are carcinomas, or cancers that form in the epithelial tissue.[5] This tissue is not vascularized, which prevents tumors from growing greater than 2mm in diameter without recruiting new blood vessels to feed itself.[6] The process of angiogenesis is dysregulated to feed the cancer cells, and as a result the vasculature formed differs from that of normal tissue.

Enhanced permeability and retention effect

The enhanced permeability and retention effect (EPR effect) is the observation that the vasculature of tumors is often leaky and accumulates molecules in the blood stream to a greater extent than normal tissue. This effect linked to inflammation is not only seen in tumors, but in hypoxic area of cardiac muscles following a myocardial infarction (MI or heart attack).[7][8] This leaky vasculature is thought to have several causes, including a dearth of pericytes and a malformed basement membrane.[8]

Hypoxia

The tumor microenvironment is often hypoxic. As the tumor mass increases, the interior of the tumor grows farther away from existing blood supply. While angiogenesis can reduce this affect, the partial pressure of oxygen is below 5 mm Hg (venous blood has a partial pressure of oxygen at 40 mm Hg) in more than 50% of locally advanced solid tumors.[9][10] The hypoxic environment leads to genetic instability, which is associated with cancer progression, via downregulating nucleotide excision repair (NER) and mismatch repair (MMR) pathways.[11] Hypoxia also causes the upregulation of hypoxia-inducible factor 1 alpha (HIF1-α), which induces angiogenesis, and is associated with poorer prognosis and the activation of genes associated with metastasis.[10]

While a lack of oxygen can cause glycolytic behavior in cells, tumor cells have also been shown to undergo aerobic glycolysis as well, in which they preferentially produce lactate from glucose even when there is abundant oxygen. This phenomenon is called the Warburg effect, in honor of its discoverer, Otto Warburg.[12] No matter the cause, this leaves the extracellular microenvironment acidic (pH 6.5-6.9), while the cancer cells themselves are able to remain neutral (ph 7.2-7.4). It has been shown that this induces greater cell migration in vivo and in vitro, possibly by promoting degradation of the ECM.[13][14]

Reactive stromal cells and the microenvironment

The stroma of a carcinoma is the connective tissue below the basal lamina. This includes fibroblasts, ECM, immune cells, and other cells and molecules. The stroma surrounding a tumor often reacts to the intrusion via inflammation, similar to how it might with a wound, leading cancer to be called "wounds that do not heal."[15] Inflammation can encourage angiogenesis, speed the cell cycle, and prevent cell death, all of which augments tumor growth.[16]

Carcinoma associated fibroblasts

Carcinoma associated fibroblasts (CAFs) are a heterogenous group of fibroblasts whose function is pirated by cancer cells and then contribute toward carcinogenesis[17] These cells usually are derived from the normal fibroblasts in the surrounding stroma but can also come from pericytes, smooth muscle cells, fibrocytes, mesenchymal stem cells (MSCs, often derived from bone marrow), or via epithelial-mesenchymal transition (EMT) or endothelial-mesenchymal transition (EndMT).[18][19][20] Unlike their normal counterparts, CAFs do not retard cancer growth in vitro.[21] Beyond simply lacking the ability of tumor inhibition, CAFs also perform several functions which support tumor growth, such as secreting vascular endothelial growth factor (VEGF), fibroblast growth factors (FGFs), platelet-derived growth factor (PDGF), and other pro-angiogenic signals to induce angiogenesis.[9] CAFs can also secrete transforming growth factor beta (TGF-β), which is associated with EMT, a process by which cancer cells can metastasize,[22] and is associated with inhibiting cytotoxic T cells and natural killer T cells.[23] As fibroblasts, CAFs are able to rework the ECM to include more paracrine survival signals such as IGF-1 and IGF-2, thus promoting survival of the surrounding cancer cells.[17] CAFs are also associated with the Reverse Warburg Effect where the CAFs perform aerobic glycolysis and feed lactate to the cancer cells.[17]

Several markers are used to identify CAFs including expression of α smooth muscle actin (αSMA), vimentin, platelet-derived growth factor receptor α (PDGFR-α), platelet-derived growth factor receptor β (PDGFR-β), fibroblast specific protein 1 (FSP-1), and fibroblast activation protein (FAP).[19] None of the factors can be used to differentiate CAFs from all other cells by itself.

Myeloid-derived suppressor cells and tumor associated macrophages

Myeloid-derived suppressor cells (MDSCs) are a heterogenous population of cells of myelogenous origin with the potential to repress T cell responses. They regulate wound repair and inflammation and are rapidly expanded in cancer, correlating with that signs of inflammation are seen in most if not all tumor sites.[24][25] Tumors can produce exosomes that stimulate inflammation via MDSCs.[26][27] This group of cells include some tumor associated macrophages (TAMs).[24] TAMs are a central component in the strong link between chronic inflammation and cancer. TAMs are recruited to the tumor as a response to cancer associated inflammation.[28] Unlike normal macrophages, TAMs lack cytotoxic activity.[29] TAMs have been induced in vitro by exposing macrophage progenitors to different immune regulatory cytokines, such as interleukin 4 (IL-4) and interleukin 13 (IL-13).[17] TAMs gather in necrotic regions of tumors where they have been associated with hiding the cancer cells from normal immune cells by secreting interleukin 10 (IL-10),[30] aiding angiogenesis by secreting vascular endothelial growth factor(VEGF) and nitric oxide synthase(NOS),[9] supporting tumor growth by secreting epidermal growth factor (EGF)[30] and remodeling the ECM.[9] TAMs show sluggish NF-κB activation, which allows for the smoldering inflammation seen in cancer.[31] An increased amount of TAMs is associated with worse prognosis.[32][33] TAMs represent a potential target for novel cancer therapies.

TAMs have recently been associating with using exosomes (vesicles used by mammalian cells to secrete intracellular contents) to deliver invasion-potentiating microRNA (miRNA) into cancerous cells, specifically breast cancer cells.[26][34]

Tumor infiltrating lymphocytes

Tumor infiltrating lymphocytes (TILs) are lymphocytes that penetrate a tumor and while have a common origin with myelogenous cells at the hematopoietic stem cell, diverge in development. Concentration is generally positively correlated.[30] However, only in instances of melanoma has autologous transplant of TILs been used successfully as a means of treatment.[35] Cancer cells have been shown to induce apoptosis of activated T cells (a class of lymphocyte) by secreting exosomes containing death ligands such as FasL and TRAIL, and via the same method, turn off the normal cytotoxic response of natural killer cells (NK cells).[27][36] This suggests that cancer cells actively work to restrain TILs.

Extracellular matrix remodeling

Fibroblasts are in charge of laying down most of the collagens, elastin, glycosaminoglycans, proteoglycans (e.g. perlecan), and glycoproteins in the ECM. As many fibroblasts are transformed into CAFs during carcinogenesis, this reduces the amount of ECM produced and the ECM that is produced can be malformed, like collagen being loosely woven and non-planar, even curved.[37] In addition, CAFs produce matrix matrix metalloproteinases (MMP), which cleave the proteins within the ECM.[9] CAFs are also able to disrupt the ECM via force, generating a track that a carcinoma cell can follow directly behind.[38] In either case, destruction of the ECM allows cancer cells to escape from their in situ location and intravasate into the blood stream where they can metastasize systematically. It can also provide passage for endothelial cells to complete angiogenesis to the tumor site.

Destruction of the ECM also modulates the signaling cascades controlled by the interaction of cell-surface receptors and the ECM, and it also reveals binding sites previously hidden, like the integrin alpha-v beta-3 (αVβ3) on the surface of melanoma cells can be ligated to rescue the cells from apoptosis after degradation of collagen.[39][40] In addition, the degradation products may have downstream effects as well that can increase tumorigenicity of cancer cells and can serve as potential biomarkers.[39] The destruction of the ECM also releases the cytokines and growth factors stored therein (for example, VEGF, basic fibroblast growth factor (bFGF), insulin-like growth factors (IGF1 and IGF2), TGF-β, EGF, heparin-binding EGF-like growth factor (HB-EGF), and tumor necrosis factor (TNF), which can increase the growth of the tumor.[37][41] Cleavage of ECM components can also release cytokines that inhibit tumorigenesis, such as degradation of certain types of collagen can form endostatin, restin, canstatin, & tumstatin, which have antiangiogenic functions.[37]

Stiffening of the ECM is associated with tumor progression.[42] This stiffening may be partially attributed to CAFs secreting lysyl oxidase (LOX), an enzyme that cross-links the collagen IV found in the ECM.[19][43]

Clinical implications

Drug development

Numerous high throughput screens for cancer therapeutics are performed in vitro on cancer cell lines without the accompanying microenvironment, but current studies are also investigating the effects of supportive stroma cells on the biology of cancer cells and their resistance to therapy.[44] These studies revealed that there are interesting therapeutic targets in the microenvironment like integrins or chemokines.[44] These were missed by initial screens for anti-cancer drugs and might also help explain why so few initially identified drugs are highly potent in vivo.

Much effort has been devoted into developing nanocarrier vehicles (~20-200 nm in diameter) for transportation of drugs and other therapeutic molecules, so that these therapies can be targeted to selectively extravasate through tumor vasculature via the EPR effect. Using a nanocarrier is now considered the gold standard of targeted cancer therapy because it targets almost all tumors besides those few that are hypovascularized, like prostate and pancreatic tumors.[8][45] These efforts include protein capsids[46] and liposomes.[47] However, as some important, normal tissues, like the liver and kidneys, also have fenestrated endothelium, great care must be taken with using the correct size (10-100 nm, with greater retention in tumors seen in using larger nanocarriers) and charge (anionic or neutral).[8] Lymphatic vessels do not usually develop with the tumor, leading to increased interstitial fluid pressure, which made abrogate the journey of these nanocarriers to the tumor.[8][48]

Current Therapies

Bevacizumab is clinically approved to treat a variety of cancer by targeting VEGF-A, which is produced by both CAFs and TAMs, thus slowing angiogenesis. Many other small molecule inhibitors exist that block the receptors for the growth factors released, thus making the cancer cell deaf to much of the paracrine signaling produced by CAFs and TAMs. These inhibitors include Sunitinib, Pazopanib, Sorafenib, and Axitinib, all of which inhibit platelet derived growth factor receptors (PDGF-Rs) and VEGF receptors (VEGFRs). Cannabidiol, a cannabis derivate without psychoactive side effects, has also been shown to inhibit the expression of VEGF in Kaposi's sarcoma cells.[49]

Natalizumab is a monoclonal antibody that was designed to target one of the molecules responsible for cell adhesion (integrin VLA-4) and has promising in vitro activity in B cell lymphomas and leukemias.[44]

Also, Trabectedin is known to have immunomodulatory effects that inhibit TAMs.[30]

Current formulations of liposomes encapsulating anti-cancer drugs for selective uptake to tumors via the EPR effect include: Doxil and Myocet, both of which encapsulate doxorubicin (a DNA intercalator and common chemotherapeutic); DaunoXome, which encapsulates daunorubicin (another DNA intercalator similar to doxorubicin); and Onco-TCS, which encapsulates vincristine (a molecule which constitutively induces formation of microtubules, dysregulating cell division). Another novel utilization of the EPR effect comes from Protein-bound paclitaxel (marketed under the trade name Abraxane) where paclitaxel (a molecule which dysregulates cell division via stabilization of microtubules) is bound to albumin to add bulk and aid delivery.

References

  1. http://www.cancer.gov/dictionary?cdrid=561725 NCI Dictionary of Cancer Terms - Tumor Microenvironment
  2. The Lancet, Volume 133, Issue 3421, 23 March 1889, Pages 571-573
  3. Halachmi, E., & Witz, I.P. Differential tumorigenicity of 3T3 cells transformed in vitro with polyoma virus and in vivo selection for high tumorigenicity Cancer Research Volume 49, Issue 9, 1989, Pages 2383-2389
  4. Isaac P. Witz, Orlev Levy-Nissenbaum The tumor microenvironment in the post-PAGET era Cancer Letters, Volume 242, Issue 1, 8 October 2006, Pages 1–10 http://dx.doi.org/10.1016/j.canlet.2005.12.005
  5. http://cancer.stanford.edu/information/cancerOverview.html Standford Medicine Cancer Institute, Cancer Overview
  6. Michael J. Duffy The biochemistry of metastasis Advances in Clinical Chemistry, Volume 32 1996, Pages 135–160
  7. T.N. Palmer, V.J. Caride, M.A. Caldecourt, J. Twickler, & V. Abdullah The mechanism of liposome accumulation in infarction Biochimica et Biophysica Acta (BBA) - General Subjects Volume 797, 1984, Pages 363–368
  8. 8.0 8.1 8.2 8.3 8.4 Fabienne Danhier, Olivier Feron, Véronique Préat To exploit the tumor microenvironment: Passive and active tumor targeting of nanocarriers for anti-cancer drug delivery Journal of Controlled Release, Volume 148, Issue 2, 1 December 2010, Pages 135–146 http://dx.doi.org/10.1016/j.jconrel.2010.08.027
  9. 9.0 9.1 9.2 9.3 9.4 Cynthia E. Weber, Paul C. Kuo The tumor microenvironment Surgical Oncology, Volume 21, Issue 3, September 2012, Pages 172–177 http://dx.doi.org/10.1016/j.suronc.2011.09.001
  10. 10.0 10.1 Mikhail V. Blagosklonny Antiangiogenic therapy and tumor progression Cancer Cell, Volume 5, Issue 1, January 2004, Pages 13–17 http://dx.doi.org/10.1016/S1535-6108(03)00336-2
  11. Ranjit S. Bindra, Peter M. Glazer Genetic instability and the tumor microenvironment: towards the concept of microenvironment-induced mutagenesis Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis, Volume 569, Issues 1–2, 6 January 2005, Pages 75–85 http://dx.doi.org/10.1016/j.mrfmmm.2004.03.013
  12. Robert A. Gatenby & Robert J. Gillies Why do cancers have high aerobic glycolysis? Nature Reviews Cancer Volume 4, November 2004, Pages 891-899 http://dx.doi.org/10.1038/nrc1478
  13. Robert van Sluis, Zaver M. Bhujwalla, Natarajan Raghunand, Paloma Ballesteros, José Alvarez, Sebastián Cerdán, Jean-Philippe Galons, Robert J. Gillies In vivo imaging of extracellular pH using 1H MRSI Magnetic Resonance in Medicine Volume 41, Issue 4, April 1999, Pages 743-750 http://dx.doi.org/10.1002/(SICI)1522-2594(199904)41:4<743::AID-MRM13>3.0.CO;2-Z
  14. Veronica Estrella, Tingan Chen, Mark Lloyd, Jonathan Wojtkowiak, Heather H. Cornnell, Arig Ibrahim-Hashim, Kate Bailey, Yoganand Balagurunathan, Jennifer M. Rothberg, Bonnie F. Sloane, Joseph Johnson, Robert A. Gatenby, and Robert J. Gillies Acidity Generated by the Tumor Microenvironment Drives Local Invasion Cancer Research, Volume 73, Issue 5, 1 March 2013, Pages 1524-1535 http://dx.doi.org/10.1158/0008-5472.CAN-12-2796
  15. Harold F. Dvorak Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing New England Journal of Medicine Volume 325, Issue 26, 25 December 1986, Pages 1650-1659
  16. Joydeb Kumar Kundu, Young-Joon Surh Inflammation: Gearing the journey to cancer Mutation Research/Reviews in Mutation Research, Volume 659, Issues 1–2, July–August 2008, Pages 15–30 http://dx.doi.org/10.1016/j.mrrev.2008.03.002
  17. 17.0 17.1 17.2 17.3 Douglas Hanahan, Lisa M. Coussens Accessories to the Crime: Functions of Cells Recruited to the Tumor Microenvironment Cancer Cell, Volume 21, Issue 3, 20 March 2012, Pages 309–322 http://dx.doi.org/10.1016/j.ccr.2012.02.022
  18. Kati Räsänen, Antti Vaheri Activation of fibroblasts in cancer stroma Experimental Cell Research, Volume 316, Issue 17, 15 October 2010, Pages 2713–2722 http://dx.doi.org/10.1016/j.yexcr.2010.04.032
  19. 19.0 19.1 19.2 Timothy Marsh, Kristian Pietras, Sandra S. McAllister Fibroblasts as architects of cancer pathogenesis Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease Online 30 October 2012 http://dx.doi.org/10.1016/j.bbadis.2012.10.013
  20. Michael Quante, Shui Ping Tu, Hiroyuki Tomita, Tamas Gonda, Sophie S.W. Wang, Shigeo Takashi, Gwang Ho Baik, Wataru Shibata, Bethany DiPrete, Kelly S. Betz, Richard Friedman, Andrea Varro, Benjamin Tycko, & Timothy C. Wang Bone Marrow-Derived Myofibroblasts Contribute to the Mesenchymal Stem Cell Niche and Promote Tumor Growth Cancer Cell, Volume 19, Issue 2, 15 February 2011, Pages 257–272 http://dx.doi.org/10.1016/j.ccr.2011.01.020
  21. Emilie Flaberg, Laszlo Markasz, Gabor Petranyi, Gyorgy Stuber, Ferenc Dicső, Nidal Alchihabi, Èva Oláh, István Csízy, Tamás Józsa, Ove Andrén, Jan-Erik Johansson, Swen-Olof Andersson, George Klein, & Laszlo Szekely High-throughput live-cell imaging reveals differential inhibition of tumor cell proliferation by human fibroblasts Cancer Cell Biology, Volume 128, 29 December 2010, Pages 2793-2802 http://dx.doi.org/10.1002/ijc.25612
  22. Christine L. Chaffer, Robert A. Weinberg A Perspective on Cancer Cell Metastasis Science, Volume 331, Issue 6024, 25 March 2011, Pages 1559-1564 https://dx.doi.org/10.1126/science.1203543
  23. Daniel G. Stover, Brian Bierie, & Harold L. Moses A delicate balance: TGF-β and the tumor microenvironment Journal of Cellular Biochemistry Volume 101, Issue 4, July 2007, Pages 851-861 http://dx.doi.org/10.1002/jcb.21149
  24. 24.0 24.1 http://www.nature.com/nri/posters/mdscs/nri1005_mdscs_poster.pdf Vincenzo Bronte and Dmitry Gabrilovich Myeloid-derived suppressor cells Nature
  25. Alberto Mantovani, Paola Allavena, Antonio Sica & Frances Balkwill Cancer-related inflammation Nature, Volume 454 24 July 2008 http://dx.doi.org/10.1038/nature07205
  26. 26.0 26.1 Rommel A. Mathias, Shashi K. Gopal, Richard J. Simpson Contribution of cells undergoing epithelial–mesenchymal transition to the tumour microenvironment Journal of Proteomics, Volume 78, 14 January 2013, Pages 545–557 http://dx.doi.org/10.1016/j.jprot.2012.10.016
  27. 27.0 27.1 Roberta Valenti, Veronica Huber, Manuela Iero, Paola Filipazzi, Giorgio Parmiani, & Licia Rivoltini Tumor-released microvesicles as vehicles of immunosuppression Cancer Research, Volume 67, 1 April 2007, Pages 2912-1915 http://dx.doi.org/10.1158/0008-5472.CAN-07-0520
  28. Frances Balkwill, Kellie A. Charles, & Alberto Mantovani Smoldering and polarized inflammation in the initiation and promotion of malignant disease Cancer Cell, Volume 7, Issue 3, March 2005, Pages 211–217 http://dx.doi.org/10.1016/j.ccr.2005.02.013
  29. Bin-Zhi Qian, Jeffrey W. Pollard Macrophage Diversity Enhances Tumor Progression and Metastasis Cell, Volume 141, Issue 1, 2 April 2010, Pages 39–51 http://dx.doi.org/10.1016/j.cell.2010.03.014
  30. 30.0 30.1 30.2 30.3 G. Solinas, G. Germano, A. Mantovani, & P. Allavena Tumor-associated macrophages (TAM) as major players of the cancer-related inflammation Journal of Leukocyte Biology Volume 86, Issue 5, November 2009, Pages 1065-1073
  31. Subhra K. Biswas, Lisa Gangi, Saki Paul, Tiziana Schioppa, Alessandra Saccani, Marina Sironi, Barbara Bottazzi, Andrea Doni, Bronte Vincenzo, Fabio Pasqualini, Luca Vago, Manuela Nebuloni, Alberto Mantovani, and Antonio Sica A distinct and unique transcriptional program expressed by tumor-associated macrophages (defective NF-κB and enhanced IRF-3/STAT1 activation) Blood, Volume 107, Issue 5, 3 November 2005, Pages 2112-2122 http://dx.doi.org/10.1182/blood-2005-01-0428
  32. Wei Zhang, Liang Wang, Daobin Zhou, Quancai Cui, Dachun Zhao, & Yongji Wu Expression of tumor-associated macrophages and vascular endothelial growth factor correlates with poor prognosis of peripheral T-cell lymphoma, not otherwise specified Leukemia & Lymphoma, Volume 52, Issue 1, January 2011, Pages 46-52 http://dx.doi.org/10.3109/10428194.2010.529204
  33. Bi Cheng Zhang, Juan Gao, Jun Wang, Zhi Guo Rao, Bao Cheng Wang, & Jian Fei Gao Tumor-associated macrophages infiltration is associated with peritumoral lymphangiogenesis and poor prognosis in lung adenocarcinoma Medical Oncology, Volume 28, Issue 4 December 2011, Pages 1447-1452 http://dx.doi.org/10.1007/s12032-010-9638-5
  34. Mei Yang, Jingqi Chen, Fang Su, Bin Yu, Fengxi Su, Ling Lin, Yujie Liu, Jian-Dong Huan, & Erwei Song Microvesicles secreted by macrophages shuttle invasion-potentiating microRNAs into breast cancer cells Molecular Cancer Volume 10, Issue 117, 22 September 2011, http://dx.doi.org/10.1186/1476-4598-10-117
  35. Simon Turcotte & Steven A. Rosenberg Immunotherapy of Metastatic Solid Cancers Advances in Surgery Volume 45, 2011, Pages 342-360
  36. Aled Clayton & Zsuzsanna Tabi Exosomes and the MICA-NKG2D system in cancer Blood Cells, Molecules, and Diseases, Volume 34, Issue 3, May–June 2005, Pages 206–213 http://dx.doi.org/10.1016/j.bcmd.2005.03.003
  37. 37.0 37.1 37.2 Thea D. Tisty & Lisa M. Coussens Tumor stroma and regulation of cancer development Annual Review of Pathology: Mechanisms of Disease Volume 1, February 2006, Pages 11-150 http://dx.doi.org/10.1146/annurev.pathol.1.110304.100224
  38. Cedric Gaggioli, Steven Hooper, Cristina Hidalgo-Carcedo, Robert Grosse, John F. Marshall, Kevin Harrington & Erik Sahai Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells Nature Cell Biology, Volume 9, 25 November 2007, Pages 1392-1400 http://dx.doi.org/10.1038/ncb1658
  39. 39.0 39.1 Serenella M. Pupa, Sylvie Ménard, Stefania Forti, & Elda Tagliabue New insights into the role of extracellular matrix during tumor onset and progression Journal of Cellular Physiology, Volume 192, Issue 3, September 2002, Pages 259–267 http://dx.doi.org/10.1002/jcp.10142
  40. Anthony M.P. Montgomery, Ralph A. Reisfeld, & David A. Cheresh Integrin αvβ3 rescues melanoma cells from apoptosis in three-dimensional dermal collagen Proceedings of the National Academy of Sciences of the United States of America, Volume 91, Issue 19, 13 September 1994, Pages 8856–8860
  41. Gabriele Bergers & Lisa M Coussens Extrinsic regulators of epithelial tumor progression: metalloproteinases Current Opinion in Genetics & Development, Volume 10, Issue 1, 1 February 2000, Pages 120–127 http://dx.doi.org/10.1016/S0959-437X(99)00043-X
  42. R. Sinkus, J. Lorenzen, D. Schrader, M. Lorenzen, M. Dargatz, D. Holz High-resolution tensor MR elastography for breast tumour detection Physics in Medicine and Biology, 45 (2000), pp. 1649–1664
  43. Kandice R. Levental, Hongmei Yu, Laura Kass, Johnathon N. Lakins, Mikala Egeblad, Janine T. Erler, Sheri F.T. Fong, Katalin Csiszar, Amato Giaccia, Wolfgang Weninger, Mitsuo Yamauchi, David L. Gasser, Valerie M. Weaver Matrix Crosslinking Forces Tumor Progression by Enhancing Integrin Signaling Cell, Volume 139, Issue 5, 25 November 2009, Pages 891–906 http://dx.doi.org/10.1016/j.cell.2009.10.027
  44. 44.0 44.1 44.2 Mraz, M.; Zent, C. S.; Church, A. K.; Jelinek, D. F.; Wu, X.; Pospisilova, S.; Ansell, S. M.; Novak, A. J.; Kay, N. E.; Witzig, T. E.; Nowakowski, G. S. (2011). "Bone marrow stromal cells protect lymphoma B-cells from rituximab-induced apoptosis and targeting integrin α-4-β-1 (VLA-4) with natalizumab can overcome this resistance". British Journal of Haematology 155 (1): 53–64. doi:10.1111/j.1365-2141.2011.08794.x. PMID 21749361.
  45. Sakae Unezaki, Kazuo Maruyama, Jun-Ichi Hosoda, Itsuro Nagae, Yasuhisa Koyanagi, Mikiho Nakata, Osamu Ishida, Motoharu Iwatsuru, Seishi Tsuchiya Direct measurement of the extravasation of polyethyleneglycol-coated liposomes into solid tumor tissue by in vivo fluorescence microscopy International Journal of Pharmaceutics, Volume 144, Issue 1, 22 November 1996, Pages 11–17 http://dx.doi.org/10.1016/S0378-5173(96)04674-1
  46. Seth Lilavivat , Debosmita Sardar , Subrata Jana , Geoffrey C. Thomas , and Kenneth J. Woycechowsky In vivo encapsulation of nucleic acids using an engineered nonviral protein capsid Journal of the American Chemical Society Volume 134, Issue 32, 15 August 2012, Pages 13152–13155 http://dx.doi.org/10.1021/ja302743g
  47. Srinivas Ramishetti & Leaf Huang Intelligent design of multifunctional lipid-coated nanoparticle platforms for cancer therapy Therapeutic Delivery Volume 3, Issue 12, December 2012, 1429-1445 http://dx.doi.org/doi: 10.4155/tde.12.127
  48. Rakesh K. Jain Transport of molecules in the tumor interstitium: a review Cancer Research, Volume 47, 1987, Pages 3039–3051
  49. Yehoshua Maor, Jinlong Yu, Paula M. Kuzontkoski, Bruce J. Dezube, Xuefeng Zhang, & Jerome E. Groopman Cannabidiol Inhibits Growth and Induces Programmed Cell Death in Kaposi Sarcoma–Associated Herpesvirus-Infected Endothelium Genes Cancer Volume 7-8, July 2012, Pages 512-520 http://dx.doi.org/10.1177/1947601912466556