Rakesh K. Jain and Dan G. Duda
A solid tumor is an organlike structure containing neoplastic and stromal cells nourished by the tumor vasculature composed of endothelial cells, basement membrane, and perivascular cells. All of these components are embedded in an extracellular matrix (Fig. 4.1). The interactions between these cells, their surrounding matrix, and their local microenvironment influence the expression of various genes. The products encoded by these genes, in turn, control the pathophysiologic characteristics of the tumor. The tumor pathophysiology affects tumor growth, invasion, and metastasis, as well as the response to radiation and other therapies. In this chapter, we will discuss various pathophysiologic parameters that characterize the vascular and extravascular compartments of a tumor as well as the molecular players involved in the formation and function of these compartments. Finally, we will point out some clinical implications of the findings and present a future perspective.
FIGURE 4.1. Schematic representation of a solid tumor—an organlike structure. The key components include cancer cells, host cells, and blood vasculature made of endothelial and perivascular cells—all embedded in a matrix bathed in interstitial fluid. The lymphatic vasculature, present in most normal tissues, is often lacking or is dysfunctional in solid tumors.

VASCULAR COMPARTMENT
Neoplastic cells, similar to normal cells, need oxygen and other nutrients for their survival and growth. Every reproductively intact normal cell in our body is located within 100 to 200 μm from a blood capillary so that it can receive adequate levels of oxygen and other nutrients by the process of diffusion. Likewise, cells undergoing neoplastic transformation depend on nearby capillaries for growth. These preneoplastic (i.e., dysplastic or hyperplastic) cells can grow as spherical or ellipsoidal cellular aggregates. However, once the size of the cellular aggregate reaches the diffusion limit for critical nutrients, the aggregate may become dormant. Indeed, human tumors may remain dormant for a number of years despite active cell proliferation because of a balance between proliferation and cell death. However, once they have access to new blood vessels, the tumor may grow and metastasize. What triggers the growth of new vessels? What molecular and cellular players are involved? How do these vessels compare with normal vessels with respect to their structure and function?
Angiogenesis
The fact that the vascular system is associated with tumor growth in animals and humans has been known for more than a century.1 Ide et al.2 and Algire and Chalkley3 provided powerful insight into the neovascularization of transplanted tumors using transparent window techniques (for reviews on this subject, see 4,5). In 1968 Rijhsinghani et al.6 and Ehrmann and Knoth7 suggested the possibility that tumors produce an “angiogenic” substance. In 1971, Folkman8,9 proposed the hypothesis that blocking angiogenesis should block tumor growth and metastasis. In 1978, Gullino10 demonstrated that a tissue acquires angiogenic capacity during neoplastic transformation and proposed that antiangiogenesis approaches be used to prevent cancer. Both of these hypotheses have been validated in a number of preclinical studies.11,12 A wide range of antiangiogenic strategies are currently being evaluated in the clinic to prevent or treat a large number of diseases, including cancer.11 Most importantly, antiangiogenic strategies have yielded overall survival benefits in patients with advanced colorectal cancer, non–small-cell lung cancer, renal cell cancer, hepatocellular carcinoma, and gastrointestinal stromal tumors and have shown increased response rates and progression-free survival benefits in advanced breast, medullary thyroid, and ovarian cancer; pancreatic neuroendocrine tumors (pNETs); and glioblastoma.13–21,22 This has led so far to U.S. Food and Drug Administration (FDA) approval for eight antiangiogenic drugs in the treatment of these diseases (Table 4.1).
The net balance between pro- and antiangiogenic factors governs both normal and pathologic angiogenic processes.11 This balance is spatially and temporally regulated under physiologic conditions so that the “angiogenic switch” is “on” when needed (e.g., during embryonic development, wound healing, formation of corpus luteum) and “off” otherwise. During neoplastic transformation and tumor progression, this regulation is deranged, which results in ectopically formed blood vessels to support the growing mass.11
Cellular Mechanisms
Several cellular mechanisms have been described in the vascularization of tumors: (a) co-option, (b) intussusception, (c) sprouting (angiogenesis), (d) vasculogenesis from endothelial precursors, (e) cancer cell lining of vessels (vascular mimicry), or (f) transdifferentiation to the endothelial cell (Fig. 4.2).11 Tumor cells can co-opt and grow around the existing vessels to form “perivascular” cuffs. However, as stated earlier, these cuffs cannot grow beyond the diffusion limit of critical nutrients, and they actually may cause the collapse of the vessels as a result of the growth pressure (referred to as “solid stress”).23,24,25 Alternatively, an existing vessel may enlarge in response to the growth factors released by tumors, and an interstitial tissue column may grow in the enlarged lumen and partition the lumen to form an expanded vascular network. This mode of intussusceptive microvascular growth has been observed during tumor growth, wound healing, and gene therapy.26,27
“Sprouting” angiogenesis is the most widely studied mechanism of vessel formation. During sprouting angiogenesis, the existing vessels become leaky in response to growth factors released by cancer or stromal cells; the basement membrane and the interstitial matrix dissolve; the pericytes dissociate from the vessel; endothelial cells (ECs) migrate and proliferate to form an array/sprout; a lumen is formed in the sprout (referred to as canalization); branches and loops are formed by confluence and anastomoses of sprouts to permit blood flow; and, finally, these immature vessels are invested in basement membrane and pericytes. During physiologic angiogenesis, these vessels differentiate into mature arterioles, capillaries, and venules, whereas in tumors they may remain immature.5,11,28
During mammalian embryonic development, a primitive vascular plexus is formed from angioblasts or endothelial precursor cells (EPCs) by a process referred to as vasculogenesis. Distinct signals specify arterial or venous differentiation.29 Several studies showed that circulating EPCs mobilized from the bone marrow or peripheral blood also can contribute to postnatal vasculogenesis in tumors and other tissues.30 Although debated, the repair of healthy adult vessels or the expansion of pathologic vessels can be aided by the recruitment of bone marrow–derived cells (BMDCs) and/or EPCs to the vascular wall.31 The progenitor cells then become incorporated into the endothelial lining in a process known as postnatal vasculogenesis. Collateral vessels, which bring bulk flow to ischemic tissues during revascularization, enlarge in size by distinct mechanisms, such as the attraction and activation of myeloid cells.31,32 However, this process appears to be of rather limited importance in tumor neovascularization.33–37
Tissues can also become vascularized by other mechanisms, but the relevance of these processes is not well understood. For example, tumor cells can line vessels—a phenomenon known as vascular mimicry. Putative cancer stemlike cells can even generate tumor endothelium.38,39–41
The challenge now is to discern the relative contribution of each of these mechanisms of new vessel formation in tumors to optimize antiangiogenic treatment of cancer.16,41,42
TABLE 4.1 FDA-APPROVED ANTI-ANGIOGENIC DRUGS

FIGURE 4.2. Modes of vessel formation in solid tumors. There are several known methods of blood vessel formation in normal tissues and tumors. A–C: Vessel formation can occur by sprouting angiogenesis (A), by the recruitment of bone marrow–derived and/or vascular wall–resident endothelial progenitor cells (EPCs) that differentiate into endothelial cells (ECs; B), or by a process of vessel splitting known as intussusception (C). D–F: Tumor cells can co-opt pre-existing vessels (D), or tumor vessels can be lined by cancer cells (vascular mimicry; E) or by endothelial cells, with cytogenetic abnormalities in their chromosomes, derived from putative cancer stem cells (F). Unlike normal tissues, which use sprouting angiogenesis, vasculogenesis, and intussusception (A–C), tumors can use all six modes of vessel formation (A–F). (Reproduced from Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature 2011;473:298–307, with permission.)

FIGURE 4.3. Molecular basis of vessel branching. The consecutive steps of blood vessel branching are shown, with the key molecular players involved denoted in parentheses. A: After stimulation with angiogenic factors, the quiescent vessel dilates and an endothelial-cell tip cell is selected (DLL4 and JAGGED1) to ensure branch formation. Tip-cell formation requires degradation of the basement membrane, pericyte detachment, and loosening of endothelial cell junctions. Increased permeability permits extravasation of plasma proteins (such as fibrinogen and fibronectin) to deposit a provisional matrix layer, and proteases remodel pre-existing interstitial matrix, all enabling cell migration. For simplicity, only the basement membrane between endothelial cells and pericytes is depicted, but in reality, both pericytes and endothelial cells are embedded in this basement membrane. B: Tip cells navigate in response to guidance signals (such as semaphorins and ephrins) and adhere to the extracellular matrix (mediated by integrins) to migrate. Stalk cells behind the tip cell proliferate, elongate, and form a lumen, and sprouts fuse to establish a perfused neovessel. Proliferating stalk cells attract pericytes and deposit basement membranes to become stabilized. Recruited myeloid cells such as tumor-associated macrophages (TAMs) and TIE-2–expressing monocytes (TEMs) can produce proangiogenic factors or proteolytically liberate angiogenic growth factors from the extracellular matrix. C: After fusion of neighboring branches, lumen formation allows perfusion of the neovessel, which resumes quiescence by promoting a phalanx phenotype, re-establishment of junctions, deposition of basement membrane, maturation of pericytes, and production of vascular maintenance signals. Other factors promote transendothelial lipid transport. (Reproduced from Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature 2011;473:298–307, with permission.)

Molecular Mechanisms
Various pro- and antiangiogenic molecules orchestrate different steps in vessel formation. Vascular endothelial growth factor (VEGF) is, perhaps, the most critical angiogenic molecule. Originally discovered in 1983 as the vascular permeability factor by Dvorak et al. and cloned in 1989 by Ferrara et al., VEGF increases vascular permeability, promotes migration and proliferation of ECs, serves as an EC survival factor, and is known to up-regulate leukocyte adhesion molecules on ECs.43–44,45 During tumor progression, the variety and concentration of angiogenic molecules produced by a tumor can increase. Thus, if VEGF were blocked, tumor growth might continue as a result of the action of other angiogenic molecules (e.g., basic fibroblast growth factor [bFGF], interleukin-8 [IL-8], stromal cell–derived factor 1α[SDF1α]).42,46,47 Other positive regulators include angiopoietins that are involved in blood vessel maturation;48 various proteases involved in extracellular matrix remodeling and growth factor release;11,28 and organ-specific angiogenic stimulators such as endocrine gland VEGF45 (Fig. 4.3).
Angiogenesis inhibitors include soluble receptors of various proangiogenic ligands as well as molecules that down-regulate stimulator expression (e.g., interferons), interfere with stimulator release, or block binding of stimulators to their receptors (e.g., platelet factor 4). Thrombospondins are among the first and best characterized endogenous inhibitors that interfere with the growth, adhesion, migration, and survival of ECs. Other endogenous inhibitors include fragments of various plasma or matrix proteins, for example, angiostatin—fragment of plasminogen;49 endostatin—fragment of collagen XVIII;50 and tumstatin—fragment of collagen IV.51
The generation of pro- and antiangiogenic molecules can be triggered by injury, metabolic stress (e.g., low partial pressure of oxygen [Po2], low pH, or hypoglycemia), mechanical stress (e.g., shear stress, solid stress), immune/inflammatory responses (e.g., immune/inflammatory cells that have infiltrated the tissue), and genetic mutations (e.g., activation of oncogenes or deletion of suppressor genes that control the production of angiogenesis regulators).52–55,56–57 These molecules can emanate from cancer cells, endothelial cells, stromal cells, blood, and extracellular matrix58,59,60 (Fig. 4.4). Because the host cells differ among organs, angiogenesis depends on host–tumor interactions.59,61–64,65 Furthermore, because the tumor microenvironment is likely to change during tumor growth, regression, and relapse after treatment, profiles of pro- and antiangiogenic molecules are likely to change with time and space.66–75 The challenge now is to develop a unified theoretic framework to describe the temporal and spatial profiles of this increasing number of angiogenesis regulators to develop effective therapeutic strategies.42,76
FIGURE 4.4. Tumor induction of host promoter activity in stromal cells. The expression of vascular endothelial growth factor (VEGF) in host cells can be examined using transgenic mice expressing green fluorescent protein (GFP) under the control of the VEGF promoter. A: A murine mammary carcinoma xenograft shows host cell VEGF expression mainly at the periphery of the tumor after 1 week. B: After 2 weeks, the VEGF-expressing host cells have infiltrated the tumor. (From Fukumura D, Xavier R, Sugiura T, et al. Tumor induction of VEGF promoter activity in stromal cells. Cell 1998;94:715–725, with permission). C: A GFP-expressing layer of host cells can be seen at the tumor–host interface. D, E: The VEGF-expressing host cells colocalize with the angiogenic tumor vessels. (From Brown EB, Campbell RB, Tsuzuki Y, et al. In vivo measurement of gene expression, angiogenesis and physiological function in tumors using multiphoton laser scanning microscopy. Nat Med 2001;7:866–870, with permission.)

Vascular Architecture
In normal tissue, blood flows in a closed circuit from an artery to an arteriole to capillaries to venules to a vein. Although the tumor vasculature originates from these host vessels and the mechanisms of angiogenesis are similar, its organization may be completely different depending on the tumor type, its location, and whether it is growing, regressing, or relapsing.77,78–79 In general, tumor vessels are dilated, saccular, tortuous, and chaotic in their patterns of interconnection. For example, whereas the normal vasculature is characterized by dichotomous branching, the tumor vasculature has many trifurcations and branches with uneven diameters.80,81 The fractal dimensions and minimum path lengths of tumor vasculature are different from those of the normal host vasculature.78,79,82
The molecular mechanisms of this abnormal vascular architecture are not entirely understood, but it seems reasonable to hypothesize that an imbalance of pro- and antiangiogenic molecules is a key contributor.11,83 By extension, modulation of this angiogenic imbalance may allow for the correction of the tumor vascular abnormalities, leaving behind a more structurally and functionally normal vascular bed. Several observations support this “normalization” hypothesis.83 Normalization of tumor xenograft vasculature is observed during therapies that lower VEGF (e.g., hormone withdrawal from a hormone-dependent tumor84), that interfere with VEGF signaling (e.g., treatment with anti-VEGF or anti-VEGF receptor-2 antibody or tyrosine kinase inhibitor85,86,87–90) (Fig. 4.5), or that mimic an antiangiogenic cocktail (e.g., trastuzumab treatment of a HER2 overexpressing tumor70). Emerging clinical data from cancer patients treated with bevacizumab, an anti-VEGF antibody, or pan-vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors AZD2171 (cediranib) or sunitinib lend even more compelling support to this hypothesis.16,41,66,67,69,71,72,73,75,83,91,92 More crucially, the extent of vascular normalization directly correlates with the survival of patients with recurrent glioblastoma.92 Moreover, the patients whose tumor blood flow increased most after cediranib treatment had the longest overall survival.93,94
Mechanical stress generated by proliferating tumor cells also may lead to partially compressed or totally collapsed vessels often found in tumors.23,95 Decompression of blood vessels by depleting cells or matrix supports this mechanical hypothesis.24,25,96 Perhaps the combination of both molecular and mechanical factors renders the tumor vasculature abnormal; thus, both must be taken into account when designing novel strategies for cancer treatment.
FIGURE 4.5. Normalization of tumor vasculature. A: Normal vessels are well organized with even diameters. B: In contrast, tumor vessels are tortuous with increased vessel diameter, length, density, and permeability. C: Antiangiogenic therapies “normalize” the tumor vascular network and ultimately may reduce the vasculature to the point that it provides inadequate support for tumor growth. (From Jain RK. Normalizing tumor vasculature with anti-angiogenic therapy: a new paradigm for combination therapy. Nat Med 2001;7:987–989; and Tong RT, et al. Vascular normalization by vascular endothelial growth factor receptor 2 blockade induces a pressure gradient across the vasculature and improves drug penetration in tumors. Cancer Res 2004;64:3731–3736, with permission.)

Blood Flow and Microcirculation
Blood flow in a vascular network—whether normal or abnormal—is governed by arteriovenous pressure difference and flow resistance. The flow resistance is a function of the vascular architecture and the blood viscosity.97Abnormalities in both the vasculature and blood viscosity increase the resistance to blood flow in tumors.80,81,98–100 As a result, overall perfusion rates (blood flow rate per unit volume) in tumors are lower than in many normal tissues.101–103
Macroscopically and microscopically, tumor blood flow is temporally and spatially heterogeneous. Macroscopically, four spatial regions can be recognized in a tumor: an avascular necrotic region, a seminecrotic region, a stabilized microcirculation region, and an advancing front12,104 (Fig. 4.6A). Microscopically, in normal tissues red blood cell (RBC) velocity is dependent on vessel diameter, but there is no such dependence in most tumors.61,65,105Furthermore, the average RBC velocity may be an order of magnitude lower in some tumors compared to the host tissue.65 In a given tumor vessel, blood flow fluctuates with time and can even reverse its direction.104,105–106
In addition to the elevated geometric and viscous (rheologic) resistance, other molecular and mechanical factors contribute to this spatial and temporal heterogeneity. These include imbalance between pro- and antiangiogenic molecules,105 solid stress generated by proliferating cancer cells,23,24,25,95,96 vascular remodeling by intussusception, and coupling between luminal and interstitial fluid pressure via hyperpermeability of tumor vessels.78,108–110 As discussed later, this heterogeneity contributes to both acute and chronic hypoxia in tumors—a potential cause of resistance to radiation and other therapies and increased metastatic potential.
Considerable effort has gone into modulating tumor blood flow to improve cancer treatment. This has been difficult to achieve reproducibly because the tumor vasculature consists of both vessels co-opted from the pre-existing host vasculature and vessels resulting from the angiogenic response of host vessels to cancer cells. The former are invested in normal contractile perivascular cells, whereas the latter either lack perivascular cells or are abnormally invested.111–113 As a result, efforts to increase tumor blood flow and the delivery of cytotoxins, by pharmacologic or physical agents, have not always been successful.97,114 In contrast, efforts to “starve” tumors by decreasing or shutting down tumor blood flow by “stealing” blood away from the passive component of the tumor vasculature by vasodilators114 as well as by vascular targeting or intravascular coagulation have shown promise in experimental systems.115 It also appears that judiciously applied antiangiogenic therapy may “normalize” the abnormal tumor vasculature and the resulting “normalized” vessels might be more responsive to vasoactive agents12,16,41,83,116 (Fig. 4.5).
FIGURE 4.6. The tumor microenvironment is heterogeneous with proliferative, quiescent, and necrotic regions. A: These regions can be characterized in terms of various physiologic parameters. Decreasing magnitude of these parameters is indicated as + + +, + +, +, +/–, and – in the adjoining table. (From Jain RK, Forbes NS. Commentary–can engineered bacteria help control cancer? Proc Natl Acad Sci U S A 2001;98:14748–14750, with permission.) B: pH and Po2 as a function of distance from a blood vessel in a tumor. The tumor environment becomes progressively more hypoxic and acidic farther away from a blood vessel. (From Helmlinger G, Yuan F, Dellian M, et al. Interstitial pH and Po2 gradients in solid tumors in vivo: high-resolution measurements reveal a lack of correlation. Nat Med 1997;3:177–182, with permission.)

Vascular Permeability
Once a bloodborne molecule has reached an exchange vessel, its extravasation occurs by diffusion, convection, and, to some extent, presumably transcytosis.117 The diffusive permeability of a molecule depends on the size, shape, charge, and flexibility of the molecule as well as the size, shape, charge, and dynamics of the transvascular transport pathway. In normal vessels, these pathways include diffusion through the EC membrane (for lipophilic solutes), trans-EC diffusion, interendothelial junctions (<7 nm), open or closed fenestrations (<10 nm), and transendothelial channels (including vesicles or vesicovacuolar organelles [VVOs]).118 Some of these pathways may be lined with glycocalyx on ECs, thus effectively reducing the size of the pathway. A basement membrane may retard further the movement of molecules. Ultrastructural studies show widened interendothelial junctions; an increased number of fenestrations, vesicles, and VVOs in tumor vessels; and a lack of normal basement membrane and pericytes.43,63,118,119–120
In concert with these ultrastructural findings, both vascular permeability to solutes and water permeability (referred to as hydraulic conductivity) of tumors, in general, are significantly higher than that of various normal tissues.65,100,121–124 Furthermore, unlike normal vessels, tumor vessels lack selectivity for the size of extravasating molecules.125 However, positively charged molecules have a higher affinity for the negatively charged angiogenic tumor vessels.126 Despite increased overall permeability, not all blood vessels of a tumor are leaky. Even the leaky vessels have a finite pore size that is tumor dependent, and ultrastructural studies show that the larger pore size in tumors represents wide interendothelial junctions.63,120 Not only do the vascular permeability and pore size vary from one tumor to the next, but also within the same tumor they vary both spatially and temporally as well as during tumor growth, regression, and relapse.63,70,84
The local microenvironment plays an important role in controlling vascular permeability. For example, a human glioma (HGL21) is fairly leaky when grown subcutaneously in immunodeficient mice, but it exhibits blood–brain barrier properties in the cranial window.65 Such site-dependent differences for other tumors have been observed in other orthotopic sites.61,64,127 One possible explanation is that the host–tumor interactions control the production and secretion of cytokines associated with permeability increase (e.g., VEGF) and decrease (e.g., angiopoietin 1).12,28,48,90,128 A better understanding of the molecular mechanisms of permeability regulation in tumors is likely to yield strategies for improved delivery of molecular medicine to tumors.129
Movement of Cells Across Vessel Walls
Both cancer cells and immune cells frequently move across the walls of blood vessels—the former in the process of metastasis and the latter during immune response or cell-based immunotherapy. Both transendothelial and periendothelial pathways have been proposed as a route for intravasation and extravasation of cells. Very little is known about intravasation, except that a tumor may shed more than a million cells per gram per day and most of these are not clonogenic, and that some may be shed as fragments along with stromal cells.103,130–132 More is known about the molecular and cellular mechanisms of extravasation.133–135 A cell within a blood vessel may continue to move with the flowing blood, collide with the vessel wall, adhere transiently or stably, and finally extravasate. These interactions are governed by both local hydrodynamic forces and adhesive forces. The former are determined by the vessel diameter and fluid velocity, and the latter by the expression, strength, and kinetics of binding between adhesion molecules and by the surface area of contact.133,136,137–140 Deformability of cells affects both types of forces.141
Rolling of endogenous leukocytes is generally low in tumor vessels, whereas stable adhesion (≥30 seconds) is comparable between normal and tumor vessels.142 However, both rolling and stable adhesion are nearly zero in angiogenic vessels induced in collagen gels by bFGF or VEGF, two of the most potent angiogenic factors. Whether the latter is due to a low flux of leukocytes into angiogenic vessels and/or down-regulation of adhesion molecules in these immature vessels is currently not known. Age may also play an important role in leukocyte–endothelial interactions.143
Further insight into the biology of cells that adhere to tumor vessels comes from studies on the localization of IL-2–activated natural killer (A-NK) cells in normal and tumor tissues in mice using positron emission tomography.144,145 Immediately following systemic injection, these cells localized primarily in the lungs, whereas a nondetectable number of cells arrived in the tumor.144 Increased rigidity caused by IL-2 activation may contribute to the mechanical entrapment of these cells in the lung microcirculation.146,147 Constitutive expression of certain adhesion molecules in the lung vasculature also may facilitate their localization in the lungs.133 One approach to reducing lung entrapment is to reduce the rigidity of these cells.141,145 Alternatively, entrapment in lung vasculature can be circumvented by injecting A-NK cells directly into the blood supply of tumors. In this case, A-NK cells, both xenogeneic and syngeneic, adhered to some blood vessels in three different tumor models145,148,149 via CD18 and very late antigen-4 (VLA-4) on the A-NK cells and intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and E-selectin on the activated endothelium of angiogenic vessels.44,150,151
These molecules can be up-regulated by a number of cytokines, including tumor necrosis factor-α (TNF-α) and a protein of 90 kD molecular weight (p90); secreted by some neoplastic cells;44,137 and down-regulated by others, for example, transforming growth factor-β (TGF-β) also, presumably, secreted by cancer cells.62,152 Surprisingly, the proangiogenic VEGF also can up-regulate these molecules, whereas another proangiogenic molecule, bFGF, can down-regulate these molecules.44,59,86,153 The challenge now is to decrease nonspecific entrapment of immune cells in normal vessels and to increase their delivery to tumor vessels to improve various cell-based therapies, including gene therapy. Judicious doses of anti-VEGF agents that “normalize” tumor vasculature can potentially realize this goal.154,155
EXTRAVASCULAR COMPARTMENT
Composition and Origin
The extravascular compartment of a solid tumor consists of neoplastic cells (parenchyma) and host cells (e.g., inflammatory cells, fibroblasts) residing in an interstitial subcompartment bathed by the interstitial fluid (Fig. 4.1). Depending on the tumor type and its stage of differentiation, neoplastic cells may be dispersed in the matrix as individual cells (e.g., lymphomas, melanomas) or as clumps or nests (e.g., carcinomas). More than 80% of tumors are carcinomas arising from epithelial cells. The remaining 20% include sarcomas arising from mesenchymal cells (e.g., bone or muscle cells), lymphomas arising from lymphoid tissue, leukemias arising from hematopoietic cells, and hemangiomas arising from endothelial cells. In a poorly differentiated carcinoma, the cancer cells may be loosely packed in clumps, whereas in a well-differentiated carcinoma, the cells may be connected with intercellular junctions and tightly packed in a nest enveloped by a basement membrane. With tumor progression, cancer cells may invade the basement membrane and spread to other regions.43
Unlike cancer cells, host cells must migrate into the tumor from normal tissue. Inflammatory cells may enter the tumor via blood vessels or may infiltrate from the adjacent tissue.131 Other host cells, such as fibroblasts, may proliferate and migrate from the adjacent connective tissue28,58,60,156,157 or from primary tumor to the metastatic site.131 Increasingly, infiltrating host cells are being recognized as critical modulators of the tumorigenic process. For example, mesenchymal cells generically termed carcinoma-associated fibroblasts have been shown to promote tumor growth, metastasis, and angiogenesis, potentially through the secretion of stromal cell–derived factor-1α.131,158–161Furthermore, tumor-associated immune/inflammatory cells such as tumor-associated macrophages (TAMs), Tie2-expressing monocytes (TEMs), or Gr-1+ myeloid-derived suppressor cells (MDSCs) have been linked to both cancer immunosurveillance and suppression as well as to tumor promotion.54,159,162,163,164 The challenge now is to establish approaches for skewing the TAMs toward a phenotype that promotes antitumor immune responses in cancer patients.54
The interstitial compartment of a tumor is bounded by the walls of the blood vessels on one side and by the membranes of cancer and stromal cells on the other. In normal tissues, the blood vessels are surrounded by a basement membrane, which is defective in tumors (see the Vascular Permeability section).28,83 In addition, functional lymphatics may be confined to the tumor margin (see the Lymphatic Transport section).165,166 Similar to normal tissues, the interstitial space of tumors is composed of a collagen and elastin fiber network that provides structural support to the tissue. Interdispersed in this cross-linked structure are the interstitial fluid and macromolecular constituents (polysaccharides hyaluronan and proteoglycans [PGs]), which form a hydrophilic gel.
Compared with our understanding of blood vessel formation, our understanding of stroma generation is minimal. Dvorak has proposed that the extravasated plasma protein fibrinogen, a key component of the tumor interstitial fluid (TIF), clots to form fibrin, which serves as a major component of the provisional stroma.43 This provisional stroma eventually is replaced by more mature connective tissue stroma. The TIF also contains several proteins including fibronectin, vitronectin, osteopontin, thrombospondin, decorin, and tenascin. These proteins are present in both free and bound forms and contain the amino acid sequence arg-gly-asp (RGD). The RGD sequence provides a binding site for adhesion that assists in the migration of various cells, including stromal cells. In addition to extravasating from the leaky tumor vessels, these proteins, along with collagen and various PGs, also are synthesized by the stromal cells, albeit in a form that is different from that in the plasma or normal tissues.43 TIF also may contain various growth factors that facilitate stroma formation. For example, in vitro studies suggest that platelet-derived growth factor-βis involved in the recruitment of fibroblasts to tumors, and TGF-β controls the production of collagen and other matrix molecules in tumors.28,167 With the increasing interest in using the fragments of matrix constituents for controlling angiogenesis, our understanding of the molecular and cellular mechanisms of stroma generation in tumors is likely to increase.156
FIGURE 4.7. Schematic of lymphatics in low (left) versus high (right) vascular endothelial growth factor-C (VEGF-C)—expressing tumors. VEGF-C secreted from tumor cells stimulates vascular endothelial growth factor receptor-3 (VEGFR-3) expressed in lymphatic endothelial cells, inducing peritumor lymphatic hyperplasia (top right). An increase in lymphatic surface area may increase the opportunity for tumor cell entry into lymphatic vessels. Augmented lymph flow enhances tumor cell delivery to draining lymph nodes (bottom right). In the absence of VEGF-C overexpression, peritumor lymphatic hyperplasia is less pronounced and fewer tumor cells are delivered to draining lymph nodes (left). Anti-VEGF-C/anti-VEGFR-3 treatment inhibits VEGF-C–induced lymphatic hyperplasia and tumor cell delivery to draining lymph nodes (bottom left). (From Hoshida T, Isaka N, Hagendoorn J, et al. Imaging steps of lymphatic metastasis reveals that vascular endothelial growth factor-C increases metastasis by increasing delivery of cancer cells to lymph nodes: therapeutic implications. Cancer Res2006;66:8065–8075, with permission.)

Interstitial Transport
Once a molecule has extravasated, its movement through the interstitial space occurs by diffusion and convection.118 Diffusion is proportional to the concentration gradient in the interstitium, and convection is proportional to the interstitial fluid velocity, which, in turn, is proportional to the pressure gradient in the interstitium. Just as the interstitial diffusion coefficient, D (cm2/s), relates the diffusive flux to the concentration gradient, the interstitial hydraulic conductivity, K (cm2/mm Hg · s), relates the interstitial velocity to the pressure gradient.118 Values of these transport coefficients are governed by the structure and composition of the interstitial compartment as well as by the physicochemical properties of the solute molecule.126,168,169–172
The value of K (interstitial hydraulic conductivity) for a human colon carcinoma xenograft (LS174 T), measured using two different methods,173,174 was found to be higher than that of a hepatoma,172 which, in turn, was higher than that of the normal liver. Using fluorescence recovery after photobleaching, the diffusion coefficient (D) of various molecules in tumors was found to be about one-third that in water175 and higher than the values in the host tissue.169 Collagen content and structure have a significant effect on D in tumors.171,173,176 This is surprising because hyaluronan and proteoglycans, and not collagen, account for most of the resistance to transport in normal tissues. Because collagen is produced by the host-derived cells (e.g., fibroblasts), the penetrability of macromolecules into a tumor will depend on the host–tumor interaction. Thus, agents that interfere with collagen synthesis and/or organization (e.g., relaxin, bacterial collagenase, losartan) may increase interstitial transport in tumors.168,177,178
The time constant for a molecule with diffusion coefficient D to diffuse across a distance L is approximately L2/4D. For diffusion of IgG (immunoglobulin G) in tumors, this time constant is on the order of 1 hour for a 100-μm distance, days for a 1-mm distance, and months for a 1-cm distance. So for a 1-mm tumor, diffusional transport across the tumor would take days, and for a 1-cm tumor, it would take months. If the central vessels have collapsed completely as a result of cellular proliferation23,24,96 and interstitial matrix rearrangement, the reduced delivery of macromolecules by blood flow would make diffusion the primary mechanism of delivery to this hypoxic center. Binding may further retard the transport in tumors.175,179,180 The role of binding is illustrated clearly by comparing the rate of fluorescence recovery of a photobleached spot in tumor tissue injected with a nonspecific versus specific IgG. In addition to the heterogeneity of D in tumors, the most unexpected result of these photobleaching studies was the large extent (30% to 40%) of nonspecific binding.175 These results collectively suggest that the interstitial compartment of a tumor can be a formidable barrier to the uniform delivery of therapeutic macromolecules (e.g., antibodies, genes using viruses, nano-therapeutics) in tumors, and strategies are needed to overcome this barrier.117,126,177,178,181
Lymphatic Transport
In most normal tissues, extravasated plasma and macromolecules are taken up by the lymphatics and returned to the central circulation. Although it is widely accepted that lymphatic vessels are present in the tumor margin and the peritumoral tissue, the hotly debated issue for nearly a century has been whether anatomically defined lymphatic vessels are present within solid tumors and, if so, whether they function.166,182 Currently available immunohistochemical markers stain for structures in some tumors that resemble lymphatic vessels. However, because many of these markers lack specificity,165,166,183 it is not clear whether they stain functional lymphatic vessels, endothelial cells from remnant lymphatic vessels, or some other structures or cell types (e.g., preferential fluid channels173). It is likely that the stress induced by proliferating cancer cells compresses and impairs lymphatic vessels that are co-opted or formed in a tumor23,24 and/or lymphatic valves are impaired by tumor growth.184 The impaired lymphatic vessels, in turn, may contribute to the interstitial hypertension characteristic of animal and human tumors (see the Interstitial Hypertension section). In addition, invasion of the functional peritumoral lymphatics is considered to be a poor prognostic factor for a number of tumors, and lymphatic metastasis is a major cause of morbidity and mortality.
Our understanding of the mechanisms of lymphangiogenesis lags behind our understanding of the mechanisms of angiogenesis. However, considerable progress has recently been made toward identifying molecular players responsible for lymphangiogenesis. VEGF-C, acting through VEGFR-3, appears to play a central role in tumor-associated lymphangiogenesis. Several experimental185–186,187–189 and clinical190 studies have demonstrated a positive correlation between VEGF-C expression and peritumoral lymphatic vessel density, lymphatic metastasis, and, in some cases, poor clinical outcomes. Like in vascular angiogenesis, other positive and negative regulators, such as VEGF,191,192 VEGF-D,186 hepatocyte growth factor, platelet-derived growth factor-BB, and angiopoietins are involved in lymphangiogenesis.11,190 Furthermore, mechanisms analogous to co-option, intussusception, sprouting, and vasculogenesis may operate in lymphatic growth11 (see the Angiogenesis: Cellular Mechanisms section). Similar to organ-specific angiogenic molecules (e.g., EG-VEGF)45 and blood vascular endothelial precursor cells,30,193 there may be organ-specific lymphangiogenic molecules and lymphatic endothelial precursor cells that contribute to tumor-associated lymphangiogenesis.194 Moreover, the proteolytic processing of lymphangiogenesis molecules, as well as the phenotype and function of the resulting lymphatics, may depend on the tumor type as well as on the host organ in which the tumor is growing.28,86,165,190
The precise roles for these lymphangiogenic molecules in the induction of lymphatic metastasis are imperfectly understood. Recent data demonstrate that tumor VEGF-C overexpression induces peritumoral lymphatic hyperplasia through activation of VEGFR-3. Consequently, lymph fluid volumetric flow increases.185 This results in increased tumor cell delivery to lymph nodes and a higher rate of lymphatic metastasis185 (Fig. 4.7). It remains unclear how VEGF-C overexpression impacts tumor cell entry into lymphatic vessels; however, an attractive hypothesis is that the increased lymphatic surface area simply increases the probability of tumor cell entry and dissemination. Alternatively, VEGF-C may stimulate the release of a chemotactic factor that recruits tumor cells into lymphatic vessels. Of potential clinical importance, VEGFR-3 blockade was shown to inhibit VEGF-C–induced lymphatic hyperplasia, tumor cell delivery to draining lymph nodes, and lymphatic metastasis when treatment was started at the time of tumor initiation. However, lymphatic metastases were not significantly reduced if VEGFR-3 blockade was started after tumor cell seeding of draining lymph nodes.185,195 These data suggest that anti-VEGFR-3 therapy may be effective in preventing, but not treating, lymphatic metastases—the more common clinical imperative—except in cases where a significant fraction of vascular endothelial cells and/or cancer cells express VEGFR-3. The challenge now is to identify alternative strategies for treating lymphatic metastases, either through combination therapy (e.g., anti-VEGFR-2/anti-VEGFR-3) or modulation of other pathways.
Mechanical signals that trigger the lymphangiogenic switch are unknown. Because lymphatic vessels help maintain the balance of fluid in tissues, hydrostatic pressure is a likely trigger.195,197 Whether the lymphatic hyperplasia seen in tumor margins is, in part, a response to the elevated hydrostatic pressure in tumors and whether the newly formed lymphatics remain open and relieve this pressure is an open question. Techniques such as microlymphangiography,4,165,166,185,188,198,199 fluorescence photobleaching lymph flow quantitation,182,185,198,200–202 and optical frequency domain imaging (OFDI)203 will allow us to answer these important questions.
Interstitial Hypertension
Unlike normal tissues, where the interstitial fluid pressure (IFP) is around 0 mm Hg, both animal and human tumors exhibit interstitial hypertension.69,71,73,118,166,174,204,205,206,207–211,212,213–215 The tumor IFP begins to increase as soon as the host vessels become leaky in response to angiogenic molecules such as VEGF.216 Thus, IFP can be lowered by inhibiting the VEGF pathway using blocking antibodies.69,71,72,73,88,217 The IFP increases with tumor size in some tumors204,206,210 and remains independent of tumor size in others.211
Three mechanisms contribute to the interstitial hypertension in tumors. In normal tissues, lymphatics maintain fluid homeostasis; thus, the lack of functional lymphatics within tumors is a key contributor. Indeed, DiResta et al. have shown that one could lower the IFP by placing “artificial lymphatics” in tumors.218 The second contributor is the leaky nature of tumor vessels. As a result, the hydrostatic and oncotic (colloid osmotic) pressures become almost equal between the intravascular and extravascular space.82,88,205,219 At least two pieces of evidence support this hypothesis. First, lowering permeability by blocking VEGF signaling lowers IFP.71,88,217 Second, IFP goes up and down with the microvascular pressure within seconds.220–222 The two mechanisms described so far can only explain hypertension up to 20 to 30 mm Hg—the microvascular pressure of most exchange vessels in our body—but IFPs as high as 94 mm Hg have been measured in human tumors.214 Because the microvascular pressure (MVP) is the driving force for IFP in tumors, these tumors must have a high MVP. Indeed, this is the case.205 There are two possible explanations for the elevated MVP in tumors: (a) the tumor vessels have reduced arterial resistance so that the MVP becomes closer to arterial pressure, and/or (b) the tumor vessels have increased venous resistance as a result of compression and tortuosity so that the whole vascular network is under hypertension. Indirect evidence for the latter comes from the decrease in IFP following decompression of tumor vessels by drug-induced apoptosis of perivascular cancer cells.24,96
The elevated pressure can compromise the tumor microcirculation and delivery of therapeutics in three ways. First, reduced transmural pressure gradients resulting from equilibrium between MVP and IFP reduce convection across tumor vessels and thus compromise the transport of macromolecules.82,88,97,205,221 Second, because IFP is nearly uniform throughout a tumor and drops precipitously in the tumor margin, the interstitial fluid “oozes” out of the tumor into the surrounding normal tissue, carrying macromolecules with it.15,35,130 Finally, transmural coupling between IFP and MVP as a result of high permeability of tumor vessels can lead to blood flow stasis in tumors without physically occluding the vessels.108–110 Thus, decreasing vascular leakiness might restore the transmural pressure gradients and potentially resume/re-establish blood flow in the nonperfused regions of tumors. Some direct and indirect antiangiogenic therapies may “normalize” the tumor vasculature through this mechanism12,16,41,67,70,71,83,116,223 (Tables 4.2 and 4.3 and Fig. 4.5).
TABLE 4.2 STUDIES REPORTING ANTIANGIOGENIC THERAPY–INDUCED IMPROVEMENT IN TUMOR OXYGENATION

Metabolic Environment
Hypoxia
A key function of the vasculature is to provide adequate levels of nutrients to the parenchymal cells and to remove waste products. Based on the anatomy of the capillary bed and a mathematical model of oxygen diffusion and consumption, the Nobel laureate August Krogh introduced the concept of a diffusion limit for oxygen of 100 to 200 μm nearly a century ago.224 This unit of tissue—a single capillary surrounded by a 100 to 200 μm radius cylinder—is referred to as a Krogh cylinder in physiology. Nearly 50 years later, Thomlinson and Gray identified similar “cords” in human lung cancer and found necrotic cells beyond 180 μm away from blood vessels, presumably due to a lack of oxygen.225 This is referred to as chronic hypoxia or diffusion-limited hypoxia. Although various hypoxia markers and microelectrodes have suggested these gradients, the first direct measurements of these perivascular Po2gradients, as well as perivascular pH gradients, became possible only with the development of phosphorescence quenching microscopy23,226 (Fig. 4.6B).
As discussed earlier, blood flow in tumor vessels is intermittent, and, thus, some regions of a tumor are periodically starved for oxygen. The resulting hypoxia is referred to as acute hypoxia or perfusion-limited hypoxia. A necessary consequence of intermittent blood flow is the resumption of blood flow after shutdown, and the resulting production of free radicals can lead to ischemia-reperfusion injury or reoxygenation injury; thus, applying additional selection pressure on cancer cells can cause them to become more locally aggressive, metastatic, and resistant to therapy.227
TABLE 4.3 STUDIES REPORTING THE IMPACT OF ANTIANGIOGENIC/VASCULAR NORMALIZATION STRATEGIES UPON DELIVERY OF THERAPEUTIC COMPOUNDS/SYSTEMICALLY ADMINISTERED MOLECULES INTO TUMORS

Low pH
Another consequence of the abnormal microcirculation of the tumor is low extracellular pH. There are at least two sources of H+ ions in tumors—lactic acid and carbonic acid.228 The former results from glycolysis, and the latter results from conversion of CO2 and H2O via carbonic anhydrase. However, the intracellular pH of cancer cells remains neutral or alkaline (≥7.2) despite the acidic extracellular pH. Because carbonic anhydrase-9 and various glucose transporters (GLUT-1, -3) and enzymes in the glycolytic pathway are up-regulated by hypoxia,227 one would expect low extracellular pH and hypoxia to track each other and to colocalize with regions of low blood flow. It is surprising that there is a lack of spatial correlation among these parameters—a discovery made possible by recent developments in optical techniques that permit the simultaneous high-resolution mapping of multiple physiologic parameters.23 A potential explanation for this lack of concordance is that some perfused tumor vessels carry hypoxic blood.23 Thus, although they may not be able to deliver enough oxygen to the surrounding cells, they may be able to carry away the waste products (e.g., lactic acid).
Therapeutic Consequences
The presence of molecular oxygen during irradiation can “fix” biologic (e.g., DNA) free radicals, making radiation-induced damage irreparable (oxygen fixation hypothesis). Thus, hypoxia reduces the radiation sensitivity of neoplastic and normal cells both in vitro and in vivo. Similarly, hypoxia can compromise the efficacy of some chemotherapeutics. Independently, hypoxia can increase the metastatic potential of cancer cells.227 Therefore, for nearly half a century considerable preclinical and clinical effort has been focused on alleviating hypoxia through a multitude of interventions such as improving tumor perfusion with mild hyperthermia or drugs, increasing oxygen content of the blood via hyperbaric oxygenation, and increasing hemoglobin/hematocrit by transfusion or exogenous erythropoietin. Unfortunately, the clinical outcomes have not met expectations. Although early studies suggested a marked benefit of transfusion in anemic cervical cancer patients undergoing definitive radiotherapy, careful analysis suggests that these studies are confounded by selection biases that preclude the conclusion that anemia correction by transfusion impacts outcome.229,230 Furthermore, erythropoietin (or analog) treatment showed encouraging survival results in anemic cancer patients receiving nonplatinum chemotherapy and in anemic lung cancer patients receiving chemotherapy.231,232 However, subsequent trials in anemic head and neck cancer patients and mainly nonanemic metastatic breast cancer patients actually suggested outcomes may be impaired by erythropoietic agents.233,234 There are multiple possible reasons such interventions have yielded mixed results. These include the inability to increase tumor Po2 as markedly as systemic Po2,235 the inability to increase Po2 in all areas of a tumor to optimal levels due to abnormal vasculature,82,116 and undesired “off-target” effects of interventions (e.g., immunosuppression with transfusion236–238). Furthermore, tumors may reoxygenate during radiation therapy with standard fractionation, potentially minimizing the impact of providing additional oxygen to the target tissue.
Similarly, low extracellular pH can adversely (or favorably) affect the uptake and cytotoxicity of some therapeutics. The pH gradient difference between tumor and normal tissue may offer a tumor-specific target for weak acid chemotherapeutics for the treatment of cancer.239,240 The development of specific drugs that exploit this pH difference and strategies to modulate pH in tumors have not yet reached the clinic but are anticipated.227
Two broad strategies targeting the unique tumor metabolic environment are emerging: (a) exploit hypoxia to activate drugs or attract tumoricidal anaerobic bacteria and (b) dissect hypoxia-induced pathways to identify novel targets for drug development. The first strategy has led to the development of drugs such as tirapazamine and to renewed interest in bacteriolytic therapy;241 both approaches are in clinical trials, but promising data are yet to emerge242 (see Trial Identifier NCT00358397 at ClinicalTrials.gov). The second strategy has revealed several molecular players in the physiologic and pathophysiologic response to hypoxia.227,243,244 The balance between hypoxia-induced apoptosis/necrosis on one hand and the increased resistance to cell death mediated by various hypoxia-induced pathways on the other determines whether a tumor can survive and even grow under hypoxic conditions. Ultimately, hypoxia selects for tumor cells that are more malignant, more invasive, and genetically unstable, rendering them resistant to various therapies. Therefore, certain players in the hypoxia-induced pathways now are being targeted in the development of diagnostic and therapeutic agents. Hypoxia-induced pathways include genes involved in oxygen delivery, glycolysis and glucose uptake, pH control, stress-response pathways, growth factor signaling, angiogenesis, transcription, apoptosis, growth inhibition, and invasion and metastasis (Fig. 4.3 and Box 4.1).11,243
Of the various molecular players involved in sensing and responding to hypoxia, hypoxia-inducible factor-1 a (HIF-1α) has received the most attention. This transcription factor is up-regulated in a number of human tumors.243,245Regulated by proline and asparagine hydroxylases, HIF-1α activates genes involved in an array of physiologic responses including angiogenesis, vasodilation, glycolysis, and RBC production by binding to the hypoxia-response element (HRE). Although HIF-1α is an attractive therapeutic target, its pleiotropic action may prove to be a major challenge for clinical exploitation. For example, teratomas arising from HIF-1α(–/–) embryonic cells grow more rapidly despite lower levels of VEGF and angiogenesis.246 This counterintuitive finding may be a result of the ability of HIF-1α(–/–) cells to survive under hypoxic conditions, instead of undergoing apoptosis.60 HIF-1α has also been shown to play an important role in determining tumor radioresponsiveness through the regulation of multiple, and sometimes opposing, processes.247 Under some circumstances, HIF-1α inhibition reduces tumor cell radiosensitivity by protecting hypoxic cells from radiation-induced apoptosis and enhancing clonogenic survival potentially through reductions in adenosine triphosphate metabolism, cellular proliferation, and p53 activation.247 Furthermore, HIF-1αserves a key function in inflammatory cell energy metabolism, and its inhibition results in profound immunodeficiency.248 Consequently, molecular therapies that target HIF-1α or HRE, as well as more selective therapies that target key downstream effectors of HIF-1α, are under intensive investigation for cancer detection and treatment.227,243,249
Box 4.1
Hypoxia and Epigenetic Regulation of Angiogenesis
The prolyl hydroxylase domain (PHD) proteins PHD1–3 are oxygen-sensing enzymes that hydroxylate the hypoxia-inducible factor (HIF) proteins HIF-1α and HIF-2α when sufficient oxygen is available. Once hydroxylated, HIFs are targeted for proteasomal degradation.268 Under hypoxia, PHDs become inactive, and HIFs initiate broad transcriptional responses to increase the oxygen supply by angiogenesis, through the up-regulation of angiogenic factors such as vascular endothelial growth factor (VEGF).269 HIFs are also activated in nonhypoxic conditions by oncogenes and growth factors, allowing tumor cells to stimulate angiogenesis before they become deprived of oxygen. In general, HIF-1α promotes vessel sprouting, whereas HIF-2α mediates vascular maintenance.269 Reduced HIF-1α levels in mice impair embryonic vascular development, revascularization of ischemic tissues, and angiogenesis in injured tissues and tumors.269 The use of HIF-1α inhibitors to block tumor or ocular angiogenesis has therefore received attention. Conversely, Hif1 α gene transfer in mice or activation of HIF-1α by pharmacologic blockade of PHDs promotes ischemic tissue revascularization.
HIF-1α also regulates tumor angiogenesis indirectly, by releasing chemoattractants such as stromal cell–derived factor 1α (SDF-1α) to recruit proangiogenic bone marrow–derived cells (BMDCs).270 Gene silencing of Phd2 in mouse tumor cells enhances vessel growth by similar mechanisms. Hypoxia also regulates the polarization and proangiogenic activity of tumor-associated macrophages (TAMs) by means of HIF-1α and HIF-2α with different effects.268 That hypoxia and inflammation are closely intertwined is illustrated by the finding that signaling by HIF-1α and nuclear factor-κB cross-activate each other. In certain cases, hypoxic up-regulation of VEGF occurs independently of HIF-1α and is mediated by the metabolic regulator peroxisome proliferator-activated receptor gamma coactivator (PGC)-1α in preparation for oxidative metabolism once the ischemic tissue is revascularized.271Because HIF signaling contributes to acquired resistance against anti-VEGF therapy, the combined blockade of VEGF and HIF-1α is being explored as a cancer treatment strategy.
There is increasing evidence for epigenetic control of angiogenesis, particularly by noncoding microRNAs (miRNAs),272 which induce messenger RNA degradation or block translation. Because miRNAs target multiple genes, they are well positioned to regulate complex processes such as angiogenesis. Endothelial cells express several miRNAs that are induced by hypoxia or VEGF. Most of those stimulate angiogenesis by hijacking proangiogenic cascades while suppressing angiostatic pathways.273 The expression of miR-126 is induced by the mechanosensitive transcription factor KLF2A and integrates the mechanosensory stimulus of blood flow to shape the vascular system.274 Endothelial cell–specific loss of DICER, an exonuclease involved in miRNA biogenesis, impairs pathologic angiogenesis. Angiogenic miRNAs seem to offer significant pro- or antiangiogenic potential.
Reproduced with permission from Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature 2011; 473:298–307.
CLINICAL IMPLICATIONS
Two major problems currently plague the nonsurgical treatment of malignant solid tumors. First, physiologic barriers within tumors impede the delivery of therapeutics and oxygen (a key radiation sensitizer) at effective concentrations to all cancer cells.41,83,250 Second, inherent or acquired resistance resulting from genetic and epigenetic mechanisms reduces the effectiveness of conventional as well as novel therapies.251 Can we take advantage of the unique pathophysiology of tumors to overcome these problems for better management of cancer? As discussed next, recent clinical data offer some hope.
Prognostic/Predictive Biomarker Implications
Multiple indices of tumor pathophysiology have been evaluated as potential predictors of treatment outcome including vessel density (reviewed in 42,252), oxygen level (reviewed in243,253), interstitial pressure,42,212,214,230 and blood or urine circulating molecules73,91 (reviewed in42). Vessel density can be evaluated in biopsies and is measured either in “hot spots” (i.e., regions of most active angiogenesis) or in the tissue as a whole. The former presumably provides a measure of a tumor’s aggressiveness, and the latter reflects the status of global oxygenation. Most studies to date show that poor outcome of radiation therapy correlates with high vessel density in “hot spots” and/or low overall microvessel density. There are, however, several studies showing a lack of correlation or an opposite correlation. This discrepancy may be the result of the morphometric techniques used or of differences in tumor types or treatment schedules.
The oxygen level in a tumor also has a potential prognostic value, and it can be directly measured with microelectrodes. Alternatively, immunohistochemical analysis of tumor tissue for endogenous or exogenous hypoxic markers (e.g., HIF-1α, glucose transporter-1, carbonic anhydrase-9, pimonidazole) can be used as a surrogate for tumor oxygenation status. However, immunohistochemical assessments of hypoxia do not necessarily correlate with oxygen status measured directly with microelectrodes.254 A concerted effort is under way to assess hypoxia using novel, noninvasive imaging techniques.255,256 Several studies have shown that tumor hypoxia is a predictor of a poor outcome of radiation therapy when used alone or in combination with other therapies. These findings are consistent with in vitro and in vivo preclinical studies showing the adverse effect of hypoxia on radiation responses.
Because the IFP is a reflection of the global physiology of tumors, a correlation between tumor IFP and the response to radiation therapy has been suggested. One cervical cancer study has shown that elevated tumor IFP can, indeed, independently predict a poor outcome of radiation therapy.230 Further studies are needed to evaluate the prognostic significance of IFP in tumors. However, one potential application of the steep rise of pressure at the tumor periphery is improved localization of tumors before their removal.
Finally, circulating biomarkers may provide information about tumor pathophysiology and its changes after treatment. Of note, some of the emerging biomarkers—such as circulating collagen IV or soluble VEGFR-1—may represent biomarkers of vascular normalization and, if validated, could be useful in treatment decisions.91,92
Although each of these approaches has advantages, key disadvantages include their invasiveness and their potential for sampling error. With rapid developments in the field of noninvasive imaging, it is likely that the measurement of various physiologic and molecular parameters in tumors will become more refined and convenient for patients. Examples of such imaging approaches include blood oxygen level–dependent magnetic resonance imaging (BOLD MRI), electron paramagnetic resonance spectroscopy/imaging, and [18F]-misonidazole positron emission tomography (FMISO-PET).255–256,257–259 The promise of such imaging approaches has just started to be realized. FMISO-PET has been evaluated in a substudy of patients with stage III or IV squamous cell carcinoma of the head and neck randomized to concurrent radiotherapy with either tirapazamine and cisplatin or infusional fluorouracil and cisplatin. Pretreatment FMISO-PET–detected hypoxia was associated with a higher risk of locoregional recurrence among patients who did not receive the tirapazamine-containing regimen compared to patients who did receive tirapazamine.259 This study suggests that FMISO-PET can provide clinically meaningful information about tumor physiology and simultaneously provides evidence that tirapazamine acts by specifically targeting hypoxic tumor cells. Such progress will continue and physiologic/molecular profiles of patients’ tumors will yield improved and better-tailored therapies for individual patients.
FIGURE 4.8. Vascular “normalization” in rectal cancer patients following treatment with the anti–vascular endothelial growth factor (VEGF) antibody, bevacizumab. A–C: Tumor vessel “normalization” following a single injection of bevacizumab is suggested by the reduction of tumor microvessel density (A), by the increase in fraction of tumor vessels with pericyte coverage (B), and by the drop in interstitial fluid pressure (C). D: Positron emission tomography reveals no change in 18-fluorodeoxyglucose (FDG) uptake after a single dose of bevacizumab and complete resolution of FDG uptake following neoadjuvant chemoradiation (bevacizumab, 5-fluorouracil, pelvic external-beam radiation therapy). The stability of FDG uptake following bevacizumab monotherapy, despite marked reductions in microvessel density, suggests the efficiency of residual tumor blood vessels after bevacizumab treatment is improved. (From Willett CG, Boucher Y, di Tomaso E, et al. Direct evidence that the VEGF-specific antibody bevacizumab has antivascular effects in human rectal cancer. Nat Med 2004;145–147, with permission.)

Therapeutic Implications
Given the physiologic barriers to the delivery and effectiveness of various therapeutics, a strategy that is gaining increasing interest is targeting the tumor vasculature. This strategy has the advantage of targeting ECs that are easily accessible to a bloodborne drug and are presumably genetically stable. In addition, each EC supports multiple cancer cells, thus providing “therapeutic amplification.” However, the inability to target all ECs in a tumor can reduce the effectiveness of antivascular therapy. Similarly, the dependence of ECs on multiple angiogenic molecules can limit the effectiveness of various antiangiogenic therapies when used alone.41,83 These challenges may explain why currently available antiangiogenic agents, although demonstrating biologic activity, are unable to provide durable tumor control when used as monotherapy.
Although of limited utility when used alone, the judicious combination of antiangiogenic therapies with conventional cytotoxic therapies has led to improved tumor control in mice and lengthened survival in certain types of human tumors14,16,17,240,260–263 (see Table 4.1). For example, in two human tumor xenograft models, a VEGFR-2–blocking antibody decreased the dose of fractionated radiation required to control 50% of tumors (TCD50) by 11 to 27 Gy without modifying in-field skin reactions.240 Thus, to maximize clinical gains, these agents must be employed in combination with radiation and chemotherapy. The challenge now is to optimally combine these therapies in patients. Destruction of tumor vasculature by antiangiogenic agents should antagonize chemo- and radiotherapy by compromising the delivery of therapeutics and oxygen, respectively. However, judiciously applied antiangiogenic therapy can prune inefficient tumor vessels and render the remaining vasculature more efficient (Fig. 4.5).12,41,83,116,223 This “normalization” of tumor vasculature has been demonstrated in various preclinical models (reviewed in41,83) and in rectal cancer, hepatocellular carcinoma, ovarian carcinoma, and glioblastoma patients66,67,69,71,73,75 (Fig. 4.8). Vascular “normalization” should result in improved delivery of cytotoxic chemotherapy and radiosensitizing oxygen, thereby improving tumor control. This principle has been rigorously tested in animal models83,89,260,264–266 and confirmatory data on the impact of vascular “normalization” on patient outcomes await mature results of ongoing and future clinical trials.
ACKNOWLEDGMENTS
This chapter is an update of the chapter published in the fifth edition of Principles and Practice of Radiation Oncology and based on a review article by Carmeliet and Jain, Nature 2011.11 The work summarized here was supported by continuous support from the National Cancer Institute since 1980 to RKJ. We want to acknowledge the support through grants P01CA80124, R01CA115767, R01CA85140, R01CA126642, and Federal Share/NCI Proton Beam Program Income (RKJ), and R21CA139168, R01CA159258, and Federal Share/NCI Proton Beam Program Income (DGD), as well as the National Foundation for Cancer Research Grant and Department of Defense Breast Cancer Research Innovator Award W81XWH-10-1-0016 (RKJ), and the American Cancer Society Research Grant RSG-11-073-01-TBG (DGD).
DISCLOSURE
R.K.J. has research grants from Dyax, MedImmune, and Roche; serves as a consultant to Noxxon Pharmaceuticals; serves on the Scientific Advisory Board of Enlight and SynDevRx; serves on the Board of Directors of XTuit; serves on the Board of Trustees of H&Q Healthcare Investors and H&Q Life Sciences Investors; and has equity in Enlight, SynDevRx and XTuit Pharmaceuticals.
REFERENCES
1. Goldman E. The growth of malignant disease in man and the lower animals with special reference to the vascular system. Lancet 1907;2:1236–1240.
2. Ide AG, Baker NH, Warren SL. Vascularization of the Brown-Pearce rabbit epithelioma transplant as seen in the transplant ear chamber. Am J Radiol 1939;42:891–899.
3. Algire GH, Chalkley HW. Vascular reactions of normal and malignant tissues in vivo. I. Vascular reactions of mice to wounds and to normal and neoplastic transplants. J Natl Cancer Inst 1945;6:73–85.
4. Fukumura D, Duda DG, Munn LL, et al. Tumor microvasculature and microenvironment: novel insights through intravital imaging in pre-clinical models. Microcirculation 2010;17(3):206–225.
5. Jain RK, Munn LL, Fukumura D. Dissecting tumour pathophysiology using intravital microscopy. Nat Rev Cancer 2002;2(4):266–276.
6. Rijhsinghani K, Greenblatt M, Shubik P. Vascular abnormalities induced by benzo[a]pyrene: an in vivo study in the hamster cheek pouch. J Natl Cancer Inst 1968;41(1):205–216.
7. Ehrmann RL, Knoth M. Choriocarcinoma. Transfilter stimulation of vasoproliferation in the hamster cheek pouch. Studied by light and electron microscopy. J Natl Cancer Inst 1968;41(6):1329–1341.
8. Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med 1971;285(21):1182–1186.
9. Folkman J. Angiogenesis: an organizing principle for drug discovery? Nat Rev Drug Discov 2007;6(4):273–286.
10. Gullino PM. Angiogenesis and oncogenesis. J Natl Cancer Inst 1978;61(3):639–643.
11. Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature 2011;473:298–307.
12. Jain RK. Normalizing tumor vasculature with anti-angiogenic therapy: a new paradigm for combination therapy. Nat Med 2001;7(9):987–989.
13. Demetri GD, van Oosterom AT, Garrett CR, et al. Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial. Lancet2006;368(9544):1329–1338.
14. Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 2004;350(23):2335–2342.
15. Llovet J, Ricci S, Mazzaferro V, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med 2008;359(4):378–390.
16. Jain RK, Duda DG, Clark JW, et al. Lessons from phase III clinical trials on anti-VEGF therapy for cancer. Nat Clin Pract Oncol 2006;3(1):24–40.
17. Sandler A, Gray R, Perry MC, et al. Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N Engl J Med 2006;355(24):2542–2550.
18. Miller K, Wang M, Gralow J, et al. Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer. N Engl J Med 2007;357(26):2666–2676.
19. Motzer RJ, Hutson TE, Tomczak P, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med 2007;356(2):115–124.
20. Cloughesy TF, Prados MD, Wen PY, et al. A phase II, randomized, non-comparative clinical trial of the effect of bevacizumab alone or in combination with irinotecan (CPT-11) on 6-month progression free survival in recurrent, treatment-refractory glioblastoma. J Clin Oncol 2008;26S:abstract 2010b.
21. Raymond E, Dahan L, Raoul JL, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med 2011;364(6):501–513.
22. Wells SAJ, Gosnell JE, Gagel RF, et al. Vandetanib for the treatment of patients with locally advanced or metastatic hereditary medullary thyroid cancer. J Clin Oncol 2010;28(5):767–772.
23. Helmlinger G, Netti PA, Lichtenbeld HC, et al. Solid stress inhibits the growth of multicellular tumor spheroids. Nat Biotechnol 1997;15(8):778–783.
24. Padera TP, Stoll BR, Tooredman JB, et al. Pathology: cancer cells compress intratumour vessels. Nature 2004;427(6976):695.
25. Stylianopoulos T, Martin JD, Chauhan VP, et al. Causes, consequences, and remedies for growth-induced solid stress in murine and human tumors. Proc Natl Acad Sci USA 2012;109:15101–15108.
26. Patan S, Munn LL, Jain RK. Intussusceptive microvascular growth in a human colon adenocarcinoma xenograft: a novel mechanism of tumor angiogenesis. Microvasc Res 1996;51(2):260–272.
27. Patan S, Munn LL, Tanda S, et al. Vascular morphogenesis and remodeling in a model of tissue repair: blood vessel formation and growth in the ovarian pedicle after ovariectomy. Circ Res2001;89(8):723–731.
28. Jain RK. Molecular regulation of vessel maturation. Nat Med 2003;9(6):685–693.
29. Swift MR, Weinstein BM. Arterial-venous specification during development. Circ Res 2009;104(5):576–588.
30. Rafii S, Lyden D, Benezra R, et al. Vascular and haematopoietic stem cells: novel targets for anti-angiogenesis therapy? Nat Rev Cancer 2002;2(11):826–835.
31. Jones R, Capen DE, Jacobson M, et al. VEGFR2+PDGFRbeta+ circulating precursor cells participate in capillary restoration after hyperoxia acute lung injury (HALI). J Cell Mol Med 2009;13(9B):3720–3729.
32. Schaper W. Collateral circulation: past and present. Basic Res Cardiol 2009;104(1):5–21.
33. Duda DG, Cohen KS, Kozin SV, et al. Evidence for incorporation of bone marrow-derived endothelial cells into perfused blood vessels in tumors. Blood 2006;107(7):2774–2776.
34. Kozin SV, Kamoun W, Huang Y, et al. Recruitment of myeloid but not endothelial precursor cells facilitates tumor re-growth after local irradiation. Cancer Res 2010;70(14):5679–5685.
35. Purhonen S, Palm J, Rossi D, et al. Bone marrow-derived circulating endothelial precursors do not contribute to vascular endothelium and are not needed for tumor growth. Proc Natl Acad Sci U S A2008;105(18):6620–6625.
36. Peters BA, Diaz LA, Polyak K, et al. Contribution of bone marrow-derived endothelial cells to human tumor vasculature. Nat Med 2005;11(3):261–262.
37. Kozin SV, Duda DG, Munn LL, et al. Is vasculogenesis crucial for the regrowth of irradiated tumours? Nat Rev Cancer 2011;11(7):532.
38. Ricci-Vitiani L, Pallini R, Biffoni M, et al. Tumour vascularization via endothelial differentiation of glioblastoma stem-like cells. Nature 2010;468:824–828.
39. Soda Y, Marumoto T, Friedmann-Morvinski D, et al. Transdifferentiation of glioblastoma cells into vascular endothelial cells. Proc Natl Acad Sci U S A 2011;108(11):4274–4280.
40. Wang R, Chadalavada K, Wilshire J, et al. Glioblastoma stem-like cells give rise to tumour endothelium. Nature 2010;468:829–833.
41. Carmeliet P, Jain RK. Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nature Rev Drug Discov 2011;10(6):417–427.
42. Jain RK, Duda DG, Willett CG, et al. Biomarkers of response and resistance to antiangiogenic therapy. Nat Rev Clin Oncol 2009;6(6):327–338.
43. Dvorak HF. Vascular permeability factor/vascular endothelial growth factor: a critical cytokine in tumor angiogenesis and a potential target for diagnosis and therapy. J Clin Oncol 2002;20(21):4368–4380.
44. Melder RJ, Koenig GC, Witwer BP, et al. During angiogenesis, vascular endothelial growth factor and basic fibroblast growth factor regulate natural killer cell adhesion to tumor endothelium. Nat Med1996;2(9):992–997.
45. Chung AS, Lee J, Ferrara N. Targeting the tumour vasculature: insights from physiological angiogenesis. Nat Rev Cancer 2010;10(7):505–514.
46. Mizukami Y, Jo WS, Duerr EM, et al. Induction of interleukin-8 preserves the angiogenic response in HIF-1alpha-deficient colon cancer cells. Nat Med 2005;11(9):992–997.
47. Yoshiji H, Harris SR, Thorgeirsson UP. Vascular endothelial growth factor is essential for initial but not continued in vivo growth of human breast carcinoma cells. Cancer Res 1997;57(18):3924–3928.
48. Yancopoulos GD, Davis S, Gale NW, et al. Vascular-specific growth factors and blood vessel formation. Nature 2000;407(6801):242–248.
49. O’Reilly MS, Holmgren L, Shing Y, et al. Angiostatin: a novel angiogenesis inhibitor that mediates the suppression of metastases by a Lewis lung carcinoma. Cell 1994;79(2):315–328.
50. O’Reilly MS, Boehm T, Shing Y, et al. Endostatin: an endogenous inhibitor of angiogenesis and tumor growth. Cell 1997;88(2):277–285.
51. Maeshima Y, Sudhakar A, Lively JC, et al. Tumstatin, an endothelial cell-specific inhibitor of protein synthesis. Science 2002;295(5552):140–143.
52. Fukumura D, Xu L, Chen Y, et al. Hypoxia and acidosis independently up-regulate vascular endothelial growth factor transcription in brain tumors in vivo. Cancer Res 2001;61:6020–6024.
53. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144(5):646–674.
54. Huang Y, Snuderl M, Jain RK. Polarization of tumor-associated macrophages: a novel strategy for vascular normalization and antitumor immunity. Cancer Cell 2011;19(1):1–2.
55. Jain RK, Duda DG. Role of bone marrow-derived cells in tumor angiogenesis and treatment. Cancer Cell 2003;3(6):515–516.
56. Kerbel RS. Tumor angiogenesis. N Engl J Med 2008;358(19):2039–2049.
57. Xu L, Fukumura D, Jain RK. Acidic extracellular pH induces vascular endothelial growth factor (VEGF) in human glioblastoma cells via ERK1/2 MAPK signaling pathway. Mechanism of low pH induced VEGF. J Biol Chem2002;277:11368–11374.
58. Fukumura D, Xavier R, Sugiura T, et al. Tumor induction of VEGF promoter activity in stromal cells. Cell 1998;94(6):715–725.
59. Tsuzuki Y, Fukumura D, Oosthuyse B, et al. Vascular endothelial growth factor (VEGF) modulation by targeting HIF-1α->HRE-> VEGF cascade differentially regulates vascular response and growth rate in tumors. Cancer Res2000;60:6248–6252.
60. Brown EB, Campbell RB, Tsuzuki Y, et al. In vivo measurement of gene expression, angiogenesis and physiological function in tumors using multiphoton laser scanning microscopy. Nat Med2001;7(7):864–868.
61. Fukumura D, Yuan F, Monsky WL, et al. Effect of host microenvironment on the microcirculation of human colon adenocarcinoma. Am J Pathol 1997;151(3):679–688.
62. Gohongi T, Fukumura D, Boucher Y, et al. Tumor-host interactions in the gallbladder suppress distal angiogenesis and tumor growth: involvement of transforming growth factor beta1. Nat Med1999;5(10):1203–1208.
63. Hobbs SK, Monsky WL, Yuan F, et al. Regulation of transport pathways in tumor vessels: role of tumor type and microenvironment. Proc Natl Acad Sci U S A 1998;95(8):4607–4612.
64. Monsky WL, Carreira CM, Tsuzuki Y, et al. Role of host microenvironment in angiogenesis and microvascular functions in human breast cancer xenografts: mammary fat pad vs. cranial tumors. Clin Cancer Res 2002;8:1008–1013.
65. Yuan F, Salehi HA, Boucher Y, et al. Vascular permeability and microcirculation of gliomas and mammary carcinomas transplanted in rat and mouse cranial windows. Cancer Res 1994;54(17):4564–4568.
66. Batchelor TT, Duda DG, di Tomaso E, et al. Phase II study of cediranib, an oral pan-vascular endothelial growth factor receptor tyrosine kinase inhibitor, in patients with recurrent glioblastoma. J Clin Oncol 2010;28(17):2817–2823.
67. Batchelor TT, Sorensen AG, di Tomaso E, et al. AZD2171, a pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor vasculature and alleviates edema in glioblastoma patients. Cancer Cell2007;11(1):83–95.
68. Gerstner ER, Eichler AF, Plotkin SR, et al. Phase I trial with biomarker studies of vatalanib (PTK787) in patients with newly diagnosed glioblastoma treated with enzyme inducing anti-epileptic drugs and standard radiation and temozolomide. J Neurooncol 2011;103(2):325–332.
69. Horowitz NS, Penson RT, Duda DG, et al. Safety, efficacy, and biomarker exploration in a phase II study of bevacizumab, oxaliplatin and gemcitabine in recurrent mullerian carcinoma. Clin Ov Cancer2011;4(1):26–33.
70. Izumi Y, Xu L, di Tomaso E, et al. Tumour biology: herceptin acts as an anti-angiogenic cocktail. Nature 2002;416(6878):279–280.
71. Willett CG, Boucher Y, di Tomaso E, et al. Direct evidence that the VEGF-specific antibody bevacizumab has antivascular effects in human rectal cancer. Nat Med 2004;10(2):145–147.
72. Willett CG, Boucher Y, Duda DG, et al. Surrogate markers for antiangiogenic therapy and dose-limiting toxicities for bevacizumab with radiation and chemotherapy: continued experience of a phase I trial in rectal cancer patients. J Clin Oncol 2005;23(31):8136–8139.
73. Willett CG, Duda DG, di Tomaso E, et al. Efficacy, safety, and biomarkers of neoadjuvant bevacizumab, radiation therapy, and fluorouracil in rectal cancer: a multidisciplinary phase II study. J Clin Oncol2009;27(18):3020–3026.
74. Yoon SS, Duda DG, Karl DL, et al. Phase II study of neoadjuvant bevacizumab and radiotherapy for resectable soft tissue sarcomas. Int J Radiat Oncol Biol Phys 2011;81(4):1081–1090.
75. Zhu AX, Sahani DV, Duda DG, et al. Efficacy, safety, and potential biomarkers of sunitinib monotherapy in advanced hepatocellular carcinoma: a phase II study. J Clin Oncol 2009;27(18):3027–3035.
76. Zhu AX, Duda DG, Sahani DV, et al. HCC and angiogenesis: possible targets and future directions. Nat Rev Clin Oncol 2011;8(5):292–301.
77. Baish JW, Stylianopoulos T, Lanning RM, et al. Scaling rules for diffusive drug delivery in tumor and normal tissues. Proc Natl Acad Sci U S A 2011;108(5):1799–1803.
78. Baish JW, Jain RK. Fractals and cancer. Cancer Res 2000;60(14):3683–3688.
79. Gazit Y, Baish JW, Safabakhsh N, et al. Fractal characteristics of tumor vascular architecture during tumor growth and regression. Microcirculation 1997;4(4):395–402.
80. Less JR, Posner MC, Skalak T, et al. Geometric resistance to blood flow and vascular network architecture in human colorectal carcinoma. Microcirculation 1997;4:25–33.
81. Less JR, Skalak TC, Sevick EM, et al. Microvascular architecture in a mammary carcinoma: branching patterns and vessel dimensions. Cancer Res 1991;51(1):265–273.
82. Jain RK, Tong RT, Munn LL. Effect of vascular normalization by antiangiogenic therapy on interstitial hypertension, peritumor edema, and lymphatic metastasis: insights from a mathematical model. Cancer Res2007;67(6):2729–2735.
83. Goel S, Duda DG, Xu L, et al. Normalization of the vasculature for treatment of cancer and other diseases. Physiol Rev 2011;91(3):1071–1121.
84. Jain RK, Safabakhsh N, Sckell A, et al. Endothelial cell death, angiogenesis, and microvascular function after castration in an androgen-dependent tumor: role of vascular endothelial growth factor. Proc Natl Acad Sci U S A1998;95(18):10820–10825.
85. Chae S-S, Kamoun W, Farrar C, et al. Angiopoietin-2 interferes with anti-VEGFR-2-induced vessel normalization and survival benefit in mice bearing gliomas. Clin Cancer Res 2010;16:3618–3627.
86. Kadambi A, Mouta Carreira C, Yun CO, et al. Vascular endothelial growth factor (VEGF)-C differentially affects tumor vascular function and leukocyte recruitment: role of VEGF-receptor 2 and host VEGF-A. Cancer Res2001;61(6):2404–2408.
87. Kamoun WS, Ley CD, Farrar CT, et al. Edema control by cediranib, a vascular endothelial growth factor receptor-targeted kinase inhibitor, prolongs survival despite persistent brain tumor growth in mice. J Clin Oncol2009;27(15):2542–2552.
88. Tong RT, Boucher Y, Kozin SV, et al. Vascular normalization by vascular endothelial growth factor receptor 2 blockade induces a pressure gradient across the vasculature and improves drug penetration in tumors. Cancer Res2004;64(11):3731–3736.
89. Winkler F, Kozin SV, Tong R, et al. Kinetics of vascular normalization by VEGFR2 blockade governs brain tumor response to radiation: role of oxygenation, angiopoietin-1 and matrix metalloproteinases. Cancer Cell2004;6:553–563.
90. Yuan F, Chen Y, Dellian M, et al. Time-dependent vascular regression and permeability changes in established human tumor xenografts induced by an anti-vascular endothelial growth factor/vascular permeability factor antibody. Proc Natl Acad Sci U S A 1996;93(25):14765–14770.
91. Duda DG, Willett CG, Ancukiewicz M, et al. Plasma soluble VEGFR-1 is a potential dual biomarker of response and toxicity for bevacizumab with chemoradiation in locally advanced rectal cancer. Oncologist 2010;15(6):577–583.
92. Sorensen AG, Batchelor TT, Zhang WT, et al. A “vascular normalization index” as potential mechanistic biomarker to predict survival after a single dose of cediranib in recurrent glioblastoma patients. Cancer Res2009;69(13):5296–5300.
93. Sorensen AG, Emblem KE, Polaskova P, et al. Increased survival of glioblastoma patients who respond to anti-angiogenic therapy with elevated blood perfusion. Cancer Res 2012;72:402–407.
94. Gerstner ER, Emblem KE, Chi AS, et al. Effects of cediranib, a VEGF signaling inhibitor, in combination with chemoradiation on tumor blood flow and survival in newly diagnosed glioblastoma. J Clin Oncol 2012;30 (suppl; abstr 2009).
95. Koike C, McKee T, Pluen A, et al. Solid stress facilitates spheroid formation. Br J Cancer 2002;86:947–953.
96. Griffon-Etienne G, Boucher Y, Brekken C, et al. Taxane-induced apoptosis decompresses blood vessels and lowers interstitial fluid pressure in solid tumors: clinical implications. Cancer Res1999;59(15):3776–3782.
97. Jain RK. Determinants of tumor blood flow: a review. Cancer Res 1988;48:2641–2658.
98. Sevick EM, Jain RK. Geometric resistance to blood flow in solid tumors perfused ex vivo: effects of tumor size and perfusion pressure. Cancer Res 1989;49:3506–3512.
99. Sevick EM, Jain RK. Viscous resistance to blood flow in solid tumors: effect of hematocrit on intratumor blood viscosity. Cancer Res 1989;49:3513–3519.
100. Sevick EM, Jain RK. Measurement of capillary filtration coefficient in a solid tumor. Cancer Res 1991;51:1352–1355.
101. Jain RK, Shah SA, Finney PL. Continuous noninvasive monitoring of pH and temperature in rat Walker 256 carcinoma during normoglycemia and hyperglycemia. J Natl Cancer Inst 1984;73(2):429–436.
102. Vaupel P, Kallinowski F, Okunieff P. Blood flow, oxygen and nutrient supply, and metabolic microenvironment of human tumors: a review. Cancer Res 1989;49(23):6449–6465.
103. Butler TP, Gullino PM. Quantitation of cell shedding into efferent blood of mammary adenocarcinoma. Cancer Res 1975;35(3):512–516.
104. Endrich B, Reinhold HS, Gross JF, et al. Tissue perfusion inhomogeneity during early tumor growth in rats. J Natl Cancer Inst 1979;62(2):387–395.
105. Leunig M, Yuan F, Menger MD, et al. Angiogenesis, microvascular architecture, microhemodynamics, and interstitial fluid pressure during early growth of human adenocarcinoma LS174 T in SCID mice. Cancer Res1992;52(23):6553–6560.
106. Brizel DM, Klitzman B, Cook JM, et al. A comparison of tumor and normal tissue microvascular hematocrits and red cell fluxes in a rat window chamber model. Int J Radiat Oncol Biol Phys1993;25(2):269–276.
107. Ramanujan S, Koenig GC, Padera TP, et al. Local imbalance of proangiogenic and antiangiogenic factors: a potential mechanism of focal necrosis and dormancy in tumors. Cancer Res 2000;60(5):1442–1448.
108. Baish JW, Netti PA, Jain RK. Transmural coupling of fluid flow in microcirculatory network and interstitium in tumors. Microvasc Res 1997;53:128–141.
109. Mollica F, Jain RK, Netti PA. A model for temporal heterogeneities of tumor blood flow. Microvasc Res 2003;65(1):56–60.
110. Netti PA, Roberge S, Boucher Y, et al. Effect of transvascular fluid exchange on arterio-venous pressure relationship: implication for temporal and spatial heterogeneities in tumor blood flow. Microvasc Res 1996;52:27–46.
111. Armulik A, Genove G, Betsholtz C. Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. Dev Cell 2011;21(2):193–215.
112. Kashiwagi S, Izumi Y, Gohongi T, et al. NO mediates mural cell recruitment and vessel morphogenesis in murine melanomas and tissue-engineered blood vessels. J Clin Invest 2005;115(7):1816–1827.
113. Morikawa S, Baluk P, Kaidoh T, et al. Abnormalities in pericytes on blood vessels and endothelial sprouts in tumors. Am J Pathol 2002;160:985–1000.
114. Jain RK, Ward-Hartley KA. Tumor blood flow: characterization, modifications and role in hyperthermia. IEEE Transactions in Sonics and Ultrasonics; Special Issue on Hyperthermia, SU-31 1984:504–526.
115. Heath VL, Bicknell R. Anticancer strategies involving the vasculature. Nat Rev Clin Oncol 2009;6(7):395–404.
116. Jain RK. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science 2005;307(5706):58–62.
117. Jain RK, Stylianopoulos T. Delivering nanomedicine to solid tumors. Nat Rev Clin Oncol 2010;7(11):653–664.
118. Jain RK. Transport of molecules across tumor vasculature. Cancer Metastasis Rev 1987;6:559–594.
119. Baluk P, Morikawa S, Haskell A, et al. Abnormalities of basement membrane on blood vessels and endothelial sprouts in tumors. Am J Pathol 2003;163(5):1801–1815.
120. Hashizume H, Baluk P, Morikawa S, et al. Openings between defective endothelial cells contribute to tumor vessel leakiness. Am J Pathol 2000;156:1363–1380.
121. Endo M, Jain RK, Witwer B, et al. Water channel (aquaporin 1) expression and distribution in mammary carcinomas and glioblastomas. Microvasc Res 1999;58(2):89–98.
122. Gerlowski LE, Jain RK. Microvascular permeability of normal and neoplastic tissues. Microvasc Res 1986;31(3):288–305.
123. Lichtenbeld HC, Yuan F, Michel CC, et al. Perfusion of single tumor microvessels: application to vascular permeability measurement. Microcirculation 1996;3(4):349–357.
124. Yuan F, Leunig M, Berk DA, et al. Microvascular permeability of albumin, vascular surface area, and vascular volume measured in human adenocarcinoma LS174 T using dorsal chamber in SCID mice. Microvasc Res1993;45(3):269–289.
125. Yuan F, Dellian M, Fukumura D, et al. Vascular permeability in a human tumor xenograft: molecular size dependence and cutoff size. Cancer Res 1995;55(17):3752–3756.
126. Chauhan VP, Stylianopoulos T, Boucher Y, et al. Delivery of molecular and nanoscale medicine to tumors: transport barriers and strategies. Ann Rev Chem Biomol Eng 2011;2:281–298.
127. Tsuzuki Y, Mouta Carreira C, Bockhorn M, et al. Pancreas microenvironment promotes VEGF expression and tumor growth: novel window models for pancreatic tumor angiogenesis and microcirculation. Lab Invest2001;81(10):1439–1451.
128. Monsky WL, Fukumura D, Gohongi T, et al. Augmentation of transvascular transport of macromolecules and nanoparticles in tumors using vascular endothelial growth factor. Cancer Res1999;59(16):4129–4135.
129. Weis SM, Cheresh DA. Pathophysiological consequences of VEGF-induced vascular permeability. Nature 2005;437(7058):497–504.
130. Bockhorn M, Jain RK, Munn LL. Active versus passive mechanisms in metastasis: do cancer cells crawl into vessels, or are they pushed? Lancet Oncol 2007;8(5):444–448.
131. Duda DG, Duyverman AM, Kohno M, et al. Malignant cells facilitate lung metastasis by bringing their own soil. Proc Natl Acad Sci U S A 2010;107(50):21677–21682.
132. Swartz MA, Kristensen CA, Melder RJ, et al. Cells shed from tumours show reduced clonogenicity, resistance to apoptosis, and in vivo tumorigenicity. Br J Cancer 1999;81(5):756–759.
133. Jain RK, Koenig GC, Dellian M, et al. Leukocyte-endothelial adhesion and angiogenesis in tumors. Cancer Metastasis Rev 1996;15(2):195–204.
134. Eichler AF, Chung E, Kodack DP, et al. The biology of brain metastases-translation to new therapies. Nat Rev Clin Oncol 2011;8(6):344–356.
135. Kienast Y, von Baumgarten L, Fuhrmann M, et al. Real-time imaging reveals the single steps of brain metastasis formation. Nat Med 2009;16(1):116–122.
136. Hiratsuka S, Goel S, Kamoun WS, et al. Endothelial focal adhesion kinase mediates cancer cell homing to discrete regions of the lungs via E-selectin up-regulation. Proc Natl Acad Sci U S A2011;108(9):3725–3730.
137. Melder RJ, Munn LL, Yamada S, et al. Selectin- and integrin-mediated T-lymphocyte rolling and arrest on TNF-a-activated endothelium: augmentation by erythrocytes. Biophys J 1995;69:2131–2138.
138. Melder RJ, Yuan J, Munn LL, et al. Erythrocytes enhance lymphocyte rolling and arrest in vivo. Microvasc Res 2000;59(2):316–322.
139. Migliorini C, Qian Y, Chen H, et al. Red blood cells augment leukocyte rolling in a virtual blood vessel. Biophys J 2002;83:1834–1841.
140. Munn LL, Melder RJ, Jain RK. Role of erythrocytes in leukocyte-endothelial interactions: mathematical model and experimental validation. Biophys J 1996;71:466–478.
141. Melder RJ, Kristensen CA, Munn LL, et al. Modulation of A-NK cell rigidity: in vitro characterization and in vivo implications for cell delivery. Biorheology 2001;38:151–159.
142. Fukumura D, Salehi HA, Witwer B, et al. Tumor necrosis factor alpha-induced leukocyte adhesion in normal and tumor vessels: effect of tumor type, transplantation site, and host strain. Cancer Res1995;55(21):4824–4829.
143. Yamada S, Mayadas T, Yuan F, et al. Rolling in P-selectin deficient mice is reduced but not eliminated in the dorsal skin. Blood 1995;86:3487–3492.
144. Melder RJ, Brownell AL, Shoup TM, et al. Imaging of activated natural killer cells in mice by positron emission tomography: preferential uptake in tumors. Cancer Res 1993;53:5867–5871.
145. Melder RJ, Jain RK. Reduction of rigidity in human activated natural killer cells by thioglycollate treatment. J Immunol Methods 1994;175:69–77.
146. Melder RJ, Jain RK. Kinetics of interleukin-2 induced changes in rigidity of human natural killer cells. Cell Biophys 1992;20(2–3):161–176.
147. Sasaki A, Jain RK, Maghazachi AA, et al. Low deformability of lymphokine-activated killer cells as a possible determinant of in vivo distribution. Cancer Res 1989;49:3742–3746.
148. Melder RJ, Salehi HA, Jain RK. Interaction of activated natural killer cells with normal and tumor vessels in cranial windows in mice. Microvasc Res 1995;50(1):35–44.
149. Sasaki A, Melder RJ, Whiteside TL, et al. Preferential localization of human adherent lymphokine-activated killer cells in tumor microcirculation. J Natl Cancer Inst 1991;83(6):433–437.
150. Munn L, Koenig GC, Jain RK, et al. Kinetics of adhesion molecule expression and spatial organization using targeted sampling fluorometry. BioTechniques 1995;19:622–631.
151. Munn LL, Melder RJ, Jain RK. Analysis of cell flux in the parallel plate flow chamber: implications for cell capture studies. Biophys J 1994;67(2):889–895.
152. Gamble JR, Khew-Goodall Y, Vadas MA. Transforming growth factor-beta inhibits E-selectin expression on human endothelial cells. J Immunol 1993;150(10):4494–4503.
153. Detmar M, Brown LF, Schon MP, et al. Increased microvascular density and enhanced leukocyte rolling and adhesion in the skin of VEGF transgenic mice. J Invest Dermatol 1998;111(1):1–6.
154. Hamzah J, Jugold M, Kiessling F, et al. Vascular normalization in Rgs5-deficient tumours promotes immune destruction. Nature 2008;453(7193):410–414.
155. Huang Y, Yuan J, Righi E, et al. Vascular normalizing doses of antiangiogenic treatment reprogram the immunosuppressive tumor microenvironment and enhance immunotherapy. Proc Natl Acad Sci USA2012;doi:10.1073/pnas.1215397109.
156. Kalluri R, Zeisberg M. Fibroblasts in cancer. Nat Rev Cancer 2006;6(5):392–401.
157. Qian BZ, Pollard JW. Macrophage diversity enhances tumor progression and metastasis. Cell 2010;141(1):39–51.
158. Dawson MR, Chae SS, Jain RK, et al. Direct evidence for lineage-dependent effects of bone marrow stromal cells on tumor progression. Am J Cancer Res 2011;1(2):144–154.
159. Hiratsuka S, Duda DG, Huang Y, et al. C-X-C receptor type 4 promotes metastasis by activating p38 mitogen-activated protein kinase in myeloid differentiation antigen (Gr-1)-positive cells. Proc Natl Acad Sci U S A2011;108(1):302–307.
160. Egeblad M, Nakasone ES, Werb Z. Tumors as organs: complex tissues that interface with the entire organism. Dev Cell 2010;18(6):884–901.
161. Orimo A, Weinberg RA. Stromal fibroblasts in cancer: a novel tumor-promoting cell type. Cell Cycle 2006;5(15):1597–1601.
162. de Visser KE, Eichten A, Coussens LM. Paradoxical roles of the immune system during cancer development. Nat Rev Cancer 2006;6(1):24–37.
163. Mazzieri R, Pucci F, Moi D, et al. Targeting the ANG2/TIE2 axis inhibits tumor growth and metastasis by impairing angiogenesis and disabling rebounds of proangiogenic myeloid cells. Cancer Cell2011;19(4):512–526.
164. Rolny C, Mazzone M, Tugues S, et al. HRG inhibits tumor growth and metastasis by inducing macrophage polarization and vessel normalization through downregulation of PlGF. Cancer Cell2011;19(1):31–44.
165. Jain RK, Padera TP. Prevention and treatment of lymphatic metastasis by antilymphangiogenic therapy. J Natl Cancer Inst 2002;94(11):785–787.
166. Padera TP, Kadambi A, di Tomaso E, et al. Lymphatic metastasis in the absence of functional intratumor lymphatics. Science 2002;296(5574):1883–1886.
167. Elenbaas B, Weinberg RA. Heterotypic signaling between epithelial tumor cells and fibroblasts in carcinoma formation. Exp Cell Res 2001;264(1):169–184.
168. Brown EB, McKee T, diTomaso E, et al. Dynamic imaging of collagen and its modulation in tumors in vivo using second-harmonic generation. Nat Med 2003;9(6):796–800.
169. Chary SR, Jain RK. Direct measurement of interstitial convection and diffusion of albumin in normal and neoplastic tissues by fluorescence photobleaching. Proc Natl Acad Sci U S A 1989;86(14):5385–5389.
170. Nugent LJ, Jain RK. Extravascular diffusion in normal and neoplastic tissues. Cancer Res 1984;44:238–244.
171. Pluen A, Boucher Y, Ramanujan S, et al. Role of tumor-host interactions in interstitial diffusion of macromolecules: cranial vs. subcutaneous tumors. Proc Natl Acad Sci U S A 2001;98(8):4628–4633.
172. Swabb EA, Wei J, Gullino PM. Diffusion and convection in normal and neoplastic tissues. Cancer Res 1974;34(10):2814–2822.
173. Boucher Y, Brekken C, Netti PA, et al. Intratumoral infusion of fluid: estimation of hydraulic conductivity and implications for the delivery of therapeutic agents. Br J Cancer 1998;78:1442–1448.
174. Znati CA, Rosenstein M, Mckee TD, et al. Irradiation reduces interstitial fluid transport and increases the collagen content in tumors. Clin Cancer Res 2003;9:5508–5513.
175. Berk DA, Yuan F, Leunig M, Jain RK. Direct in vivo measurement of targeted binding in a human tumor xenograft. Proc Natl Acad Sci U S A 1997;94:1785–1790.
176. Ramanujan S, Pluen A, McKee TD, et al. Diffusion and convection in collagen gels: implications for transport in the tumor interstitium. Biophys J 2002;83(3):1650–1660.
177. McKee TD, Grandi P, Mok W, et al. Degradation of fibrillar collagen in a human melanoma xenograft improves the efficacy of an oncolytic herpes simplex virus vector. Cancer Res 2006;66(5):2509–2513.
178. 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 U S A2011;108(7):2909–2914.
179. Baxter LT, Jain RK. Transport of fluid and macromolecules in tumors. IV. A microscopic model of the perivascular distribution. Microvasc Res 1991;41(2):252–272.
180. Juweid M, Neumann R, Paik C, et al. Micropharmacology of monoclonal antibodies in solid tumors: direct experimental evidence for a binding site barrier. Cancer Res 1992;52(19):5144–5153.
181. Olive KP, Jacobetz MA, Davidson CJ, et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 2009;324(5933):1457–1461.
182. Leu AJ, Berk DA, Lymboussaki A, et al. Absence of functional lymphatics within a murine sarcoma: a molecular and functional evaluation. Cancer Res 2000;60(16):4324–4327.
183. Mouta Carreira C, Nasser SM, di Tomaso E, et al. LYVE-1 is not restricted to the lymph vessels: expression in normal liver blood sinusoids and down-regulation in human liver cancer and cirrhosis. Cancer Res2001;61(22):8079–8084.
184. Isaka N, Padera TP, Hagendoorn J, et al. Peritumor lymphatics induced by vascular endothelial growth factor-C exhibit abnormal function. Cancer Res 2004;64(13):4400–4404.
185. Hoshida T, Isaka N, Hagendoorn J, et al. Imaging steps of lymphatic metastasis reveals that vascular endothelial growth factor-C increases metastasis by increasing delivery of cancer cells to lymph nodes: therapeutic implications. Cancer Res 2006;66(16):8065–8075.
186. Tammela T, Alitalo K. Lymphangiogenesis: molecular mechanisms and future promise. Cell 2010;140(4):460–476.
187. Padera TP, Boucher Y, Jain RK. Correspondence re: S. Maula et al., intratumoral lymphatics are essential for the metastatic spread and prognosis in squamous cell carcinoma of the head and neck. Cancer Res., 63: 1920-1926, 2003. Cancer Res 2003;63(23):8555–8556; author reply 8558.
188. Padera TP, Stoll BR, So PT, et al. Conventional and high-speed intravital multiphoton laser scanning microscopy of microvasculature, lymphatics, and leukocyte-endothelial interactions. Mol Imaging2002;1(1):9–15.
189. Wong SY, Haack H, Crowley D, et al. Tumor-secreted vascular endothelial growth factor-C is necessary for prostate cancer lymphangiogenesis, but lymphangiogenesis is unnecessary for lymph node metastasis. Cancer Res2005;65(21):9789–9798.
190. Norrmen C, Tammela T, Petrova TV, et al. Biological basis of therapeutic lymphangiogenesis. Circulation 2011;123(12):1335–1351.
191. Cursiefen C, Chen L, Borges LP, et al. VEGF-A stimulates lymphangiogenesis and hemangiogenesis in inflammatory neovascularization via macrophage recruitment. J Clin Invest 2004;113(7):1040–1050.
192. Nagy JA, Vasile E, Feng D, et al. Vascular permeability factor/vascular endothelial growth factor induces lymphangiogenesis as well as angiogenesis. J Exp Med 2002;196(11):1497–1506.
193. Lyden D, Hattori K, Dias S, et al. Impaired recruitment of bone-marrow-derived endothelial and hematopoietic precursor cells blocks tumor angiogenesis and growth. Nat Med 2001;7(11):1194–1201.
194. Religa P, Cao R, Bjorndahl M, et al. Presence of bone marrow-derived circulating progenitor endothelial cells in the newly formed lymphatic vessels. Blood 2005;106(13):4184–4190.
195. Padera TP, Kuo AH, Hoshida T, et al. Differential response of primary tumor versus lymphatic metastasis to VEGFR-2 and VEGFR-3 kinase inhibitors cediranib and vandetanib. Mol Cancer Ther2008;7(8):2272–2279.
196. Rutkowski JM, Swartz MA. A driving force for change: interstitial flow as a morphoregulator. Trends Cell Biol 2007;17(1):44–50.
197. Fukumura D, Jain RK. Tumor microenvironment abnormalities: causes, consequences, and strategies to normalize. J Cell Biochem 2007;101(4):937–949.
198. Hagendoorn J, Padera TP, Kashiwagi S, et al. Endothelial nitric oxide synthase regulates microlymphatic flow via collecting lymphatics. Circ Res 2004;95(2):204–209.
199. Jeltsch M, Kaipainen A, Joukov V, et al. Hyperplasia of lymphatic vessels in VEGF-C transgenic mice. Science 1997;276(5317):1423–1425.
200. Berk DA, Swartz MA, Leu AJ, et al. Transport in lymphatic capillaries. II. Microscopic velocity measurement with fluorescence photobleaching. Am J Physiol 1996;270(1 Pt 2):H330–H337.
201. Leu AJ, Berk DA, Yuan F, et al. Flow velocity in the superficial lymphatic network of the mouse tail. Am J Physiol 1994;267(4 Pt 2):H1507–H1513.
202. Swartz MA, Berk DA, Jain RK. Transport in lymphatic capillaries. I. Macroscopic measurements using residence time distribution theory. Am J Physiol 1996;270(1 Pt 2):H324–H329.
203. Vakoc BJ, Lanning RM, Tyrrell JA, et al. Three-dimensional microscopy of the tumor microenvironment in vivo using optical frequency domain imaging. Nat Med 2009;15(10):1219–1223.
204. Boucher Y, Baxter LT, Jain RK. Interstitial pressure gradients in tissue-isolated and subcutaneous tumors: implications for therapy. Cancer Res 1990;50(15):4478–4484.
205. Boucher Y, Jain RK. Microvascular pressure is the principal driving force for interstitial hypertension in solid tumors: Implications for vascular collapse. Cancer Res 1992;52:5110–5114.
206. Boucher Y, Kirkwood JM, Opacic D, et al. Interstitial hypertension in superficial metastatic melanomas in humans. Cancer Res 1991;51(24):6691–6694.
207. Boucher Y, Lee I, Jain RK. Lack of general correlation between interstitial fluid pressure and oxygen partial pressure in solid tumors. Microvasc Res 1995;50(2):175–182.
208. Boucher Y, Salehi H, Witwer B, et al. Interstitial fluid pressure in intracranial tumours in patients and in rodents. Br J Cancer 1997;75(6):829–836.
209. Curti BD, Urba WJ, Alvord WG, et al. Interstitial pressure of subcutaneous nodules in melanoma and lymphoma patients: changes during treatment. Cancer Res 1993;53(10 Suppl):2204–2207.
210. Gutmann R, Leunig M, Feyh J, et al. Interstitial hypertension in head and neck tumors in patients: correlation with tumor size. Cancer Res 1992;52(7):1993–1995.
211. Less JR, Posner MC, Boucher Y, et al. Interstitial hypertension in human breast and colorectal tumors. Cancer Res 1992;52(22):6371–6374.
212. Milosevic M, Fyles A, Hedley D, et al. Interstitial fluid pressure predicts survival in patients with cervix cancer independent of clinical prognostic factors and tumor oxygen measurements. Cancer Res2001;61(17):6400–6405.
213. Nathanson SD, Nelson L. Interstitial fluid pressure in breast cancer, benign breast conditions, and breast parenchyma. Ann Surg Oncol Jul 1994;1(4):333–338.
214. 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 Res1991;51(24):6695–6698.
215. Znati CA, Rosenstein M, Boucher Y, et al. Effect of radiation on interstitial fluid pressure and oxygenation in a human colon carcinoma xenograft. Cancer Res 1996;56:964–968.
216. Boucher Y, Leunig M, Jain RK. Tumor angiogenesis and interstitial hypertension. Cancer Res 1996;56(18):4264–4266.
217. Lee CG, Heijn M, diTomasso E, et al. Anti-vascular endothelial growth factor treatment augments tumor radiation response under normoxic or hypoxic conditions. Cancer Res 2000;60:5565–5570.
218. DiResta GR, Lee J, Healey JH, et al. “Artificial lymphatic system”: a new approach to reduce interstitial hypertension and increase blood flow, pH and pO2 in solid tumors. Ann Biomed Eng2000;28(5):543–555.
219. Stohrer M, Boucher Y, Stangassinger M, et al. Oncotic pressure in solid tumors is elevated. Cancer Res 2000;60(15):4251–4255.
220. Netti PA, Baxter LT, Boucher Y, et al. Time-dependent behavior of interstitial fluid pressure in solid tumors: implications for drug delivery. Cancer Res 1995;55(22):5451–5458.
221. Netti PA, Hamberg LM, Babich JW, et al. Enhancement of fluid filtration across tumor vessels: implication for delivery of macromolecules. Proc Natl Acad Sci U S A 1999;96(6):3137–3142.
222. Zlotecki RA, Boucher Y, Lee I, et al. Effect of angiotensin II induced hypertension on tumor blood flow and interstitial fluid pressure. Cancer Res 1993;53(11):2466–2468.
223. Jain RK. Taming vessels to treat cancer. Sci Am 2008;298:56–63.
224. Krogh A, Harrop GA, Rehberg PB. Studies on the physiology of capillaries: III. The innervation of the blood vessels in the hind legs of the frog. J Physiol 1922;56(3-4):179–189.
225. Thomlinson RH, Gray LH. The histological structure of some human lung cancers and the possible implications for radiotherapy. Br J Cancer 1955;9(4):539–549.
226. Torres Filho IP, Leunig M, Yuan F, et al. Noninvasive measurement of microvascular and interstitial oxygen profiles in a human tumor in SCID mice. Proc Natl Acad Sci U S A 1994;91(6):2081–2085.
227. Pouyssegur J, Dayan F, Mazure NM. Hypoxia signalling in cancer and approaches to enforce tumour regression. Nature 2006;441(7092):437–443.
228. Helmlinger G, Sckell A, Dellian M, et al. Acid production in glycolysis-impaired tumors provides new insights into tumor metabolism. Clin Cancer Res 2002;8(4):1284–1291.
229. Bush RS. The significance of anemia in clinical radiation therapy. Int J Radiat Oncol Biol Phys 1986;12(11):2047–2050.
230. Fyles A, Milosevic M, Pintilie M, et al. Long-term performance of interstial fluid pressure and hypoxia as prognostic factors in cervix cancer. Radiother Oncol 2006;80(2):132–137.
231. Littlewood TJ. The impact of hemoglobin levels on treatment outcomes in patients with cancer. Semin Oncol 2001;28(2 Suppl 8):49–53.
232. Vansteenkiste J, Pirker R, Massuti B, et al. Double-blind, placebo-controlled, randomized phase III trial of darbepoetin alfa in lung cancer patients receiving chemotherapy. J Natl Cancer Inst2002;94(16):1211–1220.
233. Henke M, Laszig R, Rube C, et al. Erythropoietin to treat head and neck cancer patients with anaemia undergoing radiotherapy: randomised, double-blind, placebo-controlled trial. Lancet2003;362(9392):1255–1260.
234. Leyland-Jones B, Semiglazov V, Pawlicki M, et al. Maintaining normal hemoglobin levels with epoetin alfa in mainly nonanemic patients with metastatic breast cancer receiving first-line chemotherapy: a survival study. J Clin Oncol 2005;23(25):5960–5972.
235. Sundfor K, Lyng H, Kongsgard UL, et al. Polarographic measurement of pO2 in cervix carcinoma. Gynecol Oncol 1997;64(2):230–236.
236. Dzik S, Mincheff M, Puppo F. Apoptosis, transforming growth factor-beta, and the immunosuppressive effect of transfusion. Transfusion 2002;42(9):1221–1223.
237. Gharehbaghian A, Haque KM, Truman C, et al. Effect of autologous salvaged blood on postoperative natural killer cell precursor frequency. Lancet 2004;363(9414):1025–1030.
238. Santin AD, Bellone S, Palmieri M, et al. Effect of blood transfusion during radiotherapy on the immune function of patients with cancer of the uterine cervix: role of interleukin-10. Int J Radiat Oncol Biol Phys2002;54(5):1345–1355.
239. Gerweck LE, Vijayappa S, Kozin S. Tumor pH controls the in vivo efficacy of weak acid and base chemotherapeutics. Mol Cancer Ther 2006;5(5):1275–1279.
240. Kozin SV, Boucher Y, Hicklin DJ, et al. Vascular endothelial growth factor receptor-2-blocking antibody potentiates radiation-induced long-term control of human tumor xenografts. Cancer Res2001;61:39–44.
241. Jain RK, Forbes NS. Can engineered bacteria help control cancer? Proc Natl Acad Sci U S A 2001;98(26):14748–14750.
242. Rischin D, Peters LJ, O’Sullivan B, et al. Tirapazamine, cisplatin, and radiation versus cisplatin and radiation for advanced squamous cell carcinoma of the head and neck (TROG 02.02, HeadSTART): a phase III trial of the Trans-Tasman Radiation Oncology Group. J Clin Oncol 2010;28(18):2989–2995.
243. Semenza GL. Oxygen sensing, homeostasis, and disease. N Engl J Med 2011;365(6):537–547.
244. Mazzone M, Dettori D, Leite de Oliveira R, et al. Heterozygous deficiency of PHD2 restores tumor oxygenation and inhibits metastasis via endothelial normalization. Cell 2009;136(5):839–851.
245. Talks KL, Turley H, Gatter KC, et al. The expression and distribution of the hypoxia-inducible factors HIF-1alpha and HIF-2alpha in normal human tissues, cancers, and tumor-associated macrophages. Am J Pathol2000;157(2):411–421.
246. Carmeliet P, Dor Y, Herbert JM, et al. Role of HIF-1alpha in hypoxia-mediated apoptosis, cell proliferation and tumour angiogenesis. Nature 1998;394(6692):485–490.
247. Moeller BJ, Dreher MR, Rabbani ZN, et al. Pleiotropic effects of HIF-1 blockade on tumor radiosensitivity. Cancer Cell 2005;8(2):99–110.
248. Cramer T, Yamanishi Y, Clausen BE, et al. HIF-1alpha is essential for myeloid cell-mediated inflammation. Cell 2003;112(5):645–657.
249. Melillo G. Inhibiting hypoxia-inducible factor 1 for cancer therapy. Mol Cancer Res 2006;4(9):601–605.
250. Duda DG, Jain RK, Willett CG. Antiangiogenics: the potential role of integrating this novel treatment modality with chemoradiation for solid cancers. J Clin Oncol 2007;25(26):4033–4042.
251. McCormick F. New-age drug meets resistance. Nature 2001;412(6844):281–282.
252. Sharma S, Sharma MC, Sarkar C. Morphology of angiogenesis in human cancer: a conceptual overview, histoprognostic perspective and significance of neoangiogenesis. Histopathology 2005;46(5):481–489.
253. Varlotto J, Stevenson MA. Anemia, tumor hypoxemia, and the cancer patient. Int J Radiat Oncol Biol Phys 2005;63(1):25–36.
254. Nordsmark M, Loncaster J, Aquino-Parsons C, et al. Measurements of hypoxia using pimonidazole and polarographic oxygen-sensitive electrodes in human cervix carcinomas. Radiother Oncol2003;67(1):35–44.
255. Serganova I, Humm J, Ling C, et al. Tumor hypoxia imaging. Clin Cancer Res 2006;12(18):5260–5264.
256. Sorensen AG, Batchelor TT, Wen PY, et al. Response criteria for glioma. Nat Clin Pract Oncol 2008;5(11):634–644.
257. Gallez B, Baudelet C, Jordan BF. Assessment of tumor oxygenation by electron paramagnetic resonance: principles and applications. NMR Biomed 2004;17(5):240–262.
258. Rajendran JG, Schwartz DL, O’Sullivan J, et al. Tumor hypoxia imaging with [F-18] fluoromisonidazole positron emission tomography in head and neck cancer. Clin Cancer Res 2006;12(18):5435–5441.
259. Rischin D, Hicks RJ, Fisher R, et al. Prognostic significance of [18 F]-misonidazole positron emission tomography-detected tumor hypoxia in patients with advanced head and neck cancer randomly assigned to chemoradiation with or without tirapazamine: a substudy of Trans-Tasman Radiation Oncology Group Study 98.02. J Clin Oncol 2006;24(13):2098–2104.
260. Ansiaux R, Baudelet C, Jordan BF, et al. Thalidomide radiosensitizes tumors through early changes in the tumor microenvironment. Clin Cancer Res 2005;11(2 Pt 1):743–750.
261. Browder T, Butterfield CE, Kraling BM, et al. Antiangiogenic scheduling of chemotherapy improves efficacy against experimental drug-resistant cancer. Cancer Res 2000;60(7):1878–1886.
262. Segers J, Di Fazio V, Ansiaux R, et al. Potentiation of cyclophosphamide chemotherapy using the antiangiogenic drug thalidomide: Importance of optimal scheduling to exploit the normalization window of the tumor vasculature Cancer Lett 2006;244:129–135.
263. Willett CG, Duda DG, Ancukiewicz M, et al. A safety and survival analysis of neoadjuvant bevacizumab with standard chemoradiation in a phase I/II study compared with standard chemoradiation in locally advanced rectal cancer. Oncologist 2010;15(8):845–851.
264. Huber PE, Bischof M, Jenne J, et al. Trimodal cancer treatment: beneficial effects of combined antiangiogenesis, radiation, and chemotherapy. Cancer Res 2005;65(9):3643–3655.
265. Salnikov AV, Roswall P, Sundberg C, et al. Inhibition of TGF-beta modulates macrophages and vessel maturation in parallel to a lowering of interstitial fluid pressure in experimental carcinoma. Lab Invest 2005;85(4):512–521.
266. Wildiers H, Guetens G, De Boeck G, et al. Effect of antivascular endothelial growth factor treatment on the intratumoral uptake of CPT-11. Br J Cancer 2003;88(12):1979–1986.
267. Helmlinger G, Yuan F, Dellian M, et al. Interstitial pH and pO2 gradients in solid tumors in vivo: high-resolution measurements reveal a lack of correlation. Nat Med 1997;3(2):177–182.
268. Majmundar AJ, Wong WJ, Simon MC. Hypoxia-inducible factors and the response to hypoxic stress. Mol Cell 2010;40(2):294–309.
269. Fraisl P, Mazzone M, Schmidt T, et al. Regulation of angiogenesis by oxygen and metabolism. Dev Cell 2009;16(2):167–179.
270. Du R, Lu KV, Petritsch C, et al. HIF1alpha induces the recruitment of bone marrow-derived vascular modulatory cells to regulate tumor angiogenesis and invasion. Cancer Cell 2008;13(3):206–220.
271. Arany Z, Foo SY, Ma Y, et al. HIF-independent regulation of VEGF and angiogenesis by the transcriptional coactivator PGC-1alpha. Nature 2008; 451(7181):1008–1012.
272. Buysschaert I, Schmidt T, Roncal C, et al. Genetics, epigenetics and pharmaco-(epi)genomics in angiogenesis. J Cell Mol Med 2008;12(6B):2533–2551.
273. Ohtani K, Dimmeler S. Control of cardiovascular differentiation by microRNAs. Basic Res Cardiol 2010;106(1):5–11.
274. Nicoli S, Standley C, Walker P, et al. MicroRNA-mediated integration of haemodynamics and Vegf signalling during angiogenesis. Nature 2010;464(7292):1196–1200.