the biology of cancer (2007) - robert a. weinberg - ch. 13

60
Chapter 13 Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis Simple ideas are wrong. Complicated ideas are unattainable. Paul Valery, poet, 1942 A Simple and powerful conceptual paradigm pervades most of the previous discussions in this book: cancer is a disease of cells, and the phenotypes of cancer cells can be understood by examining the genes and proteins within them. The origins of this idea are clear, being traceable directly back to bacterial and yeast genetics. These two research specialties thrived because they were well served by the postulate that cell genotype determines all aspects of cell phenotype. Indeed, virtually all the attributes of individual bacterial and yeast cells could be shown to derive directly from the genes that these microorgan- isms carry in their genomes. Applying this concept to metazoans and their tissues has had obvious advan- tages for biologists. Metazoan organisms are complex almost beyond measure, and this complexity has frequently prevented researchers from extracting sim- ple and irrefutable truths about organismic function. In response, many researchers, notably molecular biologists, cell biologists, and biochemists,

Upload: bcastle1

Post on 16-Apr-2015

108 views

Category:

Documents


13 download

TRANSCRIPT

Page 1: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13

Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

Simple ideas are wrong. Complicated ideas are unattainable. Paul Valery, poet, 1942

ASimple and powerful conceptual paradigm pervades most of the previous discussions in this book: cancer is a disease of cells, and the phenotypes of

cancer cells can be understood by examining the genes and proteins within them. The origins of this idea are clear, being traceable directly back to bacterial and yeast genetics. These two research specialties thrived because they were well served by the postulate that cell genotype determines all aspects of cell phenotype. Indeed, virtually all the attributes of individual bacterial and yeast cells could be shown to derive directly from the genes that these microorgan­isms carry in their genomes.

Applying this concept to metazoans and their tissues has had obvious advan­tages for biologists. Metazoan organisms are complex almost beyond measure, and this complexity has frequently prevented researchers from extracting sim­ple and irrefutable truths about organismic function. In response, many researchers, notably molecular biologists, cell biologists, and biochemists,

527

Page 2: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

embraced the credo of reductionist science: when working with complex sys­tems, the best way to arrive at solid and rigorous conclusions is to take apart these systems into simpler, more tractable components and study each sepa­rately. While the lessons learned may relate only to small parts of very large sys­tems, at least these lessons are solid and definitive and will not require substan­tial revision each time a new generation of researchers revisits these complex systems and their component parts.

Such reductionism has allowed many areas of cancer research to thrive. Witness the progress described in the early chapters of this textbook: when the first proto-oncogene was discovered three-quarters of the way through the twentieth century (Section 3.9), almost nothing was known about the genetics and molec­ular biology of cancer. By the end of the century, information was available in abundance about how cancers begin and progress to highly aggressive, malig­nant states. Much of this avalanche of information came from taking the reduc­tionist paradigm to its limits-disassembling normal and neoplastic tissues into component cells and the cells into component molecules.

As mentioned above, the reductionist pact embraced by many in the cancer research community treated cancer as a cell-autonomous process: all the attrib­utes of cancer cells can be understood in terms of the genes that these cells carry. Actually, there was yet another notion embedded in their thinking: all of the traits of a tumor can be traced directly back to the behavior of individual cancer cells within the tumor mass.

By the end of the twentieth century, it became increasingly clear that many of the traits of tumors could not in fact be traced directly back to individual cancer cells and the genes they carry. The grand simplifications agreed upon a genera­tion earlier by cancer researchers had begun to lose their utility. Increasingly, evidence turned up that cancer is actually a disease of tissues, in particular, the complex tissues that we call tumors.

The reductionists' way of thinking had, all along, denied certain realities that clinical oncologists and pathologists confronted on a daily basis, namely, that carcinomas-which constitute more than 80% of the human cancer burden­derive from epithelial tissues of very complex microscopic structure. Histopathological characterizations of these epithelial tumors reveal that they are composed of a number of distinct cell types. In fact, even cursory examina­tions under the microscope indicate that most carcinomas are as complex his­tologically as the normal epithelial tissues from which they have arisen; exam­ples of this complexity can be seen in Figure 13.1. In some cases of commonly occurring carcinomas, such as those of the breast, colon, stomach, and pan­creas, the non-neoplastic cells, which constitute the tumor stroma, may account for as many as 90% of the cells within the tumor mass. An extreme ver­sion of this is presented by the cells of another type of solid tumor-Hodgkin's disease, a lymphoma in which non-neoplastic cells account for 99% of the cells in a tumor mass (Figure 13.2).

One's first instinct is to dismiss these non-neoplastic cells as distracting con tam ­inants that confound rather than enlighten attempts at understanding the biol­ogy of tumors. To do so now seems increasingly unwise. Recent research results make it clear that non-neoplastic cells, notably the stromal cells of carcinomas, are active, indeed essential collaborators of the neoplastic epithelial cells within tumor masses, having been recruited and then exploited by the cancer cells. The biology ofthese recruited cells now appears to be almost as important as that of the neoplastic cells in enabling growth of tumors.

This means that the disease of cancer is far more than cancer cells talking to themselves in endless monologues. We know, instead, that in most tumors, the neoplastic cells are in continuous communication with their non-neoplastic

528

Page 3: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Tumors are complex tissues

(A)

~ , ,: , '"

.~

neighbors. In this chapter, we focus our discussions specifically on carcinomas, in which the interactions of neoplastic and non-neoplastic cells within tumors are far better understood than in the many other types of cancer that afflict humans.

Figure 13.1 Stromal components of several commonly occurring carcinomas A variety of common carcinomas contain significant proportions of both epithelial cancer cells and recruited stromal cells, This is apparent in the carcinomas shown here, (A) A high-grade invasive ductal carcinoma of the breast, in which the membranes (arrows) of the epithelial cells are stained brown, while the nuclei of the stromal cells are light blue, (B) A colon carcinoma (right side) and normal colonic mucosa (N, left side), in which the tumor-associated stroma (TAS, right) is immunostained dark brown for PINCH (a protein associated with the cytoplasmic tails of integrins), while the nuclei of epithelial cancer cells lining ducts are light blue, (C) A lobular carcinoma in situ of the breast showing small clusters of cancer cells (light blue, arrow) surrounded by thin layers of stromal fibroblasts that have been immunostained for PINCH antigen (dark brown), (D) An adenocarcinoma of the stomach in which the cancer cells (purple, lower left) are adjacent to extensive stroma, which has been stained for collagen (blue) (A, from A Gupta, CG, Deshpande and S, Badve, Cancer 972341-2347,2003; Band C, from J Wang-Rodriguez, AD, Dreilinger, G,M, Alsharabi and A Rearden, Cancer 951387-1395, 2002; D, from S, Ohno, M, Tachibana, T. Fujii et ai., tnt. J. Cancer 97: 770-774, 2002)

Figure 13.2 Rare neoplastic cells in Hodgkin's lymphoma Normal, non­neoplastic cells may constitute the majority of living cells in many tumor masses, An extreme form of this is illustrated here by Hodgkin's disease, a lymphoma in which as many as 99% of the cells are recruited normal lymphocytes, which surround a rare neoplastic Reed-Sternberg cell (arrow) (From AT. Skarin, Atlas of Diagnostic Oncology, 3rd ed, Philadelphia: Elsevier Science Ltd" 2003,)

529

Page 4: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

13.1 Normal and neoplastic epithelial tissues are formed from interdependent cell types

The true complexity of the stroma of epithelial cancers becomes apparent only at high magnification under the microscope. In addition to the neoplastic epithelial (i.e., carcinoma) cells, a number of stromal cell types populate the tumor. These stromal cells include fibroblasts, myofibroblasts, endothelial cells, pericytes, smooth muscle cells, adipocytes, macrophages, lymphocytes, and mast cells (Figure 13.3; see also Section 13.3) . Because some of these cell types constitute only a small proportion of the stroma of certain tumors, their pres­ence becomes apparent only through use of immunostaining with antibodies that bind cell type-specific antigens.

The diverse stromal cell types within tumors are all members of several mes­enchymal ceUlineages that generate both connective tissue and various types of immune cells in the blood and immune tissues, and therefore are biologically very different from the epithelial cells whose transformation drives the growth of carcinomas. (In the case of non-epithelial tumors such as sarcomas, where the cancer cells are themselves of mesenchymal origin, the boundaries between the malignant cell compartment and the nonmalignant cells within the tumor mass are less easily defined.)

(F)

, " ,"

,. , ,

Figure 13.3 A variety of distinct cell types in the stroma of carcinomas Various antibodies ca n be used to immunostain specific mesenchymal cell types in the st roma of carcinomas. (A) CD4 antigen-positive T lymphocytes (brown) are apparent in the stroma of a non-small-cell lung ca rcinoma (NSCLO The stroma of squamous cell carcinomas of the oral cavity, pharynx, and larynx may contain (B) CD34 antigen-positive fibrocytes (brown), (C) CD117 antigen-positive mast cells (brown), and (D) <x-smooth muscle actin-positive myofibroblasts (brown). (E) The stroma of an in situ ductal carc inoma of the breast (DC IS) shows PINCH antigen-positive fibroblasts (dark brown; see Figure 131 B) arrayed

530 in a ring immediately around the carci noma (arrow), w hich is

surrounded, in turn, by adipocytes (light blue rims). (F) Monocytes, the precursors of macrophages, can be detected through their display of the CD 11 b+ antigen in this human colorectal adenocarcinoma. The epithelial cancer cells (light blue) form a duct outlined in the upper left. (A, from O. Wakabayashi, K. Yamazaki, S. Oizumi et al., Cancer Sci. 941003-1009, 2003; B, C, and 0, from P.J. Barth, T. Schenck zu Schweinsberg, A. Ramaswamy and R. Moll, Virchows Arch. 444:231-234, 2004; E, from J. Wang­Rodriguez, A.D. Dreilinger, G.M. Alsharabi and A. Rearden, Cancer 951387-1395, 2002; F, from E. Barbera-Guillem, J.K. Nyhus, c.c. Wolford et ai, Cancer Res. 62:7042-7049, 2002)

Page 5: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Tumors are formed from interdependent cells

In the absence of other information, we might explain the presence of these var­ious stromal cell types within a carcinoma in two ways. They might represent the remnants of cells that resided in the stroma of a tissue before tumor devel­opment began. During the subsequent expansion of the tumor cell population, groups of cancer cells may have inserted themselves between these preexisting normal stromal cell layers, thereby generating the richly textured tissues that are seen when most carcinomas are examined microscopically.

The alternative explanation for the presence of these numerous stromal cells is more intriguing and is likely to be the correct one for the great majority of tumors. The rationale behind this second model depends on our current under­standing of how normal, architecturally complex tissues maintain their struc­ture and function. In such tissues, the proper proportions of the various compo­nent cells must depend on the continuous exchange of signals among them. These interactions involve multiple distinct cell types, each following its own particular differentiation program. Such communication between dissimilar cell types is termed heterotypic signaling and is used by each cell type to encourage or limit the proliferation of the other types of cells nearby.

Extending this thinking, we might speculate that many of the heterotypic interac­tions operating in normal tissues continue to play important roles in the biology of the tumors arising in these tissues. This would suggest that, like normal epithe­lial cells, carcinoma cells continue to control the populations of stromal cells near them, perhaps by recruiting the latter from adjacent normal tissues and then encouraging their proliferation. Operating in the other direction, stromal cells may also influence epithelial cell proliferation and survival in the tumor mass.

In normal tissues, these heterotypic signaling channels depend in large part on the exchange of (1) mitogenic growth factors, such as hepatocyte growth factor (HGF), transforming growth factor-a (TGF-a), and platelet-derived growth fac­tor (PDGF); (2) growth-inhibitory signals, such as transforming growth factor-~ (TGF-~); and (3) trophic factors, such as insulin-like growth factor-1 and -2 (IGF-1 and -2), which favor cell survival (Figure 13.4).

(A) (B)

Met

Figure 13.4 Heterotypic ligand­receptor signaling (A) In epithelial tissues, the synthesis of PDGF-A and its receptor (PDGF-Ra) is usually confined to distinct cell types. In the testis, as in many bona fide epithelial tissues, expression of PDGF is confined to the cells of the tubular epithelium (above), while expression of its receptor is confined to the surrounding mesenchymal cells (below). (B) The reciprocal expression of the Met receptor and its ligand, termed variously hepatocyte grovvth factor (HGF) or scatter factor (SF), is shown here in the embryonic mouse ileum (the lower section of the small intestine). In each fingerlike villus, seen in longitudinal section, HGF/SF mRNA, detected by in situ hybridization, is made by the mesenchymal cells in the core of the villus (above) while Met is expressed by the epithelial cells coating the surface of the villus (below). (A, from L. Gnessi, S. Basciani, S. Mariani et ai, I Cell BioI. 1491019-1026,2000; B, courtesy of S. Britsch and C. Birchmeier) 531

Page 6: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

During the initial formation of tumors, heterotypic interactions play an impor­tant role in driving many types of tumor promotion. Thus, stromal cells, such as macrophages, neutrophils, and lymphocytes, participate in an inflammatory response that involves the release ofTNF-cx and prostaglandins; these molecules then stimulate the proliferation of nearby epithelial cells (Section 11.16).

Many heterotypic interactions continue to operate after tumors are formed. To cite some examples, epithelial cells within a carcinoma often release PDGF, for which stromal cells-notably fibroblasts, myofibroblasts, and macrophages­possess receptors; the stromal cells reciprocate by releasing IGF-l (insulin-like growth factor-I), which benefits the growth and survival of the nearby cancer cells. Similarly, the neoplastic cells within melanomas release PDGF, which elic­its IGF-2 production from nearby stromal fibroblasts; this IGF-2 helps to main­tain the viability of the melanoma cells. Stromal cells in breast cancers release the factor CXCLl2 (a chemokine; see Section 13.5) and the HGF/SF growth fac­tor, which stimulate the proliferation and survival of nearby epithelial cancer cells. By regulating each other's numbers and positions, epithelial and stromal cells can ensure the optimal representation and localization of each cell type within normal and neoplastic tissues.

The stromal and epithelial cells in normal epithelial tissues collaborate in the construction of the specialized extracellular matrix (ECM) that lies between them, called variously the basement membrane or basal lamina (Figure 13.5; see also Figure 2.3). For example, the epithelial cells of the skin, the ker­atinocytes, express genes specifying many of the major protein components of the basement membrane, including type IV collagen and laminins; the stromal cells also contribute to its construction, doing so in ways that have yet to be documented in detail. Certain proteoglycans in the basement membrane, such as perlecan, provide it with increased hydration and with sites to which growth factors can be attached for long-term storage. Continuous tethering to the basement membrane, mediated largely by integrins and the hemidesmosomes that they construct (see Figure 13.5A), is essential for the survival of many kinds of epithelial cells, and loss of this tethering often provokes anoikis, the form of apoptosis resulting from loss of anchorage to a solid substrate (see, for exam­ple, Figure 9.21).

The endothelial cells, which assemble to form the linings of the walls of capil­laries and larger blood vessels (Figure 13.6A) as well as lymphatic ducts, repre­sent a vital component of the normal and neoplastic stroma. As discussed in greater detail later, their proliferation is encouraged by other cells in both the epithelium and stroma in order to guarantee access by all of these cells to an adequate blood supply. Once capillaries are assembled and become functional, they provide essential nutrients and oxygen to nearby cells. We were introduced to these interactions earlier, when we read that cells lacking adequate access to oxygen release angiogenic factors that stimulate the ingrowth of capillaries (Section 7.12).

While capillary formation is proceeding, the endothelial cells secrete growth factors that stimulate the proliferation of nearby nonendothelial cell types. Most important, endothelial cells release PDGF and HB-EGF (heparin -binding EGF), which enables them to attract the peri-endothelial cells called pericytes and the vascular smooth muscle cells that together create the outer cell layers of capillaries, sometimes called the mural cells to distinguish them from the luminal endothelial cells (Sidebar 13.1). Once in place, the pericytes (which closely resemble smooth muscle cells) reciprocate by releasing vascular endothelial grovvth factor (VEGF) and angiopoietin-l (Ang-l), which provide important survival signals to the endothelial cells that recruited them in the first place. In addition, these mural cells provide the endothelial tubes with structural stability and an ability to resist the forces exerted by the pressure of blood (Figure 13.6).

532

Page 7: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Tumors are formed from interdependent cells

(A) keratinocyte nuclei

collagen hemidesmosomes basement membrane fibers

(B)

Figure 13.5 The basement membrane (A) In epithelial tissues, the specialized extracellular matrix (ECM) termed the basement membrane (BM) separates the epithelial cells (above) from the stroma (below) As shown in this transmission electron micrograph of the skin of a newborn mouse, the epithelial cells of the skin (termed keratinocytes) are tethered to one side (above) of the BM, being anchored in part through structures termed hemidesmosomes, which are composed of clusters of integrins that bind BM proteins, notably laminin; the keratinocyte nuclei are labeled here. Below the basement membrane are the collagen fibers and portions of mesenchymal cells constituting the stroma of the skin, i.e., the dermis. (B) This schematic drawing indicates that the basement membrane is composed largely of four major ECM proteins, namely, laminin, collagen type IV, perlecan, and nidogen. This highly permeable molecu lar meshwork allows a variety of molecules to pass through in both directions. A specialized basement membrane, not shown here, underlies endothelial cells in capillaries. (A, courtesy of HA Pasolli and E. Fuchs; B, from B. Alberts et al., Molecular Biology of the Cell, 4th ed. New York : Garland Science, 2002, based on H. Colognato and P.O. Yurchenko, Dev. Dynamics 218:213-234, 2000.)

C;::::8;> nidogen

~ ~

perlecan

b ;c type IV collagen

integrin

The locations and large populations of stromal cells within carcinomas provide clear indication that as the epithelial components of these tumors expand, this growth is accompanied by coordinated proliferation of stromal cells. In fact, stromal cells are even found to be layered between carcinoma cells in most metastases-the secondary tumors that become established in anatomical sites

533

Page 8: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

.

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

(B)

(0) pericyte precursors endothelial ECM tube ~~ t .

wt POGF-B

pericyte precursors

ECM

~~ t

mutant POGF-B

534

Page 9: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Tumors are formed from interdependent cells

Figure 13.6 Pericytes, smooth muscle cells, and endothelial cells of the microvessels (A) Microvessels can be studied using at least three types of microscopy. Immunofluorescent microscopy (left) of the capillaries in a normal mouse trachea (using an antibody that recognizes the endothelial cell-specific CD31 antigen) reveals a well -organized network of vessels. At the level of scanning electron microscopy (middle), a capillary is seen to resemble a smooth-walled cylindrical tube w ith pericytes attached here and there to its surface (arrows). In the transmission micrograph (right), a section of a capillary reveals that its wall is constructed by the folded cytoplasm of an endothelial cell. (B) This immunofluorescence micrograph resolves the pericytes and endothelial ce ll s. Normally, the pericytes and smooth muscle cells (both red orange) coat the outside of the tubes of endothelial cells (green), forming normally structured venules and arterioles (left panel). In these vessels, coverage of the endothelial cells by pericytes and smooth muscle cells is often so complete that it is difficult to visualize the endothelial cells lying under the pericytes. In contrast, in capillaries (right panel), the pericytes are more sparsely disposed, but are nonetheless tightly attached to the endothelial ce lls forming the capillaries. (C) Normally, the pericytes (yellow green) are tightly attached to capillaries (red, upper panel). However, mice have been genetically altered to ma ke mutant PDGF-B molecules that cannot attach to the extracellular matrix and therefore diffuse away from the endothelial cells making this growth factor. As a consequence, the pericytes are no longer attracted to the endothelial cells and lack intimate contact w ith the capillary tubes (arrows, lower panels). (D) This close juxtaposition of pericytes w ith the capillaries depends on the localization of PDGF-B, w hich is secreted by endothelial cells (lighter green) . Normally (above), this PDGF-B (dark green) is trapped immediately adjacent to the endothelial cells in the extracellular matrix (ECM, brown), w here it serves to attract pericyte precursors (pink) that attach themselves (red) tightly to the capillary tubes formed by the endothelial cells. However (below), if PDGF-B is deprived of the amino acid sequences that normally tether it to the ECM (purple), it diffuses away from the capillary tubes, and recruited pericytes are scattered at some distance from the capillaries. (A, from T. Inai, M. Mancuso, H. Hashizume et al., Am. 1. Pathol. 165:35-52,2004; B, from S. Morikawa, P Baluk, T Kaidoh et al., Am. 1. Pathol. 160:985-1000, 2002; C. from A. Abramsson, P Lindblom, and C. Betsholtz, 1. Clin. Invest. 112: 1142-1151, 2003.)

far away from the initially formed, primary tumors. Consequently, even those cancer cells that are independent enough to leave the primary tumor and to found new tumors at distant sites usually find it necessary to recruit stromal cells and encourage their proliferation (Figure 13.7A), ostensibly to aid in the construction of these new tumor colonies. On occasion, metastatic cells wiU insinuate themselves into the existing stroma of the tissue in which they have landed (Figure 13.7B), once again demonstrating their need to secure stromal support.

In the extreme version of this scenario, we might imagine that all of the het­erotypic interactions needed to maintain normal tissue function continue to operate within carcinomas, being required for the neoplastic cells to thrive and multiply within these tumors. However, such a depiction cannot be literally true, since many ofthe acquired traits that we associate with cancer cells (Chapter 2),

Sidebar 13.1 Localization of growth factors is important - for proper heterotypic interactions Bighly localized con­centrations of PDGF are needed to ensure that the peri­cytes recruited to the outer layers of capillaries become directly apposed to the endothelial cells rather than dis­persed randomly some distance away (Figure 13.6C andD). Like many other growth factors, the PDGF-B molecule­the specific form of PDGF that operates here-contains a "retention motif," which ensures its binding to proteogly­cans in the extracellular matrix (ECM) . Consequently, soon after 'PDGFcB molecules are secreted by endothelia'l cells

-into the space around capillaries, these molecules are trapped in the nearby ECM' and therefore accumulate in

high concentrations immediately next to the endothelial cells. Once liberated from this tethering by proteases, the resulting high local concentrations of activated PDGF-B molecules ensure the recruitment of pericytes to locations directly adjacent to the endothelial cells. However, in mice expressing a mutant PDGF-B that lacks this retention motif, the secreted PDGF-B molecules diffuse away from endothelial cells, causing the recruitment of pericytes to the general area of the endothelial cells but not closely apposed to them. These endothelial cells therefore lack the structural support afforded by the pericytes, aswell as the VEGF supplied by thepericytes that is required.for long" term endothelial cell survivaL ..

535

Page 10: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

(B)

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

normal liver tumor stroma tumor epithelium breast cancer metastasis normal liver

(A)

Figure 13.7 Metastasis and dependence on stroma (A) This micrograph of a metastasis originating from a primary colon carcinoma reveals that the metastasis is as complex histologically as the primary tumors seen in Figure 13.1. Here, we see that the carcinoma cells that have formed a metastasis (light purple, right) have constructed the same ductal structures as are seen in primary adenocarcinomas of the colon (see, for example, Figure 13.1 B). In addition, the carcinoma cells have recruited substantial stroma (middle), w hich includes macrophages (dark brown). (B) Most breast cancer metastases to the liver (left) behave quite differently from colon carcinoma metastases to this organ, in that the mammary carcinoma cells infiltrate into normal liver tissue (right) and displace resident hepatocytes, thereby taking advantage of the existing stroma, including its vasculature . These metastatic cells appear to be as dependent on stromal support as the colorectal carcinoma cells of (A) but acquire it in a totally different way. (From F. Stessels, G. van den Eynden, I. van der Auwera et al., Brit. J. Cancer 90 1429-1436, 2004)

including a decreased dependence on mitogens, an increased resistance to apoptosis, and acquisition of anchorage independence, are certain to affect the interactions between epithelial and stromal compartments and to lessen their interdependence. So, we conclude that these acquired traits reduce the depend­ence of epithelial cancer cells on stroma but do not seem to eliminate it.

13.2 The cells forming cancer cell lines develop without heterotypic interactions and deviate from the behavior of cells within human tumors

The continued dependence of carcinoma cells on stromal support explains the enormous difficulties that researchers have experienced when attempting to adapt tumor cells to in vitro culture conditions. Their goal here has been to cre­ate cancer cell lines-populations of cancer cells that can be propagated in tis­sue culture indefinitely. To do so, cells are prepared directly from tumor biopsies and placed into culture, with the hope that colonies of vigorously growing can­cer cells will eventually emerge in culture dishes. In most cases, however, such cell colonies do not appear, foiling attempts to generate carcinoma cell lines. More often than not, the cells that do thrive in culture are of stromal origin­specifically the fibroblasts whose growth is favored by the platelet-derived growth factor (PDGF) that is present in the serum component of standard tissue culture medium.

It is clear that human carcinoma cells propagated under standard conditions of tissue culture are being forced to proliferate in an environment very different from the one experienced by their ancestors that resided in the tumors of can­cer patients. For example, the high concentration of serum present in most cul­ture media is growth-inhibitory for many types of normal and neoplastic epithe­lial cells, which rarely experience large concentrations of the serum-associated factors in vivo except acutely after wounding. Even more important, carcinoma cells are being selected for their ability to proliferate in vitro without intimate contact 'v\lith stroma, since the proper balance of epithelial and stromal cells present in tumors cannot be maintained in the tissue culture dish.

On rare occasion, vigorously proliferating colonies of carcinoma cell popula­tions do indeed emerge follOwing extended culture of tumor fragments in the Petri dish. Because they have been selected in vitro for their ability to grow

536

Page 11: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Tumors resemble wound-healing sites

autonomously, and therefore independently of stromal support, such cancer cells have evolved beyond the stage of tumor progression that is reached in vivo by the neoplastic cells of most human tumors. In fact, many of the successes in establishing human tumor cell lines have depended on culturing cells from the few tumors that had already progressed in vivo to a stage where they no longer depended on stroma for their survival and proliferation (Sidebar 25 . ).

Such human cancer cell lines-there are several dozen in common use-are standard reagents used in many types of cancer research. Cells from virtually all of these can be implanted into immunocompromised host mice, where they proliferate, often vigorously, and form tumors. These tumors are often termed xenografts, since they result from grafting the tissues of one species into a host animal of another. Mice of the Nude, NOD/SCID, and RAG1/2 strains, lacking functional immune systems, are used in these experiments, because they tolerate the growth of introduced, genetically foreign cells. The resulting tumor xenografts growing in these mice can then be tested for their responsiveness to anti-cancer drugs under development (Sidebar 13.2). The outcome of such experiments is not necessarily predictive of eventual clinical responses to these drugs.

13.3 Tumors resemble wounded tissues that do not heal

As argued above, heterotypic signaling governs much of the biology of carcino­mas, and experimental models of cancer that ignore this important process gen­erate tumors that are biologically very different from those found in cancer patients. We now proceed to examine in greater detail the nature of this signal­ing within tumors and the cell types that participate in it. Actually, the het­erotypic interactions operating within tumors are highly complex and involve the exchange of dozens of distinct molecular species that mediate cell-to-cell signaling between the various cell types that together form these growths. Importantly, many of these signals are not routinely exchanged by the cell types constituting normal, undamaged tissue and therefore seem, at first glance, to be unique to tumors.

This complexity raises some fundamental questions: How do cancer cells learn to release and respond to an array of diverse heterotypic signals? Are the com­plex Signaling programs and resulting biological responses invented anew, being cobbled together piece by piece, each time normal cells evolve progres­sively into cancer cells? Or do cancer cells exploit preexisting biological pro­grams that are normally used by tissues for other purposes?

One attractive solution to these questions has come from an insight gained in 1986. A researcher studying the histological appearance of both tumors and wounds noted the striking resemblance between many of the signaling processes involved in tumor progression and those that occur during wound healing. If the similarities between these two biological programs could indeed be docu­mented, this would explain much of the cleverness of tumor cells: they simply activate a complex, normal physiologic program-wound healing-that is already encoded in their genomes. By accessing and exploiting this preexisting biological program, cancer cells are spared the task of re-inventing it anew each time a tumor arises.

Wound healing has been studied most extensively in the context of the skin. Following the formation of a superficial wound to the skin and underlying tis­sues, blood platelets aggregate and release granules containing, among other factors, platelet-derived growth factor (PDGF) and transforming growth factor­~ (TGF-~; see Figure 5.3) . Wounding also causes release of vasoactive factors, which increase the permeability of blood vessels near the wound. This helps the wound site to acquire fibrinogen molecules from the blood plasma, which,

537

Page 12: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

Sidebar ' 13.2 The development of anti-cancer therapies has been imperfectly served by the use ofexist­ing human cancer cell lines Human cancer cell lines, when grown as tumor xenografts in immunocompro­mised mice, often respond to the anti­proliferative effects of both un targeted cytotoxic drugs (which affect a wide spectrum of normal and neoplastic cell types) and drugs that are designed to affect only cancer cells. However, the ability of these xenograft models to predict the responses of patients to these drugs is limited. A 2001 retro­spective study of 39 different anti-can­cer drugs showed a weak correlation between the responses of xenografted tumors to these drugs and patient responses observed in the oncology clinic. For example, candidate drugs that were effective in halting the growth of more than one-third of a disparate collection of mouse tumor xenografts had only a 50% likelihood of showing any therapeutic activity (including halting tumor growth) in some human tumor types treated in the clinic. And drugs that affected smaller proportions of the mouse xenograft models had essentially no meaningful activity in the clinic.

These disappointing outcomes are not surprising, in Light of the fact that the human tumor cell lines commonly used in such drug testing experiments bear little resemblance to cells in the

tumors frequently encountered in the oncology clinic. Thus, the tumors that these cell.lines form often look quite different histologically from those routinely encountered in a clinical pathology laboratory (Figure 13.8).

Another clifficulty arises from the facrthat cells of various human tumor cell iines are usually implanted in mice .subcutaneously (beneath the skin). While this location affords researchers the ability to readily monitor the growth of these tumors, it almost always corresponds to an ectopic site within the host, that is, an anatomical location quite different from the one in which the cancer cells initially arose. For example, pancreatic carcinoma cells may behave differently when growing under the skin than when they are implanted into the host animal's pancreas, their natural, orthotopic site, presumably because the stromal envi­ronments in these two locations are so different. Similarly, breast cancer cells may generate tumors having more his­tological resemblance to clinical breast tumors if these cells are implanted into the mammary stromal fat pad instead of other locations.

Research into cancer stem cells suggests yet another disparity between cancer cell lines and the neo­plastic cell populations within human tumors, this one not related to het­erotypic interactions: if commonly occurring tumors are organized in a

hierarchical fashion, involving cancer stem cells and transit-amplifying cells with Hmited self-renewal capacity, this organization. is unlikely to be reestab­lished by cancer cell lines that are propagated in vitro (see Section 11.6).

One experimental response to these difficulties involves attempts at introducing fragments of tumors directly into immunocompromised mice without intervening propagation in tissue culture. These implanted tumor grafts contain both epithelial and stromal cells, which may co-prolif­erate to form histologically complex tumors in host mice that resemble the original tumors from which they derive (see Figure 13.8). Unfortunately, how­ever, like the cancer cells in actual human tumors, the cells in these xenografts usually proliferate very slowly, and their propagation involves labor-intensive transplantation from one host animal to another. These properties preclude their routine use in the testing of anti-cancer drugs.

The inadequacies of currently available human tumor models and the resulting inability to accurately predict how clinically effective anti­cancer drugs will be (before they are actually used in humans) cost the pharmaceutical industry hundreds of millions of dollars annually and there­fore represent one of the major impediments to the development of new anti-cancer drugs.

when converted to fibrin , create the scaffolding of the blood clot (Figure 13.9). The resulting fibrin bundles, which become entwined around clumps of platelets, help to stanch further bleeding.

The PDGF released by the platelets attracts fibroblasts and stimulates their pro­liferation (Figure 13.10). Thereafter, the platelet-derived TGF-~ activates these fibroblasts and induces them to release a class of secreted proteases termed matrix metalloproteinases (MMPs; Table 13.1) . Unlike most secreted proteases, which have a serine in their catalytic clefts, MMPs carry zinc ions to aid in catal­ysis; indeed, these ions inspired their name. Activated fibroblasts also secrete mitogens, such as various fibroblast growth factors (FGFs) that can stimulate the proliferation of certain epithelial cells.

Once released, the MMPs begin to degrade specific components of the extracel­lular matrix (ECM; Table 13.1). This degradation has two major consequences. On the one hand, it allows for structural remodeling of the ECM, thereby carv­ing out space for new cells. On the other, it results in the release of a variety of growth factors that have been tethered in an inactive form to the proteoglycans of the ECM and now become solubilized and activated. Included among these

538

Page 13: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Tumors resemble wound-healing sites

PROSTATE TUMORS '· .COLON TUMORS . . ..

eng rafted cell line PC-3M eng rafted cell line Col0205

are basic fibroblast growth factor (bFGF), TGF-~1, PDGF, several EGF-related factors, and interferon-y (IFN-y) . (In addition, virtually every MMP has been found to act on a variety of non- ECM proteins in the extracellular space; among these substrates are the latent proenzyme forms of other proteases, which are converted into active enzymes following cleavage by MMPs.)

The growth factors released by platelets and those mobilized from the ECM then attract monocytes, macrophages, and another class of phagocytes, termed neu­trophils, which infiltrate the wound site. (Yet other types of immune cells, including eosinophils, mast cells, and lymphocytes, are also recruited to this site.) These recruited cells scavenge and remove foreign matter, bacteria, and tissue debris from the wound site; at the same time, they release and activate mitogenic factors, such as fibroblast growth factors (FGFs) and vascular endothelial growth factor (VEGF; see also Section 7.12). Such factors proceed to stimulate endothelial cells in the vicinity to multiply and to construct new cap­illaries-the process of angiogenesis (sometimes termed neoangiogenesis).

While all this is occurring, largely in the stromal region of a wound site, the epithelial cells around the edges of the wound have been undergoing their o\lvn

Figure..13.S Tumors and derived xenografted tumor cell lines The histology of a primary prostate carcinoma (left) and of a colon carcinoma (right) that were removed surgically from patients are shown in the top row. Below these (middle row, left, right) are the histologies arising from implantation of chunks of tumor at subcutaneous sites in immuno­compromised SOD mice; these chunks contain both epithelial and stromal tumor components. Three to six months after implantation, these eng rafted tumor samples, which have grown from a 2-mm to a 1-cm diameter, still retain the architecture of the primary tumors from which they derive. However, a prostate and a colon cancer cell line (bottom row, left, right), which have been propagated extensively in vitro as pure cancer cell populations, bear little histological resemblance to the tumors typically encountered in the oncology clinic and shown above. (Courtesy of B.L Hylander and E.A. Repasky.)

539

Page 14: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

Figure 13.9 Scanning EM of a blood clot This scanning electron micrograph, w hich has been colorized, shows that clots are composed of dense netw orks of fibrin fibers (light brown) that have trapped platelets (red) Prior to clotting, platelets undergo activation that leads to t he release of the granules seen in Figure 5.3. (CNRI/Photoresearchers; courtesy of J. Weisel, Philadelphia.)

EPITHELIUM

damage to epithelial cells vasculature TGF-~1' MMPs

from stroma

r~--,)ll,---.., I exposure of ) morph into

mesenchymal cells platelet plasma to tissue platelet (EMT)aggregation factor in degranulation

p",cchym, ,-Jj~ ! move into wound

site and cover Hb'iooAb'iO J PDGF TGF-p w ound

(""___________J vf~~~~:sr j clot

macrophage fibroblast recruitment recruitment

epithelium

formationLlf mYOfiLb''''l ~ , ~

VEGF SDF-11 woundmatrix FGFs meta Iloproteinases

(MMPs)

~ liberation and activation of

growth factors

! morph into

epithelial cells (MET)

! reconstruct

CXCL12 contraction Figure 13.10 Flowchart of wound healing These flowcharts indicate many of the major events occurring after anrecruitment

of endothelial epithelial tissue is wounded . Many of precursor cells the changes occur in the stroma of the

wounded tissue (left) in order to .1 angiogenesis reconstruct it and thereby provide a and cytokines -----------1-----------.·

(bFGF, TGF-~1, foundation for reconstructing the PDGF, EGFs, IFN-y) ----./ capillaries epithelial cell layer (right) .

540

Page 15: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Tumors resemble wound-healing sites

Table 13.1 Some matrix metalloproteinases and their extracellular matrix substrates

Name Alternative ECM substratesa

of MMP name of MMP

NlMP:1 collagenase-l various collagens, gelatin, entactiri, aggrecan, tenascin MMP-2 gelatinaseA ' elastin, fibronectin, various collagens, laminiri,

aggrecan, vitroneCtin MMP-3 stromelysin-1 . . proteoglycans, laminin, fibronectin, gelatin, various

collagens, fibrinogen, entactin, tenascin, vitronectin MMP-7 matrilysin same as MMP-3 MMP-9 gelatinase 8 same as MMP-2 MMP~11 stromelysin-3 inactive serpin

aln addition to ECM substrates, most of the MMPs also cleave other substrates present in the extracellular space. For example, MMP-3 has been reported to cleave pro-HB-EGF, pro-IL-1~, plasminogen, E-cadherin, IGFBP-3, a 1-anti-chymotrypsin , a 1-proteinase inhibitor, pro-MMPs-1, -3, -7, -8, -9, and -13, and pro-TNF-a.

Adapted from U. McCawley and L.M . Matrisian, Curr Opin. Cell 8iol. 13:534-540, 2001.

alterations. Their goal is to reconstruct the epithelial sheet that existed prior to wounding. To do so, epithelial cells reduce their adhesion to the ECM, especially to the basement membrane (Section 2.2 and Figure 2.3) that separates them from the stromal compartment (Figures 13.5 and 13.11). By severing these con­nections, the epithelial cells gain increased mobility.

The epithelial cells also sever their attachments to one another. These side-by­side associations are stabilized in part by adherens junctions (Figure 13.12), which are assembled as associations ofE-cadherin molecules that are displayed by adjacent epithelial cells and tether the cells' apposed plasma membranes to one another. Accordingly, E-cadherin expression is suppressed in the epithelial cells that are situated around the edges of a wound site and is often replaced by N-cadherin, another cell-cell adhesion molecule that is normally displayed by mesenchymal cells, notably fibroblasts. (These N-cadherin molecules, though related structurally to E-cadherin, do not reestablish adherens junctions between the epithelial cells, since they bind much more weakly to one another.)

While shifting their display of cell-surface cadherins, the epithelial cells at the edge of a wound undergo a major change in phenotype, which causes them to assume, at least superficially, a fibroblastic appearance (Figure 13.13). This pro­found shift is termed the epithelial-mesenchymal transition (EMT) and enables the epithelial cells to become motile and invasive. The traits acquired during the EMT allow these cells to move into the wound site, where they fill in the gap in the epithelium created by the wounding.

Stromal cells in the wound site help to trigger the EMT of nearby epithelial cells. The matrix metalloproteinases (MMPs) secreted by stromal cells liberate and activate latent growth factors, such as TGF-~l, that have been stored in the extracellular matrix. In ways that are poorly understood, these released factors seem to stimulate and reinforce expression of the EMT program in epithelial cells. Importantly, the EMT is only a temporary shift in cell phenotype. After the

Figure 13.11 Attachments of epithelial cells to the structures around them Epithelial cells are anchored via their basal surfaces to underlying basement membrane (yellow) and to adjacent epithelial cells via their lateral surfaces, which form several distinct types of connections (anchoring junctions, black) that physically tie the cytoskeletons of neighboring celis (green lines) to one another; these cytoskeleton attachments involve both actin filament and microtubule components. (From B. Alberts et ai, Molecular Biology of the Celi, 4th ed. New York: Garland Science, 2002.)

apical surface cytoskeletal filaments

anchoring junctions ,

basement membrane

541

Page 16: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

(A) actin filaments inside microvillus microvilli extending

LUMEN from apical surface

-ilIr-it1--l1l+- t i g ht j u nct io n

bundle of actin filaments

lateral plasma membranes of adjacent epithelial cells

basal surface

figure 13.12 Adherens junctions (A) Thi s drawing illustrates the adhesion belt that is formed by adherens junctions between the lateral surfaces of adjacent epithelial cells. E-cadherin molecules are responsible for forming these adherens junctions in many epithelial cell types. (B) In more detail, these cell-cell connections depend on oligomerization between cadherin molecules displayed by two adjacent cells. E-cadherin (light green) plays a dominant role in most epithelia in forming the resulting adherens junctions. (C) As visualized in this transmission electron micrograph, the adherens junctions (black arrows) between two closely apposed plasma membranes form between the lateral surfaces of adjacent epithelial ce lls, in this instance those creating the intestina l lining of the worm Caenorhabditis elegans. Virtually identical structu res are seen in mammalian cells. (D) This immunofluorescence micrograph illustrates the interactions between the lateral surfaces of neighboring keratinocytes. E-cadherin "zippers" (yellow) form adherens junctions between neighboring cells and are attached via their cytop lasmic domains and other intermediary proteins to the act in cytoskeletons of these cells (red) Nuclei are stained in blue. (A and B, from B. Alberts et al., Molecular Biology of the Cell, 4th ed. New York: Garland Science, 2002; C, courtesy of D. Hall; D, from M. Perez-Moreno, C. Jamora and E. Fuchs Cell 112 :535-548,2003)

migrating cells have moved into position and covered the wound site, they reconstruct the epithelium by reverting to an epithelial state via the program termed the mesenchymal-epithelial transition (MET). As a consequence, once wound healing is complete, the cells in the reconstituted epithelium show no trace of having passed transiently through a mesenchymal state,

If we compare these processes (Figure 13.14) with the interactions of epithelial cancer cells and their stromal neighbors, we find striking parallels between wound healing and tumorigenesis . One clear similarity between the two processes derives from the presence of clumps of fibrin in the tumor-associated stroma. In the case of tumors, this does not derive from traumatic damage to

(B) plasma

CELL 1 CELL 2membrane

actin cadherin anchor proteins filament dimers

(C)

(D)

542

Page 17: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Tumors resemble wound-healing sites

(A) (B)

+ MMP-3 for 3 days

(C) Oh 24 h 48 h

72h 96 h 8d

Figure 13.13 The epithelial-mesen­chymal transition (A) When epithelial cells in monolayer culture undergo an epithelial-mesenchymal transition (EMT), they shut dow n expression of many typical epithelial cell markers, such as cytokeratins (red) and E-cadherin, and induce expression of mesenchymal proteins, such as vimentin (green), fibronectin, and N-cadherin. In addition, they change their typically polygonal shape (top) to a fibrob lastic shape (bottom) and often acquire motil ity and invasiveness. In this case, the EMT was provoked by a 3-day-long exposure to matrix metalloproteinase-3 (MMP-3), which may initiate the EMT through its ability to degrade E-cadherin . (B) In some cell types in culture, individual cells may spontaneously shift from an epithelial phenotype, indicated by cytokeratin expression (red), to a mesenchymal phenotype, indicated by a-smooth muscle actin expression (green) . This mirrors a plasticity that is apparent in certain cells during early embryogenesis .

(C) A confluent monolayer of MCF1 OA cells, a line of nontransformed, immortalized human mammary epithelial cells (MECs), has been disturbed by removal of a patch of cells from the monolayer (removed from the left of each image). Initially, no vimentin is expressed by these cells. However, soon after the monolayer has been wounded, the epithelial cells at the edge of the wound undergo a partial EMT, express vimentin (characteristic of mesenchymal cells; red) and migrate into and fill the wound site. After 8 days, at which time the cell monolayer has been restored, these cells revert to a fully epithelial phenotype and shut down vimentin expression (indicating a mesenchymal-epithelial transition, an MET). (A, from M.D. Stern licht, A Lochter, c.J Sympson et al., Cell 98:137-146, 1999; B, from O.W Petersen, H.L. Nielsen, T Gudjonsson et al., Am. I Pathor 162:391-402,2003; C, from C. Gilles, M. Polette, J.M. Zahm et al., I Cell Sci. 1124615-4625,1999 )

543

Page 18: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

Figure 13.14 The program of wound healing This drawing depicts schematically the heterotypic signaling occurring after the skin, a typical epithelial tissue, is wounded. In the site of the wound, a fibrin clot that carries platelets has initially filled the wound site . Day 3 of the wound-healing process is sometimes termed its inflammatory phase, because of the involvement of a number of cell types that are often associated with inflammation, notably neutrophils and monocytes; they are involved in removing cell debris, the clot, and invading bacteria. In addition, macrophages, w hich derive from differentiation of monocytes, release TGF-~1 and TGF-a; these factors stimulate the proliferation of nearby epithelial cells from the epidermis and induce them to undergo an epithelial-mesenchymal transition (EMT) Tongues of these cells, now exhibiting mesenchymal behavior, are invading under the clot from both sides in order to cover the wound site. At the same time, TGF-~l and PDGF-A and -B, which have been released by the platelets during initial clot formation, are stimulating the proliferation of fibroblasts in the underlying dermal stroma and inducing them to convert into myofibroblasts (not shown); because of their contractility, myofibroblasts will pull the sides of the wound together. Myofibroblasts also release angiogenic factors, notably VEGFs and FGF-2 (fibroblast growth factor-2). which stimulate the formation of new blood vessels in the stroma underlying the wound site . Finally, after the wounded stroma is covered, the cells that had previously undergone an EMT revert, via a mesenchymal-epithelial transition, (MET) to an epithelial state, thereby reconstituting the epithelium (not shown). (From AJ . Singer and RAF. Clark, N. Eng/. ) Med 341738-746, 1999.)

platelet plug neutrophil macrophage epidermis

dermis

\ \

-

I fibrin clot

j

'1\ \:~;!;1 VEGF

PDGF

( I t,blood GF vessel c<IG'') 'I i~~~~~PDjGFJ

- macrophage . FGF-2 ~ 1

neutrophil '-~ LTGF-~l :J'\ ~

fibroblast 0 GF-2

fat

blood vessels. Instead, the capillaries within tumors are constitutively leaky, unlike those in normal tissues (Figure 13.1SA). Later, we will discuss the causes of this leakiness. For the moment, suffice it to say that the permeability of the capillary walls and venules (small veins) allows fibrinogen molecules from the plasma to come in direct contact with cancer cells; this provokes, through a series of intermediate reactions, the conversion of fibrinogen to fibrin and the formation of large bundles of fibrin strands (Figures l3.1SB and C).

Many kinds of cancer cells, including those forming carcinomas of the breast, prostate, colon, and lung, continuously release significant levels of PDGF. This contrasts to the situation in wounds, in which PDGF is released in a brief burst by platelets as they form the initial blood clot. As is the case in wound healing, the targets of the PDGF produced by cancer cells are mesenchymal cells in the stroma that display PDGF receptors, including smooth muscle cells, fibroblasts, and macrophages. PDGF functions here both as an attractant and as a mitogen for these stromal cells and appears to be the most important signaling molecule used by carcinoma cells to recruit and stimulate the proliferation of stromal cells. In breast cancers, for example, the level of PDGF expression generally increases with increased tumor progression, which may explain the high degree of stromalization of many advanced tumors.

The PDGF released by carcinoma cells initially succeeds in recruiting fibroblasts into the fibrin matrix. At the same time, the fibrin matrix (see Figure l3.1SB and C) provides an important scaffolding that helps these recruited mesenchymal cells to attach, via integrins, and migrate. The invading stromal cells remodel this "provisional matrix" by degrading many of the initially formed fibrin mole­cules and replacing them with a more permanent matrix that is assembled from the collagen secreted by the fibroblasts. This closely parallels the sequence of steps occurring during wound healing.

544

Page 19: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Tumors resemble wound-healing sites

(A)

no tumor tumor

(8) tumor stroma

fibrin bundle fibroblasts cancer cells

(C)

One ofthe tasks of the stromal cells in wound healing involves the physical con­traction of the wound site in order to close wounds. This contraction is medi­ated by the specialized class of fibroblasts termed myofibroblasts, which express a-smooth muscle actin (a-SMA) and are capable of using the actin-myosin contractile system (related to the one operating in muscle celis) to generate the mechanical tension needed for wound closure (Figure 13.16A). Myofibroblasts are also present in sites of chronic wounding, that is, continu­ously inflamed tissues (Figure 13.16B). Provocatively, essentially identical myofibroblasts are prominent components of the stroma present in the major­ity of advanced carcinomas (Figure 13.16C-E).

Some of the tumor-associated myofibroblasts seem to arise from normal stromal fibroblasts. In fact, myofibroblasts can be produced in vitro simply by exposing normal fibroblasts to TGF-~l. This suggests that the TGF-~l released by many types of carcinoma cells, especially those that have progressed to higher levels of malignancy, is a major factor responsible for the formation of myofibroblasts in the tumor-associated stroma. In the context of wounds, the major source ofTGF­~1 is likely to be the platelets; as mentioned earlier, they release large amounts of this factor when they pa11icipate in clot formation. The origins of many stromal myofibroblasts and fibroblasts are unclear (Sidebar 13.3).

The tumor stroma formed by myofibroblasts differs substantially in appearance from the stroma present in normal epithelial tissues. This distinctive appear­ance has caused pathologists to label it a "reactive" or desmoplastic stroma. The latter term refers to the hardness of the tumor mass as a whole, which results from the deposition of extensive extracellular matrix (ECM) by the myofibrob­lasts. As the progression of a carcinoma advances to a higher, more aggressive grade, the proportion of stroma that is desmoplastic often increases in parallel (Figure 13.17).

Figure 13.15 Capillary leakiness and deposition of fibrin bundles in the tumor-associated stroma (A) Injection of red dextran dye into normal microvasculature reveals that the walls of associated capillaries have re latively low permeability, as indicated by the sharply delineated profiles of these vessels (left). However, when injected into tumor­associated vasculature, the dye leaks out of the capillaries and diffuses into the nearby parenchyma, yielding diffusely staining outlines of vessels (right). (B) The leakiness of tumor-associated microvasculature results in the continuous leaking of thrombin and fibrinogen molecules from the plasma into the parenchyma surrounding blood vessels. Tissue factor is a protein displayed on the surface of cancer cells and many other cell types. Thrombin from the plasma is activated upon contact with tissue factor and converts fibrinogen into fibrin. This results in the extensive network of fibrin bundles (dark red) seen here at the border between actively grow ing breast cancer cells (below) and the tumor stroma (above) (C) The fibrin bundles (above and to left of cluster of cancer cells) form an extracellular matrix to which migrating cells, such as fibroblasts (below cluster of cancer cells) as well as endothelial cells (not seen), can attach, using it as a substrate on w hich they can move forward. In many tumors, such as this guinea pig liver carcinoma of bile duct origin, the fibrin matrix may eventually be dissolved by the process of fibrinolysis and replaced by a collagenous matrix. (A, courtesy of R. Muschel; B, from e.G. Colpaert, P.B. Vermeulen, P. van Beest et al., Histopathology 42530-540, 2003; C, from H.E Dvorak, Am. I Pathol 1621747-1757,2003 )

545

Page 20: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

The myofibroblasts construct the desmoplastic stroma through the secretion of large amounts of collagen types I and III, fibronectin, proteoglycans, and gly­cosaminoglycans (GAGs), which together give this stroma its characteristic appearance at the microscopic level. In addition, these cells secrete urokinase plasminogen activator (uPA, a protease) and a series of matrix metallopro­teinases (MMPs), which help to mobilize growth factors that were previously sequestered in the ECM. As the desmoplastic stroma matures, many of the ini­tially present stromal cells disappear, being replaced progressively by the dense,

(C)(A)

(D) (E)

Figure 13.16 Myofibroblasts Myofibroblasts arise in sites of wound healing and in the tumor-associated stroma through the transdifferentiation of stromal fibroblasts and through the recruitment of myofibroblast precursors, possibly fibrocytes, from the circulation. Their presence is revealed typically by their expression of a-smooth muscle actin (a-SMA), the antigen that is being stained here. (A) a-SMA-positive myofibroblasts are present in abundance 3 days after the skin of a mouse has suffered a wound (reddish brown). (B) Chronically inflamed tissues acquire a fibrotic stroma, such as the cirrhotic liver seen here stained with anti-a-SMA antibody, which revea ls the presence of numerous myofibroblasts (brown) (C) A sedion of a hepatocellular carcinoma stained w ith anti-a-SMA antibody (brown). This revea ls the clear resemblance of the stroma of a chronically inflamed tissue to the stroma of a carcinoma arising in that tissue. (D) a-SMA (green ) is

expressed by the numerous smooth muscle cells present in the stroma of the normal human prostate gland. Unlike myofibroblasts, smooth muscle cells are know n to lack significant vimentin staining (red). The few areas of overlapping expression of these two proteins are indicated in yellow; these represent the occasional pericytes surrounding a blood vessel. The nuclei of epithelial cells lining the ducts are in blue. (E) In prostate carcinomas, in contrast, the stroma is filled with myofibroblasts that express both vimentin (red) and a-SMA (green). The overlap between these two is indicated by the yellow. Epithelial cell nuclei are indicated in blue. (A, from P. Martin, D. D'Souza, J. Martin et aI., Curr. BioI. 131122-1128,2003; Band C, from A. Desmouliere, A. Guyot and G. Gabbiani, Int. I Dev BioI. 48:509-517; D and E, from JA Tuxhorn, GE . Ayala, M.l Smith et al., Clin. Cancer Res. 82912-2923, 2002 .)

546

Page 21: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Tumors resemble wound-healing sites

Sidebar 13.3 Fibroblasts and myofibroblasts may be recruited from afar It remains unresolved precisely where most stromal fibroblasts . and myofibroblasts originate. During wound healing, fibroblasts may initially be recruited from-the stro·ma of adjacent normal tissues; myofibroblasts may then be produced through the transdifferentiation of some of these recruited fibroblasts. However, an intriguing alternative, still to be rigorously documented, is that many myofibroblasts and fibroblasts within wounds derive from recruited fibrocytes-Iess differentiated, collagen-produc­ing precursor cells that are present in the circulation.

Fibrocytes, which originate in the bone marrow, are known to be capable of homing to areas of tissue damage, where they settle and seem to differentiate into myofibroblasts and, quite possibly, fibroblasts, thereby contributing to the rapid reconstruction of stroma. This organization ensures that large numbers of cells can be mobilized rapidly from sources outside a wound site in order to reconstruct dam­aged stroma. The strong parallels between wound healing and tumorigenesis, described here, suggest that fibrocyte recruitment also plays a significant role in generating tumor-associated stroma.

acellular, collagenous ECM that is the hallmark of this type of tumor-associated stroma. The processes leading to the formation of the tumor-associated desmo­plastic stroma seem to be similar to those operating in sites of wound healing. However, in the tumor, these processes operate continuously over many months and years, rather than transiently over periods of several days, as happens dur­ing wound healing.

Another hint of parallels between wound healing and tumorigenesis comes from several studies indicating that agents known to inhibit tumor-associated angiogenesis also antagonize wound healing. Included among these antago­nists are certain soluble inhibitors of the growth factor receptors that play key roles in angiogenesis, as well as an ECM component, termed thrombospondin­1 (Tsp-1), which is a potent anti-angiogenic molecule. These common mecha­nisms shared by wound healing and by the epithelial-stromal interactions in tumors also have implications for clinical practice (Sidebar 13.4).

The term "carcinoma-associated fibroblasts" (CAFs) is sometimes used to describe the mixed populations of fibroblasts and myofibroblasts that are pres­ent in the stroma of epithelial tumors. Analyses of the gene expression patterns of CAPs underscore the strong similarities between these cells and the fibrob­lasts present in wound sites. In addition, these studies provide an indication of the role of these cells as key players in tumor progression.

These analyses initially determined the gene expression pattern of serum-stim­ulated fibroblasts propagated in vitro, that is, fibroblasts that had been starved of serum and then exposed to high concentrations of fresh serum. This exposure re-creates the environment of fibroblasts during the initial stages of wound healing (a time when the various serum factors released by activated platelets act on stromal fibroblasts at the wound site, converting them, at least tran­siently, into myofibroblasts) . A relatively small cohort of serum-induced and ­repressed genes was extracted from these data and used to represent the char­acteristic "signature" of serum-stimulated fibroblasts (Figure 13.18A) .

Figure 13.17 Micrograph of normal stroma and desmoplastic stroma The histologically complex stroma of normal tissue may eventually be replaced by the desmoplastic stroma of an adva nced carcinoma. Here we see normal human prostate tissue (left panel) stained with Masson 's trichrome stain, w hich reveals the extens ive smooth muscle cells in the stroma as pink; normal ducts are scattered throughout this stroma. In contrast, an advanced pros tate carcinoma (right panel) stained identically and viewed at the sa me magnification reveals the extensive desmoplastic stroma (blue purple), which is rich in extracellular matrix, notably collagen I. Islands of prostate ca rcinoma cells forming small ducts (p ink) are scattered throughout this desmoplastic stroma, which lacks significant numbers of viable cells such as myofibroblasts and fibroblasts. (From G. Ayala, JA Tuxhorn, 1M. Wheeler et al. , Clin. Cancer Res. 94792-4801 , 2003 )

547

Page 22: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

Sidebar 13.4 Breast cancer surgery may lead to the stimulation of tumor growth Epidemiologic studies of breast cancer patients undergoing surgery (i.e., partial or total mastec­tomy) for removal of their primary tumors have revealed a peak of recur­rence of breast cancers at the primary site as well as in distant anatomical sites occurring about 3 years after sur­gery. The timing of these relapses sug­gests that the surgery is responsible for stimulating their formation. Examination of the fluids draining from the wound sites in the breast cre­ated by the surgery has demonstrated the presence of potent mitogenic fac­·tors, which are associated with the wound-healing process. Among these

are mitogens for breast cancer cells, especially those cancer cells that over­express the HERZ/Neu protein. In addition, a burst of vascular endothe­lial growth factor (VEGF) synthesis occurs following surgery; as discussed later in this chapter, VEGF is a potent stimulant of tumor angiogenesis. It is therefore plausible that surgery stimu­lates the proliferation of residual micrometastases (small deposits of metastatic cells) that are not detected and removed by the surgeons when they excise primary tumors. Indeed, some have argued that the clinical relapses stimulated by surgery nullify much of the therapeutic benefit that should, by all rights, be achieved by the removal of primary tumors and

nearby lymph nodes. The importance of such surgery-stimulated tumor pro­gression remains a matter .of great contention.

Another clinical observation may eventually be found to bear on this dis­cussion: when Herceptin (a therilpeu­tic monoclonal antibody that blocks the action of the HERZ/Neu receptor; see Section 15.19) is applied postoper­atively in women who have borrie rela­tively small, low-grade tumors, the rate of clinical relapse is reduced by as much as 50%. This stunning therapeu­tic success may one day be traced to the ability of Herceptin to block stimu­lation of residual cancer cells by the growth factors elaborated during heal­ing of the surgical wound site.

The RNA expression patterns of a large group of human carcinomas were then analyzed. In each case, these analyses determined whether a tumor expressed the signature of serum-stimulated fibroblasts. (Since all the genes being ana­lyzed are associated specifically with fibroblasts, these analyses, by necessity, reflected the RNA expression patterns of the CAPs in each tumoL) Many carci­nomas were indeed found to express the signature of serum-stimulated fibrob­lasts. Significantly, the carcinomas that expressed this signature more intensely were associated with a grimmer clinical prognosis (Figure l3.18B and C). This correlation suggests that the activated, myofibroblast-rich stroma represents a force that can drive aggressive tumor progression.

13.4 Stromal cells are active contributors to tumorigenesis

Several biological experiments provide even more direct demonstrations of the profound influence that recruited stromal cells exert on epithelial cell tumorigenesis. In one study, previously non-tumorigenic, immortalized ker­atinocytes were forced to secrete PDGF at high levels (achieved through the introduction of a PDGF expression vector) . The released growth factor had no effect on the proliferation of these epithelial cells in vitro, because they do not display PDGF receptors on their surface. However, when these cells were implanted into host mice, they acquired the ability to form robustly growing tumors, clearly derived from the ability of the released PDGF to recruit and activate stromal cells. The stromal cells then reciprocated by driving the pro­liferation of the PDGF-secreting keratinocytes, eventually causing the latter to undergo malignant transformation.

In a complementary experiment, genetically engineered alterations of the mouse germ line enabled investigators to selectively inactivate the TGF-~ type II receptor in the stromal fibroblasts in a variety of tissues. As a consequence, these stromal cells were no longer susceptible to TGF-~-mediated growth inhi­bition, and stromal hyperplasia occurred in many of these tissues. In some of these, the hyperproliferating fibroblasts drove nearby epithelial cell layers to proliferate and, eventually, to develop into carcinomas (Figure 13.19). This demonstrates, once again, the powers of stromal cells to stimulate epithelial cell proliferation, doing so in ways that can lead to neoplastic transformation of the epithelial cells.

548

Page 23: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Stromal cells contribute to tumorigenesis

The important role of stromal fibroblasts in supporting tumor growth can be demonstrated by yet another type of experiment: transformed, weakly tumori­genic human mammary epithelial cells (MECs) were found to require more than two months to form a tumor after being introduced into immunocompromised host mice (Figure 13.20). However, if these cancer cells were mixed with human mammary stromal fibroblasts (from normal breast tissue) prior to injection into

(A) time after serum addition (B) breast carcinoma stage 1 ..c ~

~ ~ ..c ..c \- \- '- '­1o....1..t"\ 1.... ,-,-,-\-1......c..c..c..c..c..c core serum ..c~~..cLr\..c..c..c..c..cON<.DO"<t<.D 00 0'11"""" NM-=:;t I..CCO..-.-.-NNM cell cycle 'II""""

0.8 eSR

&' 0.6 :.0 .D'" e 0.4 eSR+ a.

0.2

0 0 5 10 15

metastasis-free survival (years)various genes

(e) J I~~~--c~';'cinoma

0.8

eSR­>­~ 0.6 :.0 .D '" e 0.4 a.

0.2

-2 -1 o 2 0 0 10 20 30 40 50 60

..............................1­-- extent of

0.25 0.5 2.0 4.0 induction or repression

survival (months)

- repressed induced -

Figure 13.18 Gene expression arrays of tumor-associated myofibroblasts and serum-activated fibroblasts The analysis of the spectrum of genes expressed in a cell population or a tissue is often called functional genomics. (A) In this gene expression analysis, the expression of cellular mRNAs was analyzed following the addition of fresh serum to previously quiescent, serum-starved human fibroblasts; this caused the entrance by these cells into the active cell cycle and the induction or repression of the expression of a large cohort of genes. Gene expression was measured at various times (above) after addition of serum. The cohort of genes analyzed (not named) is arrayed vertically from top to bottom Genes that were induced or repressed late after the addition of serum were classified as " cell cycle genes" (orange lines, right vertical bar), w hereas genes that w ere induced or repressed early and did not fluctuate w ith cell cycle phase were classified as "core serum response" (CSR) genes (blue lines, right vertical bar) . The horizontal bar (below) is a key indicating the degree of induction (red) or

repression (green) follow ing serum addition, this being indicated logarithmically above the bar and in absolute numbers below the bar. (Genes that are induced more than 4-fold above or repressed more than 4-fold below these levels are simply registered here as being induced 4-fold or repressed 4-fold ) (B ) The expression pattern of the CSR genes was analyzed in a series of tumors in women presenting with stage 1 (i.e., early-stage) breast cancer. As indicated here, those tumors expressing the CSR gene pattern (red line) showed a much greater probability for developing metastases in the years following initial treatment compared w ith those w hose tumors did not show this gene expression pattern (blue). (C) Similarly, those patients w hose lung adenocarcinomas (including all stages of tumor progression at the time of diagnosis) showed the CSR gene expression signature (red line) suffered dramatically higher mortality rates compared with those whose tumors did not show this gene expression signature (blue line) (From HY Chang, J.B. Sneddon, AA Alizadeh et al., PLoS Bioi. 2:E7, 2004.)

549

Page 24: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

Figure 13.19 Prostatic tumors from mice with genetically altered stromal fibroblasts The gene encoding the TGF·~ type II receptor has been inactivated selectively in the fibroblasts present in a variety of epithelial tissues of the mouse. (A) The prostates of male mice normally exhibit a thin layer of epithelial cells lining the lumina of the ducts and a relatively thin layer of stromal cells (asterisk) outside the epithelial cell layers. (B) As a consequence of TG F-~ type II receptor inactivation in the stromal fibroblasts, both cell layers became hyperplastic. The epithelial cells soon created tissue that closely resembled the prostatic intraepithelial neoplasia (PIN) commonly seen in humans (see Figure 1340). This suggests that the hyperplastic stroma released proliferative signals to the nearby epithelium. Indeed, production by the stromal cells of hepatocyte growth factor (HGF)-a potent epithelial cell mitogen­increased threefold after they lost their responsiveness to the inhibitory effects of TGF-I3. In the stomachs of these mice, in w hich the TGF-~ type II receptor was inactivated in the gastric stroma, the hyperplastic epithelia that formed became dysplastic and soon progressed to form carcinomas. Insets at higher magnification. (From N.A. Bhowmick, A Chytil, D. Plieth et al ., Science 303848-851, 2004)

Figure 13.20 Admixed fibroblasts and tumor growth When human mammary epithelial cells (HMECs) were transformed through the introduction of the SV40 early region, the hTERT gene, and a weakly expressed ras oncogene (Section 11.12), the resulting transformed HMECs (txHMECs) formed tumors, albeit with a long lag time after injection, and then only in about half of the mice into which they had been injected (orange curve). This slow development was accelerated a bit and tumor-forming efficiency was doubled when a preparation of extracellular matrix (Mat rigel) produced by fibroblasts was mixed vvith these cells prior to introduction into host mice (blue curve) However, when mammary stromal fibroblasts from a normal human breast were admixed to the transformed human MECs prior to injection, the cancer cells formed tumors rapidly and with 100% efficiency (red curve) . This illustrates that the recruitment of stromal fibroblasts is an important rate-limiting step in tumor formation . (From B. Elenbaas, L Spirio, F. Koerner et al.,

550 Genes Dev. 15:50-65, 2001.)

wild type TGF-~RII inactivated

hosts, the cells formed tumors in one-third of the time. These admixed fibrob­lasts clearly obviated the need of the MECs to spend time recruiting fibroblasts from host mice-a process that usually requires many weeks' time; in the absence of these fibroblasts, tumor growth could not take off.

This experiment did not address a related issue, specifically, whether the stro­mal cells of normal epithelial tissues can accelerate tumor formation as effec­tively as the stromal cells present in carcinomas. This issue has been addressed by comparing the actions of stromal fibroblasts extracted from a normal epithelial tissue with the carcinoma-associated fibroblasts (CAFs) prepared from carcinomas (in which myofibroblasts are abundant; see Figure 13.16). In one set of experiments, fibroblasts were purified from the stroma of normal human prostate glands, while the CAFs were prepared from the stroma of human prostate carcinomas. Each of these cell populations was then mixed with otherwise non-tumorigenic, SV40 large T antigen-immortalized human prostate epithelial cells and then implanted in immunocomprornised Nude mice. The results, summarized in Figure 13.21, showed dramatic differences in the growth of these mixed tissue grafts. In particular, the grafts containing CAFs plus immortalized prostate epithelial cells formed tumors that were as much as 500 times larger than those containing normal prostate fibroblasts plus immor­talized prostate epithelial cells. (When injected alone, the CAFs formed no tumors at all.)

This experiment demonstrated that these CAFs were functionally very different from the stromal fibroblasts present in normal prostatic tissue. Stated differ­ently, during the course of tumor progression, stromal cells become increasingly

700 txHMEC + fibroblasts 100%

600 txHMEC + Matrigel 100%

E 500

-5 txHMEC 52% OJ 400 E

0 ~

> 300 0 ~

E ~ +-' 200

100

0 0 10 20 30 40 50 60 70

time (days)

Page 25: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Macrophages contribute to tumorigenesis

adept at helping their epithelial neighbors to survive and proliferate. Similar observations have since been made with CAFs extracted from human breast cancers.

Still, these experiments do not reveal precisely how the myofibroblast-rich CAF populations accelerate tumor growth. Once established within the tumor-asso­ciated stroma, the myofibroblasts are likely to confer mUltiple benefits on nearby epithelial cancer cells. Possibly the most important of these benefits is angiogenesis. Myofibroblasts aid tumorigenesis through their ability to release stroma-derived factor-1 (SOF-l), also called CXCLl2; this factor (a chemokine) serves to recruit circulating endothelial precursor cells (EPCs) into the tumor stroma (Figure 13.22). The VEGF secreted by myofibroblasts helps to induce these recruits to differentiate into the endothelial cells that form the tumor neo­vasculature. Because angiogenesis is usually a rate-limiting step in tumor for­mation, the tumor-stimulating effects of admixed CAFs may be largely due to their ability to accelerate tumor angiogenesis.

In some tumors, the functionally altered stroma may be traced to a mechanism quite different from the one described above: stromal cells in advanced carcino­mas, having coexisted with epithelial cancer cells for many years, may change their genotype and acquire traits that genetically normal stromal cells cannot achieve. This suggests that stromal cells co-evolve with their neoplastic neigh­bors during these long periods of tumor development by altering their genomes in order to adapt to the physiologic stresses present within tumors.

For example, analyses of a large group of human breast cancers for mutations in the PTEN and TP53 tumor suppressor genes have shown the presence of somat­ically mutated alleles of these genes in stromal cells isolated from the tumors. These experiments exploited the procedure of laser capture microdissection (LCM; Figure 13.23) to isolate small patches of epithelial or stromal cells from these tumors. In some tumors, distinct mutant TP53 alleles were found in the epithelial and in the stromal compartments. In others, mutant alleles were found in one cell population but not the other. These experiments have opened the door to the possibility that the well-documented genetic evolution of neo­plastic epithelial cells (Chapter 11) is often accompanied by changes in the genomes of nearby stromal cells.

13.5 Macrophages represent important participants in activating the tumor-associated stroma

The active recruitment of macrophages into tumor masses would seem, at first glance, to be counterproductive for tumor formation, since macrophages are usually deployed by the immune system to scavenge and destroy infectious agents and abnormal cells, as we will explore in more detail in Chapter 15. However, increasing evidence indicates that these immune cells also play important roles in furthering tumor development.

In more detail, monocytes from the myeloid lineage in the bone marrow enter into the general circulation, from which they are recruited by cancer cells into a tumor; once ensconced there, the monocytes are induced to differentiate into macrophages (see Figure 13.14). This recruitment depends on attraction signals that are conveyed by chemotactic factors. By definition, these factors provide directional cues to motile cells rather than mitogenic stimulation. In the case of leukocytes (white blood cells such as monocytes), the relevant chemotactic fac­tors are often called chemokines, that is, chemotactic cytokines.

The chemokine known as monocyte chemotactic protein-1 (sometimes called macrophage chemoattractant protein; MCP-l) is expressed in significant quan­tities by a \Jvide range of neuroectodermal and epithelial cancer cell types. It

1000

Ci 5 +"'

~ 100 QJ

:s: o E ::J +"' 10 ........ ....

-:;-­

. ........

1<'7 '<.'" .. <J '" J 1<'7-t' ~f .. f~ (J;p 1<'7 '0 ,,0 ...:s'V f .,IJ'.;§> 1<'7

r? '0 ~ Q, f Q, r? 7f "­.t ~1V 0 r:; l)

~ .~"o " 1....",6,' ... ;- Q 0.rR ",'>! 7i ~ ~' "J:'9, <:'

1....'" c?

Figure 13.21 Effects of prostatic stromal cells on immortalized prostatic epithelial cells When normal human prostate epithelial cells (HPEs) that had been immortalized by SV40 T antigen (Tag-immortalized HPEs) were introduced into Nude mice, they formed growths of -10 mg after many weeks, not significantly larger than the initial inoculum (red dots). However, if these same cells were mixed w ith carcinoma­associated fibroblasts (CAFs) prepared from a human prostate carcinoma, tumors arose and the median w eight of these growths w as 10 times larger (Tag-immortalized HPEs + CAFs), blue dots). In contrast, w hen these T antigen-immortalized epithelial cells were mixed w ith stromal fibroblasts from a normal human prostate, the median weight of the resulting grow th was -7 mg, possibly even less than the starting inoculum (orange dots). And if CAFs were mixed with normal human prostate epithelial cells prior to inoculation, the median weight after many weeks was, once again, only -10 mg (purple dots) (From A.F. Olumi, G.D. Grossfeld, S.w. Hayward et ai, Cancer Res. 59:5002-5011, 1999)

551

Page 26: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

Figure 13.22 Recruitment of endothelial precursor cells by mammary CAFs (A) Tumors have been assembled by admixing stromal fibroblasts to cells of the human MCF7­ras breast cancer cell line. When normal human mammary stromal fibroblasts are admixed to the breast cancer cells (left panel), the resulting tumors show relatively small numbers of blood vessels (red) amid the tumor-associated stroma (blue) However, admixture of carcinoma-associated fibroblasts (CAFs; right panel) results in highly vascularized tumors with large vessels and associated erythrocytes (red). The resulting access to the circulation is likely to greatly facilitate tumor growth . (B) The ability of the CAFs, w hich are essentially myofibroblasts, to attract endothelial cells could be demonstrated by an in vitro experiment, in which green fluorescent protein (G FP)-Iabeled bone marrow cells expressing surface antigens characteristic of endothelial precursor cells (EPCs) were placed above a permeable membrane in a Boyden chamber. Either normal mammary stromal fibroblasts or CAFs from a breast cancer were placed on the bottom surface of the lower chamber. After 18 hours, the number of EPCs that had been recruited to the bottom of the lower chamber was gauged by fluorescence microscopy. (C) Analysis of the cells attached to the lower chamber indicated that the CAFs (right panel) are able to attract far more GFP-Iabeled EPCs than the normal mammary stromal fibroblasts (Jeft panel) This recruitment could be reduced by 60% by placing antiserum that neutralized the SDF­1/CXCL 12 chemokine released by the CAFs in these chambers, indicating its important role in mediating this recruitment. (From A. Orimo, P.B. Gupta, D.C. Sgroi et ai, Cell 1211-14, 2005)

__-1-----1,

(B)

admixed carcinoma-associated fibroblasts

18 hours

EPCs

permeable membrane

fibroblasts (C)

normal mammary fibroblasts carcinoma-associated fibroblasts

appears to be a critical signal for attracting monocytes to some tumors and inducing their differentiation into macrophages. In other tumors, vascular endothelial growth factor (VEGF) , colony-stimulating factor-l (CSF-l; often called M -CSF, macrophage-colony-stimulating factor), and the PDGF released by tumor cells also seem to help in this recruitment, while CSF-l aids in stimu­lating the monocyte-to-macrophage differentiation (Figure 13.24).

Once established in the tumor stroma, macrophages play important roles in stimulating angiogenesis. Thus, in some cancers, such as breast carcinomas, there is a direct correlation between the level of MCP-l produced, the number of macro phages present, and the level of angiogenesis induced by the various

552

Page 27: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Macrophages contribute to tumorigenesis

LCM +-

section on slide removed with cap

Figure 13.23 Laser capture microdissection The procedure of biochemically. As seen here, the section of a human colon laser capture microdissection (LCM) allows an investigator to use a carcinoma shown in the left panel contains both epithelial regions laser beam to microdissect tissue sect ions that have been affixed to (light blue) and stromal regions (brown) In this instance, the LCM a microscope slide. As part of the LCM procedure, a transparent procedure caused the epithelial areas to be left behind on the slide material, termed a "cap," is layered over a microscope slide to (middle panel) while the stromal regions were lifted up (nght which a tissue section has been affixed. A motor-driven laser beam panel). Both groups of cells could then be studied by various then irradiates chosen areas, which adhere to the cap; the cap can analytical procedures, including gene expression arrays. (Courtesy then be lifted w ith the adherent cells, w hich can then be analyzed of H. Dolznig .)

tumors; in other tumors , the density of infiltrating tumor-associated Figure 13.24 Recruitment of

macrophages is correlated with microvessel density (see, for example, Figure macrophages by CSF-1 In some tumors, colony-stimulating factor-l 13.2SA, B, and C; this density can be gauged by counting the number of capillar­(CSF-l) released by tumor cells serves asies per microscopic field in a tumor section.) A major role of macrophages in the key attractant for monocytes and driving tumor progression is suggested by numerous reports demonstrating a macrophages. In the transgenic mice

direct correlation between the presence of a high density of macrophages in studied here, mammary tumorigenesis tumor masses and a poor prognosis for cancer patients carrying such tumors. By was driven by a transgene, which was now, this correlation has been documented in gliomas and in carcinomas of the constructed from an MMTV (mouse breast, ovary, prostate, cervix, bladder, and lung. mammary tumor virus) transcriptiona l

promoter that drives expression of the Such evidence, however, is only correlative. More compelling evidence of the polyoma middle T (PyMT) oncogene.

causal role of macropbages, at least in tumor angiogenesis, is supported by (A) This tumor arose in a transgenic

experiments in which cancer cells are forced experimentally to express higher PyMT mouse that was heterozygous at the Csfl locus, since it carried one null levels of MCP-I. The expression of this chemokine allows the cancer cells to allele of the CSF-l-encoding gene, attract more macrophages, which proceed to secrete important angiogenic fac­termed Csf10p The presence oftors, notably VEGF and interleukin-8 (IL-8), resulting in marked increases in macrophages in the stroma of a tumor is

angiogenic activity and the formation of more extensive tumor-associated vas­ revealed by a monoclonal antibody that culature. In addition, when lung cancer cells are exposed in vitro to factors that detects macro phages (reddish brown).

(B) This tumor arose in a transgenic mouse that was genetically Csf7oplop and therefore lacked all CSF-1 activity. Consequently, the tumor cells in the

~

v mice also were unable to release this chemoattractant. In this mammary - tumor, the macrophages are barely" detectable. The absence of macrophages .., did not affect the growth of the primary tumor but strongly suppressed

...,t­

progression to a metastatic state (From EY Lin, AV Nguyen, R.G. Russell et ai, I Exp. Med 193727-740,2001)

.. . ;.,..-

553

Page 28: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

-----------------------------------------------------------

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

(A) (8)

(C) carcinoma cells

600

'0 •QJ 500 'i= x o § 400

$ • 'v; C QJ 300

"0 QJ

01

'" 200.s::. Q.

e u

100'" • •E r =0.743, P < .001

100 150 200 250

microvessel counts (200x field)

Figure 13.25 Macrophage proteins and angiogenesis in the tumor stroma (A) The(E) catalytic domain presence of tumor-associated macrophages (TAMs; red) can be demonstrated, as is seen here

C in this section of a human breast cancer, through the use of an antibody that specifically recognizes the HIF-2a protein displayed by these cells in the stroma of breast cancers. Like its cousin, HIF-l a (see Section 7.12), HIF-2a is involved in the up-regulation of angiogenic proteins in hypoxic cells. (B) In certain breast cancers (left panel), the cancer cells synthesize the potent angiogenic factor VEGF (vascular endothelial growth factor); as seen here, the islands of carcinoma cells stain positively (dark areas) with an anti-VEGF monoclonal antibody. However, in other breast tumors (righ t panel), VEG F production originates in isolated macrophages within the tumor-associated stroma (arrows) (C) In a series of human non-small-cell lung carcinoma (NSCLC) specim ens, each represented here by a point on the graph, the density of TAMs per microscope field has been plotted versus the density of microvessels (i.e., capillaries) per microscope field. Such correlations add further weight to the notion that TAMs playa key role in fostering tumor-associated angiogenesis. (D) Macrophages in the stroma of a human colorectal adenocarcinoma produce matrix metalloproteinase-9 (MMP-9; brown spots), a key enzyme in angiogenesis and invasiveness. MMP-9 can promote angiogenesis by liberating angiogenic factors from the extracellular matrix. (E) MMP-9 is synthesized as a pro-enzyme that becomes activated through cleavage by another protease, w hich results in release of a propeptide (yel/ow green, right) . The active MMP-9 is then able to cleave a diverse set of substrates in the intercellular space The zinc ion, which gives the MMPs their name, is shown as a black sphere; calcium ions are seen as gray spheres. (A, from R.D. Leek and AL. Harris, J. Mammary Gland BioI. Neoplasia 7: 177-189, 2002; B, from RD . Leek, N.C. Hunt et ai, J. Pathol. 190430-436, 2000; C, from J.J.W Chen, Y C. Lin, PL. Yao et ai, J. Gin. Oncol. 22:953-964,2005; D, from B.S. Nielsen, S. Timshel, L. Kjeldsen et al., Int. J. Cancer 65:57-62, 1996; E, from PA Elkins, YS. Ho, WW Smith et al., Acta Crystal. Series 0 58: 1182-1192, 2002)

554

Page 29: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Macrophages contribute to tumorigenesis

have been secreted by cultured macrophages, the cancer cells respond by pro­ducing IL-8 and a number of other proteins that promote angiogenesis and celi invasiveness.

Hypoxic areas within tumors attract macrophages, which appear to tolerate hypoxia quite well. Once established in hypoxic regions of tumors, these inflam­matory celis begin to secrete significant amounts ofVEGF (see Figure l3.25B and Section 7.l2), which reduces the hypoxia by bringing in endothelial celis, capil­laries, and thus oxygen-rich blood. The fact that hypoxic areas of tumors often remain poorly vascularized indicates that the recruited macrophages, on their own, are unable to fully cure local defects in angiogenesis. Thus, in rapidly grow­ing tumors, the macrophages and the vasculature that they induce cannot keep pace with the rate of tumor expansion, and large areas within a tumor mass even­tually become necrotic (Le., filled with dead and dying celis) as a consequence.

Like myofibroblasts (Section l3.3), macrophages are adept at secreting matrix metalloproteinases (MMPs; see Table l3.1). MMP-9, prominently involved in cancer progression, is produced by tumor-associated macrophages (TAMs; Figure 13.25D) and, once activated, proceeds to cleave a number of important protein substrates in the extracellular space. In certain invasive carcinomas, including those of the breast, bladder, and ovary, TAMs have proven to be the major source of this enzyme. It contributes to tumor progression by enhancing angiogenesis, by disrupting existing tissue structure and thereby carving out space for expanding tumor masses, and by liberating critical mitogens that have been immobilized through tethering to proteoglycans of the ECM. MMP­9 can also cleave IGFBPs (insulin-like growth factor-binding proteins; Sidebar 9.7) , which normally sequester IGF molecules in the extracellular space. This liberates the IGFs, notably IGF-1, which then provide survival signals to nearby cells, including cancer cells (Figure l3.26). In advanced breast cancers, tumor-

CARCINOMA j EMT-- invasiveness,CELLS I metastasisproliferation

! chemotactic

factors (MCP-1, CSF-1,

PDGF)

~~ circulating'----""'" tumor-,----....... tumor­monocytes associated associated

monocytes macrophages (TAMs)

)!L( -.

EGF mat angiogenic metallopr< ses factors

(e.g., M (VEGF, IL-8)

~ (

)! ~

cleave IGFBPs disrupt angiogenesis

~ liberate IGFs !

liberate "jmitogenic ogenic

factors rs

reate space for

new vessels

Figure 13.26 Contribution of macrophages to tumorigenesis Macrophages playa major role in releasing mitogenic factors for carcinoma cells as well as reorganizing the tumor stroma in order to facilitate angiogenesis and, in some tumors, carcinoma cell invasiveness.

555

Page 30: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

associated macrophages are also able to help the carcinoma cells directly, since they are the major source of the epidermal growth factor (EGF) that drives the proliferation of EGF-R-expressing carcinoma cells.

While there is abundant evidence implicating some macrophages as active col­laborators in tumor progression, it is also clear that another, morphologically distinct subset of macrophages, by acting as deputies of the immune system, can detect and kill cancer cells. Yet they clearly fail to do so in many types of tumors. It may be that some cancer cells acquire the ability to inactivate or blunt the tumoricidal (cancer-killing) actions of this second class of macrophages, while leaving intact the functions of the first type that are involved in aiding tumor progression. Alternatively, cancer cells may engage in evasive maneuvers that render them invisible to the macrophages that have been dispatched by the immune system to destroy them. We will return to the role of macrophages and other types of immune cells in blocking tumor development in Chapter 15.

13.6 Endothelial cells and the vessels that they form ensure tumors adequate access to the circulation

The most obvious stromal support required by tumors has already been cited repeatedly in this chapter: like normal tissues, tumors require access to the cir­culation in order to grow and survive. As early as the mid-1950s, pathologists noted that cancer cells grew preferentially around blood vessels. Those tumor cells that were located more than about 0.2 mm away from blood vessels were found to be nongrowing, while others even farther away were seen to be dying (Figure 13.27A).

We now realize that this threshold of approximately 0.2 mm represents the dis­tance that oxygen can effectively diffuse through living tissues. Cells located within this radius from a blood vessel can rely on diffusion to guarantee them oxygen; those situated further away suffer from moderate or severe hypoxia (Figure 13.27B and C). Tissues suffering from hypoxia are in danger of becoming necrotic, as discussed above and illustrated in Figures 13.27 and 13.28. p53-trig­gered apoptotic death also threatens hypoxic cells (Chapter 9), and the inactiva­tion of the p53 signaling system often enables cancer cells to survive beyond the small perimeter surrounding each capillary.

Hypoxia is only one price that is paid by cells lacking close proximity to the cir ­culatory system. Cells also require effective interactions with the vasculature in order to acquire nutrients and to shed metabolic waste products and carbon dioxide. The capillary networks threading their way through many normal tis­sues are arrayed so densely that virtually all cells in a tissue are no more than several cell diameters away from the nearest capillary (Figure 13.29). In some normal tissues with an especially high metabolic activity, most cells enjoy direct contact with at least one capillary. This intimate association means that their access to oxygen and critical nutrients is not dependent on the diffusion ofthese molecules over large distances and through densely packed cell layers.

These observations illustrate the central importance of the vasculature to the growth and survival of all types of tissue, normal and neoplastic. The process of developing this vasculature through angiogenesis can be observed during embryonic development, implantation of the placenta, wound healing, and cer­tain pathological conditions such as diabetic retinopathy, psoriasis, rheumatoid arthritis, and, of course, tumorigenesis.

During embryonic development, the overall size and architecture of an organ or tissue is genetically programmed, but the precise locations of its individual com­ponent cells are not. This applies as well to the vasculature: the genome deter­mines the overall layout of the major vessels and delegates to the individual cells 556

Page 31: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Endothelial cells enable neovascularization

\i thin the tissue the task of designing and assembling local capillary networks and slightly larger vessels. Heterotypic interactions operating over short dis­:ances determine the routing of individual capillaries constituting the microvas­culature. More specifically, capillaries appear to form wherever they are needed by the nonvascular (Le. , parenchymal) cells in a tissue.

A. variant of this plan operates during tumorigenesis. The overall architecture of rumors cannot be determined by the genome of an organism, since each tumor

\,­,.~' })

~

..'\~ ' ~ ; , . . . ~, ",

" rat prostate cancer

Figure 13.27 Hypoxia and necrosis of cells in poorly vascularized sections of tissues Various techniques have been used to demonstrate hypoxia and necrosis in regions of tissues surrounding capillaries. (A) This micrographic image reveals capillaries (green) as w ell as the degrees of oxygenation in the surrounding tumor parenchyma. Immunostaining using an antibody reactive with EF5, a molecule that localizes to hypoxic regions of tissues, reveals areas that are hypoxic (red) at some distance from the capillaries, w hile those cells that are well oxygenated are unstained and therefore appear dark brown. (B) These sections of a human melanoma (left panel) and rat prostate carcinoma (right panel) have been immunostained with an anti-CD31 antibody, which reveals capillaries (brown). Immediately around each capillary, a region of healthy cells is apparent. However, beyond a perimeter (dashed line) surrounding each capillary a region of necrosis (granula r area) is apparent. The necrotic region begins as close as 85 11m from the melanoma capillary (left) and 110 J.1m from the prostate carcinoma capillary (right). This necrosis reveals the limitations of diffusion in conveying oxygen and nutrients from capillaries to cells in the tumor parenchyma . (C) The dynamics of oxygenation on a larger scale are apparent here in this human

o 100 200 300 400

distance from vessel (J.1m)

_ pH - P02(mmHg)

squamous cell head and neck tumor. Blood vessels (blue spots) provide good oxygenation of the tumor, which appears black. However, in areas of poor vascularization and moderate hypoxia, a carbonic anhydrase enzyme is expressed (red), w hile in areas of extreme hYPoxia, the pimonidazole dye is detectable (green) . Overlap between these two markers appears orange. Areas of necrosis, located even further away from the tumor vasculature, are indicated by "N." (D) Use of indicator dyes and intra-vital microscopy makes it possible to measure pH and P02 (partial oxygen tension) in microenvironments located at various distances from a blood vessel. These measures explain the prime causes of necrosis in poorly vascularized areas: oxygen pressure fa lls to zero at a distance of more than 0.2 11m, w hile pH drops to levels that are incompatible w ith normal cellular metabolism, largely a consequence of lactic acid production occurring under anaerobic conditions. (A, courtesy of B.M . Fenton; B, left, courtesy of N. Almog and M .l. Folkman; B, right, from L. Hltaky, P Hahnfeldt and J. Folkman J. Nat!. Cancer Inst. 94:883-893, 2002; C. from l.H. Kaanders, K.l. W ijffels, HA Marres et ai, Cancer Res. 62:7066-7074, 2002; D, from G. Helmlinger, F. Yuan, M. Dellian and R.K. Jain, Nat. Med. 3177-182, 1997) 557

Page 32: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

Figure 13.28 Large-scale necrosis within a tumor The inability of large sectors of a tumor to gain access to the vasculature present in the stroma is demonstrated by this high -grade adenocarcinoma of the prostate. As seen most clearly in the growth in the center, a nest of adenocarcinoma cells (dark red) is surrounded by extensive stroma (light red) In the middle of the nest of adenocarcinoma cells is a large clump of necrotic tissue (light red) that has shrunk away from the viable adenocarcinoma cells that surround it; the latter thrive because they have better access to the stroma and associated vasculature. (From A.T Skarin, Atlas of Diagnostic Oncology, 3rd ed. Philadelphia : Elsevier Science Ltd., 2003)

Figure 13.29 Dense microvasculature in liver and muscle Because of high metabolic rates, certain tissues develop an extremely high density of capillaries, ensuring almost every cell direct contact with endothelial cells and the capillaries that they form. (A) Virtually every hepatocyte in the liver is adjacent to a space, termed a sinusoid, that is lined w ith endothelial cells and serves as a capillary. (B) Each muscle fiber, immunostained with an anti-Iaminin antibody, is a single syncytial cell, formed by the fusion of a number of myoblasts. Each muscle fiber lies adjacent to one or more capillaries (arrows, dark brown spots). (A, from Loyola University Medical Education Network; B, from Washington University of 51. Louis Neuromuscular Disease Center.)

necrosis

nests of adenocarcinoma

that arises is a novel invention whose design is not anticipated by the genome. This has implications for tumor angiogenesis as well: tumors cannot rely on some genetic blueprint to aid them in configuring their blood supply and must, instead, design the layout of their own vasculature, doing so step-by-step as they grow. In particular, when assembling their own blood supply, tumors depend totally on the localized heterotypic interactions between the cells of the vascu­lature (including endothelial cells, pericytes, and smooth muscle cells) and the nonvascular cells in tumors (including the neoplastic cells and the other cell types of the supporting stroma).

We have already learned of two ways by which tumors assemble vasculature. Myofibroblasts in the tumor-associated stroma can release chemotactic signals, such as SDF-lICXCLl2, which helps to recruit circulating endothelial precursor cells into the stroma (see Figure 13.22). This recruitment is likely to be aided by the release of VEGF, which also helps these cells to mature into functional endothelial cells.

As was first described in Section 7.12, production ofVEGF is governed by the availability of oxygen. Thus, many types of cells take stock of their own intracel­lular oxygen tension through the actions of the VHL protein and its partners. Under conditions of hypoxia, this complex of proteins allows functional HIF-1 transcription factor to accumulate, which in turn drives the expression of a

558

Page 33: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Endothelial cells enable neovascularization

Figure 13.30 Endothelial cells and the formation of capillaries This trans­mission electron micrograph reveals the cross section through a capillary in the midst of cardiac muscle and shows how the cytoplasms of two endothelial cells have become joined (arrows) to form the lumen of a capillary. The tight sealing of the plasma membranes of adjacent endothelial cells to one another results in capillary tubes whose walls are continuous and usually without gaps. The nucleus of one of the endothelial cells is also seen here. (From D.W Fawcett, J. Histochem. Cytochem. 13:75-91, 1965)

number of genes whose products encourage angiogenesis. Prominent among these is VEGF. Production of this key angiogenic factor has been associated with tumor cells, macro phages, and myofibroblasts, depending on the tumor type and its stage of progression. (In fact, there are two structurally related, function­ally similar proteins termed VEGF-A and VEGF-B. We will continue here to use the generic term VEGF to refer to both of these.)

Like many other growth factors, VEGF functions as a ligand of tyrosine kinase receptors-in this case, VEGF receptor-l (also termed FIt-I) and VEGF receptor­2 (known also as Flk-l/KDR). Both are displayed on the surfaces of endothelial cells. Similarly, basic fibroblast growth factor (bFGF), another important angio­genic factor, binds to its own cognate receptors displayed by endothelial cells. Once stimulated by these angiogenic factors, the endothelial cells proliferate and contort their cytoplasms to construct the cylindrical walls of capillaries (Figure 13.30). These capillaries also penetrate through existing tissue layers, moving toward the highest localized concentration of angiogenic factors.

Similar mechanisms appear to operate during the formation of lymphatic ves­sels (Sidebar 13.5). As we will see later in this chapter, lymph ducts are impor­tant regulators of fluid balance in the tumor stroma. In the next chapter, they assume great importance, because they provide avenues for cancer cells to escape primary tumors and disseminate to distant sites in the body.

The existence and powers of angiogenic factors were first revealed by implant­ing small chunks of tumor on the cornea or the ears of laboratory animals such as rabbits. Within a matter of days, dense networks of capillaries and larger ves­sels were seen to emerge from preexisting capillary beds and to converge on the implanted tumor chunks. Images like these (Figure 13.32A) have strongly influ­enced our thinking about the behavior of tumors and their vasculature, because they demonstrate so vividly that tumors actively recruit blood vessels into their midst.

The formation of blood capillaries is actually far more complicated than is sug­gested by these descriptions of angiogenesis. On the one hand, this complex morphogenetic process involves a number of other factors in addition to the VEGFs, which clearly playa major role in attracting blood vessels to tumors (Figure 13.32B). These other factors include several forms of TGF-~, basic

559

Page 34: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

Sidebar 13.5 Endothelial cells also construct lymph ducts The lymph

. ducts have t\,yo major functions in normal physiology. They drain fluid from the interstices between cells and empty this fluid into the venous circu­lation. In addition, they allow antigen ­presenting cells of the immune system to convey antigens from ' various tis­sues to the lymph nodes, where immune responses are often initiated (Chapter 15).

Interestingly, lymph ducts are assembled from endothelial cells orig­inating in the same embryoniC stem cell population that yields the endothelial cefls of capillaries and larger blood vessels. During embry­onic development, lymphatic vessels can often be observed to bud from developing capillaries before they sep­arate. and construct their own parallel network of interconnecting vessels (Figure 13.31). In addition, the factors

that stimulate lymphangiogenesis­vascular endothelial growth factors C . and D (VEGF-C and VEGF~D)~are homologous to VEGF-A and -B, which play a major role in stimulating the angiogenesis that creates the blood vasculature.

As. might be expected, the receptor for VEGF-C and VEGF-D displayed by lymphatic endothelial cells-VEGF receptor 3 (VEGF-R3)-is structurally related to the dominant VEGF recep­tor of blood capillaries, VEGF-R2. In addition, there is clear evidence that VEGF-D, which is mainly responsible for driving lyrnphangiogenesis, may also help stimulate angiogenesis by binding and activating the VEGF-R2. So these two systems-the blood and lymphatic networks-derive from common evolutionary roots, develop from common precursors in the embryo, and continue to interact with one another in complex ways within adult tissues.

Figure 13.31 Networks of lymphatic vessels and capillaries Most normal tissues are interlaced with networks of capillaries (green) and lymphatic vessels (red orange). As is apparent here, the diameters of lymphatic vessels are. far larger than those of capillaries. Unlike capillaries, however, the endothelial cells of lymphatic vessels are not slJpported by underlying mural cells---':'pericytesa'nd smooth muscle cell s. (Courtesy of 1 Tammela and K. Alitalo. l

fibroblast growth factors (bFGFs), interleukin-8 (IL-8), angiopOletm, angio­genin, and PDGF. Moreover, several distinct cell types in addition to endothelial cells contribute to the construction of capillaries and larger vessels. As men­tioned, the endothelial cells form the lumen of a capillary; these cells, in turn, are surrounded by the mural pericytes and vascular smooth muscle cells (see Figure 13.6).

The systematic covering of capillaries by pericytes seen in normal tissues (see 13.6B) can be contrasted with their chaotic dispersion near tumor-associated capillaries (Figure 13.33). Capillaries in tumors typically have diameters that are three times greater than their normal counterparts. In addition, the overall lay­out of blood vessels around and within tumor masses is quite chaotic (Figure 13.34). Often vessels stop abruptly in dead-end pouches or circle back and attach to themselves. At the submicroscopic level, it is also apparent that the capillaries in tumor masses are assembled haphazardly. This is because the plasma membranes of adjacent endothelial cells do not contact one another to form a seamless lining around the capillary lumen, but instead leave gaps,

560

Page 35: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Capillaries are complex structures

often several microns wide (Figure 13.35), which allow direct access of the blood plasma to the cells surrounding the capillary. The resulting leakage seems to be responsible for the deposition of fibrin in the tumor parenchyma described earlier.

Quantitative measures indicate that the walls of capillaries in tumors are about 10 times more permeable than those of normal capillaries. Some of this leakiness is attributable to the defective assembly of capillary walls noted here. But this, on its own, does not suffice to explain all of this leakiness. Instead, most of seems to be due to the deregulated production ofVEGF within tumors (Sidebar 13.6).

The precise reasons why capillaries and larger vessels within tumors are so hap­hazardly constructed are unclear. One possible factor may lie in the balance between two mutually antagonistic growth factors, angiopoietin-l and -2. While VEGF is responsible for initiating the growth of capillaries by attracting and stimulating endothelial cells, a mix of angiopoietin-l and -2 induces endothelial cells to recruit the pericytes and smooth muscle cells that enable newly formed capillaries to mature into well-constructed vessels containing appropriate pro­portions of these three cell types. An imbalance of these two antagonistic

(A)

Figure 13.32 Recruitment of capillaries by an implanted tumor bearing human adenocarcinoma cell xenografts. The w idespread (A) These images show the grow th of a small group of pink background in the untreated control mouse, which is indicative subcutaneously implanted human colorectal adenocarcinoma cells of numerous capillaries, and the number of major vessels entering over a period of 20 days. The growth of the tumor-associated into a tumor mass (top) are strongly decreased in the presence of vessels is observed through a window that has been inserted in the the drug (bottom). (A, from M . Leunig, F. Yuan, M.D. Menger et ai , skin of the mouse above the tumor. (B) Such vascularization can be Cancer Res. 52 :6553-6560, 1992; B, from S.R. Wedge, D.J. Ogilvie, suppressed by antagonists of the VEGF receptor. Seen here are the M . Dukes et ai, Cancer Res. 62:4645-4655, 2002) effects of ZD6474, an inhibitor of the VEGF receptor, in mice

561

Page 36: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

Figure 13.34 The chaotic organization of tumor-associated vasculature (A) This image, obtained by injecting a resin into blood vessels prior to tissue fixation, shows that the vasculature feeding a mass of tumor cells (black area, far right) is tortuous and poorly organized, in contrast to the well-organized vasculature in normal tissue (turquoise area, top left). (B) Techniques of imaging living tissues (intravital imaging) have made it possible to visualize the capillary beds of normal and neoplastic tissues. This technique reveals that the capillary networks in a normal tissue are well organized (left), w hile those in a tumor are chaotic (right). (A, courtesy of L. Heiser and R. Ackland, University of Louisville; B, from R.K. Jain, Nat. Med 9: 685-693, 2003)

Figure 13.33 Tumor-associated pericytes This fluorescent micrograph reveals the structure of a tumor-associated microvessel, in this case a vessel formed in a mouse by Lewis lung carcinoma (LLC) cells. The endothelial cells forming the lumen of this vessel (green) are only partially overlain by pericytes and smooth muscle cells (red orange). The loose attachment of pericytes to the capillaries can be contrasted to their tight attachment seen in Figure 13.6B. (From S. Morikawa, P Baluk, T Kaidoh et ai, Am. I Pathol. 160985-1000, 2002)

angiopoietins, together with the greatly elevated levels ofVEGF within tumors, is suspected to be responsible for much of the defective construction of the cap­illaries and larger vessels within neoplasms.

The leakiness of tumor-associated capillaries leads to the accumulation of sub­stantial amounts of fluid in the parenchymal spaces within a tumor. In normal tissues, these fluids are drained by the lymphatic vessels, which eventually empty their contents into the venous circulation (Sidebar 13.5). However, within solid tumors, the ongoing expansion of cancer cell populations exerts pressure on those few lymphatic vessels that do succeed in forming, causing their col­lapse (Figure 13.36). (Blood capillaries are more capable of resisting this pres­sure, because of their own significant internal hydrostatic pressure, which lym­phatic vessels lack.) The resulting defective lymphatic drainage within the cores of solid tumors further exacerbates the elevated accumulation of fluid caused by

(B)

normal tissue tumor 562

Page 37: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Progression depends on the angiogenic switch

capillary leakage, generating relatively high fluid pressure in the nonvascular parts of tumors. This pressure, in turn, greatly complicates the administration of anti-cancer therapeutic drugs (Sidebar 26. ).

13.7 Tripping the angiogenic switch is essential for tumor expanslOn

The descriptions of angiogenesis given above, as well as those in an earlier chap­ter (see Section 7.12) , would seem to suggest that the release of angiogenic fac­tors is virtually automatic: whenever groups of cells, induding cancer cells, suf­fer hypoxia, they release angiogenic factors and thereby provoke the growth of capillaries into their midst. This cures the hypoxia and results in an appropriate density of capillaries in the tissue harboring these cells.

In fact, the ability to attract blood vessels seems to be a trait that many tumor cell populations initially lack and must acquire as tumor progression proceeds. This idea was initially suggested by the observation, cited above, that in certain tumors, cancer cells thrive near capillaries, but those that are located further than 0.2 mm from capillaries stop growing and may enter apoptosis or become

Sidebar 13.6Vascular endothelial growth factor (VEGF) causes capillary per­meability Two independent lines of j'esearch led to the initial cloning of the gene encodingVEGF-A. One research project characterized a factor secreted by cancer cells that caused blood vessels to leak fluid and was therefore termed vascular permeability factor (VPF). The other work focused on a factor released by tumors that functioned as an attractant and mitogen for the endothelial cells that assemble to fOI'm the luminal walls of capillaries; this agent was called vas­cular endothelial grovvth factor (VEGF)-the name that prevails today. The two research efforts culminated in 1989 in the independent isolations of the VPF­and VEGF-encoding genes and the discovery that they are one and the same. Cancer cells, as well as macrophages and fibroblasts in the tumor-associated stroma, continuously release significant amounts ofVEGF, which are responsi­ble for much of the elevated permeability of the tumor-associated capillaries and venules.

The precise mechanisms responsible for VEGF-induced permeability remain unclear. VEGF may cause the plasma membranes of adjacent endothe­lial cells to separate slightly, generating gaps between them. In addition, it may stimulate endothelial cells to transport fluid and solutes through their cyto­plasm from the luminal to the abluminal surface.

Figure 13.35 Scanning EM of gaps in tumor vasculature These scanning electron micrographs reveal that the sheets of endothelia l cel ls, which form a continuous, uninterrupted surface around the walls of a normal capillary (not shown), fail to do so in the capillary within a tumor. Instead, the tumor­associated endothelial cells overlap one another and, as indicated here (arrows), show gaps of significant size between them . Th e box in the left panel is shown at higher magnification in the right panel. Such gaps permit the seepage of plasma fluids into the interstitial spaces between the cancer cells, contributing to high hydrostatic pressure in these spaces. (From H. Hashizume, P. Baluk, S. Morikawa et ai, Am. J. Pathol. 156: 1363-1380, 2000.)

563

Page 38: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

Figure 13.36 Absence of lymphatic vessels within solid tumors Analysis of a section of a hepatocellular carcinoma (HCC; liver cancer), using an antibody to specifically stain lymph ducts (dark red), reveals that these ducts are present in the normal tissue above the tumor margin (dotted line) but are absent within the tumor mass itself . This absence may be attributed (1) to the fact that these ducts were not generated initially during tumor formation and/or (2) to the collapse and degeneration of these ducts because of the high hydrostatic pressure within solid tumors. (From C. Mouta Carreira, S.M. Nasser, E. di Tomaso et aI., Cancer Res. 618079-8084, 2001.)

Figure 13.37 The Rip-Tag model of islet cell tumor progression Multi-step tumorigenesis in the Rip-Tag transgenic mouse model proceeds in distinct stages. A normal pancreatic islet (also known as an islet of Langerhans; left) carries a small number of capillaries to support the pcells. By 5-7 weeks of age, about ha If of these islets become hyperplastic, but the density of blood vessels is not increased. During a subsequent period from 7 to 12 weeks, about 10% of the hyperplastic islets become angiogenic, as indicated by the greatly increased density of capillaries in the surrounding, nearby exocrine pancreas. Finally, by 12-14 weeks of life, 2-4% of the initially formed hyperplastic islets have become invasive carcinomas that grow rapidly and invade the surrounding exocrine pancreas. The exocrine pancreas, which surrounds the islets, is not shown here. (From D. Hanahan and J Folkman, Cell 86353-364, 1996)

~. ... .,:

tUrr.lo:r " /

necrotic (Section 13.6). So, even though these cancer cells experience hypoxia, they lack the ability to induce the formation of nearby capillaries.

We know much about these dynamics from detailed study of experimental mod­els of tumorigenesis. The most informative of these has been the Rip-Tag trans­genic mouse. It carries a transgene in its germ line, in which the expression ofthe SV40 large T antigen (Sections 8.5 and 9.1) and small T antigen (Sidebar 18 . ) is driven by the promoter of the insulin gene. This promoter ensures expression of these viral oncoproteins in the ~ cells that form the islets of Langerhans in the pancreas (in which insulin is normally produced).

Tumor progression in the 400 or so islets of the mouse pancreas can be easily followed, because these islets can readily be distinguished from the surround­ing tissue of the exocrine pancreas, which is involved in manufacturing and secreting digestive enzymes. As many as half of these islets in a Rip-Tag mouse form hyperplastic nodules by 10 weeks of age, and 8 to 12% of the hyperplastic islets eventually progress to become angiogenic, that is, they acquire the abil­ity to recruit new blood vessels into their midst. By 12 to 14 weeks, about 3% of the initially formed hyperplastic islets finally progress to form carcinomas (Figure 13.37).

Early in tumor progression in the Rip-Tag mice, the hyperplastic pancreatic islets begin to expand slightly to a small diameter of about 0.1-0.2 mm and then halt their forward march (see Figure 13.37), at least for a while. In these small nests of tumor cells, cell division continues unabated, being driven by the onco­genic trans gene. However, the overall size of the tumor cell population within each islet remains constant, because of a compensating attrition of cells occur­ring through apoptosis. This mouse model suggests that in humans, small tumor nests may also remain in this dynamic but nongrowing state for many years, unable to break through the barrier that is holding them back.

In principle, the barrier to expansion of the tumor cell nests might be a physical one-lack of adequate space within the tissue for these cancer cells to multiply. But detailed histological analysis of these small nests of cancer cells reveals

5-7 weeks -50%

7-12 weeks 10%

12-14 weeks 2-4%

tumors

< 5 weeks 100%

normal islets hyperplastic islets angiogenic islets

564

Page 39: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Progression depends on the angiogenic switch

something quite different. Because these cells have not yet become angiogenic, they lack vasculature. The resulting hypoxia that they experience triggers p53­dependent apoptosis, which explains their high rate of attrition. (It is possible that other sub-optimal conditions within these poorly vascularized cell nests, including inadequate nutrient supply, high levels of carbon dioxide and meta­bolic wastes, and low pH caused by lactic acid accumulation, also contribute to the death of these cells.)

At some point in time, however, small clusters of these pre-neoplastic islet cells suddenly acquire the ability to provoke neoangiogenesis (see Figure 13.37). Once these cells learn how to induce capillaries to form nearby, they and their descendants seem to be liberated from the major constraint that has been hold­ing back their multiplication. This sudden, dramatic change in the behavior of the small tumor masses has been termed the "angiogenic switch."

These phenomena suggest an interesting idea, really a speculation: the body purposefully denies its cells the ability to readily induce angiogenesis. By doing so, the body erects yet another impediment to block the development of large neoplasms. According to such thinking, the angiogenic switch-a clearly impor­tant step in tumor progression-represents the successful breaching of this defensive barrier and the acquisition by cancer cells of a forbidden fruit: the ability to induce blood vessel growth at will.

One might conclude that the angiogenic switch in these transgenic mice is driven by the pre-malignant ~ cells' suddenly acquiring the ability to express and release VEGFs. Actually, these cells make large amounts of VEGFs long before the angiogenic switch occurs, as do fully normal pancreatic islets. However, the VEGF molecules secreted by these ~ cells are efficiently sequestered by the surrounding extracellular matrix (ECM). As a consequence, the VEGF molecules are unable to stimulate angiogenesis.

This sequestered state of the VEGF explains why the angiogenic switch in the Rip-Tag pancreatic islets is accompanied by the sudden appearance of substan­tial amounts of matrix metalloproteinase-9 (MMP-9; Section 13.5). MMP-9 acts in a targeted fashion to cleave specific structural components of the ECM, thereby releasing and mobilizing VEGF for active signaling to nearby endothe­lial cells. This MMP-9 is synthesized and released by inflammatory cells-mast cells and, quite possibly, macrophages-that have been attracted to the pre­malignant islets (Sidebar 13.7). Hence, in this particular tissue, tripping of the angiogenic switch ultimately depends on an acquired ability to recruit inflam­matory cells.

If the gene encoding VEGF is selectively deleted from the islet cells through genetic engineering, the islets survive and the early steps of tumor progression still proceed normally, but angiogenic switching never occurs. This reinforces the conclusion that VEGF molecules of islet cell origin playa critical role in trig­gering the onset of angiogenesis, and that recruited stromal cells cannot com­pensate for this absence ofVEGF by bringing in some of their own.

This scenario (Figure 13.38) involves heterotypic interactions among three dis­tinct cell types: (1) the release of still-unidentified signals from the pre-malig­nant islet cells that recruit mast cells and, quite possibly, macrophages; (2) the release of MMP-9 by these inflammatory cells to activate previously latent VEGF made by the pre-malignant islet cells; and (3) the proliferative response of endothelial cells to the activated VEGE In fact, yet other cell types are likely to be partners in islet cell angiogenesis. Thus, many of the endothelial cells may derive from recruited endothelial precursor cells in the circulation (EPCs, some­times called circulating endothelial progenitors, or CEPs); and the capillaries that arise are eventually covered, albeit haphazardly, with another cell type­pericytes-whose origins are unclear.

Sidebar 13.7 Mast cells from the bone marrow can playa key role in the angiogenic switch Rip-Tag mice canying the insulin-SV40 transgene in their germ line can be bred with oth­ers that lack the Kit growth fac­tor receptor (see Sidebar 5.7). Kit serves as the receptor for stem cell factor (SCF). an important growth factor that triggers the development of cer­tain subclasses of hematopoi­etic cells. Among other deficits, Kit-!- mice lack the ability to form mast cells. In Rip-Tag mice lacking Kit receptor, islet cell tumors are initialed at rates rou­tinelyobserved in standard Rip­Tag mice. However, these tumors never succeed in becoming angiogenic, and the rate of cell proliferation in these small growths is compensated by an equal rate of apoptotic death. Consequently, these tumors remain at a very · small size (0.1-0.2 mm diameter).

If these mice are provided with a bone marrow transplant containing wild-type hemato­poietic precursor cells, which cures their mast cell deficit, angiogenesis is initiated in the pancreatic islets, tumor cells in the islets are no longer lost through apoptosis, and large, life-threatening neoplasms appear soon thereafter. This proves that the abiiity to pro­voke angiogenesis depends on the actions of at least one nonendothelial component of the tumor-associated stroma­the mast cells originating in the bone marrow. The main contri­bution of these mast cells to the angiogenic switch seems to be the release of MMP-9, which proceeds to mobilize latent VEGF, thereby provoking angio­genesis (see Figure 13.38).

565

Page 40: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

Figure 13.38 The angiogenic switch and recruitment of inflammatory cells (A) The normal islet of Langerhans (circular area, center of left panel) is poorly vascu larized and is sustained largely through diffusion from the microvessels surround ing it. Following tripping of the angiogenic switch, there is an increase in the number of cells in the islet, due to tumor expansion, and, as seen here, a dramatic induction of vessel formation (right panel). Imaging was achieved by w holemount microscopy of lectin-perfused vessels. (B) According to this scheme, a pre­angiogenic, hyperplastic islet sends signals to the bone marrow and/or circulation (not illustrated) that lead to the recruitment of mast cells and macrophages. Once in the vicinity of the islet, these release metalloproteinases, notably MMP-9 (see Figure 1325E). MMP-9 then cleaves extracellular matri x (ECM) components, liberating vascular endothelial grow th factor (VEGF) from its sequestered state . VEGF proceeds to induce angiogenesis around the islet, thereby tripping the angiogenic switch. (A, from G. Bergers, D. Hanahan and L.M. Coussens, Int. 1. Dev. Bioi. 42995-1002, 1998; B, adapted from data in G. Bergers, R. Brekken, G. McMahon et aI., Nat. Cell Bioi. 2:737-744, 2000; VEGF YA Muller, B. Li , H.W Christinger et ai, Proc. Nat/. Acad. Sci. USA 94:7192-7197,1997)

Importantly, the Rip-Tag model does not typify the angiogenic mechanisms occurring during the formation of all types of tumors. For example, in some tumors, angiogenesis appears to increase progressively, as if a controlling rheo­stat is gradually being turned. This contrasts with the behavior of Rip-Tag islets, in which a binary, On-Off switch seems to be tripped.

Other tumors may depend on different angiogenic factors to provoke angiogen­esis. For example, when transformed mouse embryonic stem (ES) cells are deprived of both copies of the VEGF-A-encoding gene, they lose almost all their power to make malignant teratomas. In contrast, transformed adult mouse der­mal fibroblasts remain highly tumorigenic after they have been deprived of both copies of this gene. And the sarcomas generated by these transformed fibrob­lasts continue to grow even when the mice bearing them have been treated \"fith an antibody that binds and inactivates VEGF-R2 (the primary endothelial cell receptor that confers responsiveness to both VEGF-A and VEGF-B). This behav­ior is likely to be explained by the fact that transformed dermal fibroblasts can make a complex mixture of angiogenic factors-including VEGF-B, acidic and basic fibroblast growth factors (aFGFs and bFGFs), and transforming growth factor-ex (TGF-ex). This deployment of mUltiple angiogenic factors (see Table

(A)

(B)

recruiting signals -

bone ' ~marrow mast cells macrophages

hyperplastic islet y

angiogenic switch

angiogenic islet

in the ECM

inactive VEGF sequestered

soluble, active VEGF

566

Page 41: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

The angiogenic switch is a complex process

Table 13.2 Important angiogenic factors

Name Mol. wt. (kD)

Vascular endothelialGF (VEGF) 40- 45 Basic fibroblast growth factor (bFGF) 18 Acidic fibroblast growth factor (aFGF) 16.4 Angiogenin 14.1 Transforming growth factor-a (TGF-a) 5.5 Transforming growth factor-~ (TGF-~) 25 Tumor necrosis factor·a (TNFca) 17 Platelet-derived growth factor (PDGF) 45 Gran u locyte-.colony-sti mu lati ng' factor 17 Placental growth factor 25 Interleukin-8 (lLe8) 40 Hepatocyte growth factor (HGF) 92 Proliferin 35 Angiopoietin 70 Leptin 16

13.2), often observed in advanced human cancers, complicates the develop­ment of anti-angiogenesis cancer therapies, as we will see later.

13.8 The angiogenic switch initiates a highly complex process

Angiogenesis begins in the stroma surrounding the Rip-Tag tumors long before the basement membrane has been broken down. This behavior typifies that of many tumors in both mice and humans (Figure 13.39). Somehow, angiogenic

(A) (B)

normal skin

I

hyperplasia

dysplasia

Figure 13.39 Signaling through the basement membrane early in tumor progression (A) A human ductal in situ breast carcinoma (DC IS) contains a collection of carcinoma cells (purple) that are noninvasive and therefore have not yet breached the basement membrane (BM) that surrounds them and separates them from the mammary stroma. As is apparent, this DCIS has nonetheless succeeded in transmitting angiogenic signals through the BM into the nearby stroma that have resulted in the growth of a small vessel (dark brown) that surrounds the tumor mass but does not penetrate into the tumor itself because of the continued integrity of the BM. (B) In this transgenic mouse model of skin carcinogenesis, the human papillomavirus (HPV) 16 early region is expressed under the control of a keratin-14 gene promoter. In the early hyperplastic stage of tumor progression, the noninvasive carcinoma cells on the epithelial side (above) are able to transmit signals through the BM (dashed line) that provoke angiogenesis on the stromal side, as evidenced by the increased density of capillaries (red), detected in this case through their display of the CD31 antigen. Dysplastic tissue, in which the BM (dashed line) has not yet been breached, shows even more intensive angiogenesis in the nearby stroma. (A, courtesy of A.L. Harris; B, from L.M. Coussens, WW Raymond, G. Bergers et aI., Genes Oev. 13:1382-1397,1999.)

567

Page 42: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

(A) (8)

•. stroma .' ~

Figure 13.40 Recruitment of myofibroblasts on the stromal vimentin (dark brown) (B) The identification of many of these side of the basement membrane Myofibroblasts also foster stromal cells more specifically as myofibroblasts is indicated by the angiogenesis, in part through their abi lity to recru it endothelial fact that they can be immunostained w ith both an anti-vimentin precursor cells (EPCs) to the tumor stroma. (A) In human prostate antibody (red) and (X-smooth muscle actin (green); cells intraepithelial neoplasia (PIN)- a counterpart of ductal carcinoma in co-expre ssing both markers, and therefore identified as situ (DC IS) in the breast-the carcinoma cells (darker blue nuclei) myofibroblasts, are seen in yellow. These cells can also be identified remain on the epithelial side of the still -i ntact basement membrane as myofibroblasts through their expression of collagen I (not (BM), yet they have stimulated the accumulation of stromal shown) (From J Tuxhorn, G.E. Aya la, M.J Smith et aI., C/in. Cancer fibroblasts beyond the BM, as indicated by their display of Res. 8,2912-2923 , 2002.)

Signals are dispatched by benign cancer cells through the porous basement membrane (EM) in order to encourage increased angiogenesis on the stromal side of this membrane. Yet other signals transmitted through the basement membrane recruit myofibroblasts to the nearby stroma (Figure l3.40). As we read earlier, these myofibroblasts can also help to foster angiogenesis.

Nevertheless, this early angiogenesis is circumscribed, and it is clear that intense angiogenesis can begin only when cancer cells become invasive, pene­trate the basement membrane, and acquire direct, intimate contact with stro­mal cells (Figure 13.41). This suggests that tumor invasiveness and intense

(A)

Figure 13.41 Angiogenesis before and after acquisition of invasiveness Tumor angiogenesis is circumscribed as long as human carc inomas remain benign. However, once they become invasive, the intensity of angiogenesis increases, leading to a higher density of capillaries (brown) threading their way through tumors. (A) The capillaries ringing a benign prostatic intraepithelial neoplasia (PIN) lesion (left) are far fewer than those in an invasive prostate carcinoma (right). (B) Similarly, those in a benign ductal in situ breast carcinoma (DC IS; left) are far fewer than those in an invasive ductal carcinoma (righ t). (A, courtesy of J. Folkman; B, from N. Weidner, J.P Semple, W. R. Welch and J Fol kma n, N. Eng/. J. Med 324: 1-8, 1991.)

'''7 I

, . .. ..~

prostate cancer (PIN; in situ) invasive prostate cancer

human breast cancer (in situ) invasive human breast cancer 568

Page 43: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

The angiogenic switch is a complex process

(A) (8)

1.01.00~ 0.9~ n :::: ~'~: ' ': ~ ~ 0.75 .. ' nil n' rc HER2-NEGF

::1...­V"I:'=: .~ 0.8 Q)ii

~ ~ 0.50 ::J 0.7Vl , 0 Q)

Q) ... > ~ Cl. Q) - low microvessel density ~ 0.6025 .~ ::I "0 - high microvessel density ~ 0.5

u

00 5 10 15 20 0.4

time (years) 0.30 20 40 60 80 100

time (months)

angiogenesis are often tightly coupled processes. We will study tumor invasive­ness in detail in the next chapter.

In many human tumor types, the density of capillaries per microscope field increases in lockstep ,;vith increasing degrees of malignancy. For example, among human breast carcinomas that have already grown to a considerable size, those that have managed to attract dense networks of capillaries into their midst are indicative, on average, of a far worse prognosis than those that are poorly vascularized (Figure 13.42A). Moreover, patients with breast tumors that express large amounts ofVEGF (in addition to HER2) also fare badly following initial diagnosis and treatment (Figure 13.42B). Altogether, this suggests that the angiogenic switch is ooly the first of many gradations that enable tumors to become progressively more angiogenic and hence increasingly vascularized.

These striking correlations are actually susceptible to two alternative interpreta­tions. It is possible that intense vascularization enables cancer cells to grow more aggressively, thereby leading to poor clinical outcomes. Alternatively, intense angiogenesis is only a marker of an underlying aggressive phenotype but is not causally involved in driving high-grade malignancy. Available clinical data do not allow a clear resolution between these alternatives.

We have spoken of the angiogenic switch as if small numbers of cells within a tumor undergo this shift and proliferate to ultimately dominate within the tumor mass, thereby imparting the angiogenic phenotype to the tumor as a whole. In fact, analyses of cells isolated from explanted human tumors indicate great heterogeneity in the angiogenic powers of different subpopulations of cancer cells within a given tumor, with some cancer cells being highly angio­genic while others are poorly so. Even within tumor cell lines, individual sub­cloned cell populations show greatly differing angiogenic powers when engrafted in vivo (Figure 13.43). This introduces yet another idea: that within a tumor mass, long after the tripping of the angiogenic switch, the weakly angio­genic cancer cells rely on help from their friends-their strongly angiogenic neighbors-in order to acquire adequate vasculature.

(A) (8)

::2 400 Q)r::: fj~i // II ~ 300 Q)

E 600 ) ( " i I I Vl

:::J I 'I' , ' ~ 200 ] 400 I /! # 'f ! I o... !t li /; '- I .t::i 100~ 200 r ~I ; . ! E Z 0 ~j:;wr<(,~Jc _

o 10o 40 80 120 160 100 1000 10,000

Figure 13,42 Clinical outcomes and the intensity of angiogenesis (A) Breast carcinomas were analyzed for the density of capillaries (microvessel count), which was determined as the number of capillaries per microscopic field. This Kaplan-Meier graph demonstrates that those patients whose tumors had a higher microvessel count in their tumors (blue curve) had a markedly lower probability of disease-free survival in the 20 years following initial diagnosis than did those whose tumors had a lower microvessel count (red curve). (B) As indicated earlier (see Figure 4 .6), breast cancer patients w hose tumors overexpress HER2/Neu have a markedly poorer prognosis than those w ho do not. In the group analyzed here, all of the patients show ed metastatic cancer cells in one or more of the lymph nodes draining the breast. The differences in survival are even more dramatic w hen the levels of VEGF produced by their tumors are also measured . In this relati vely small clinical study, more than 80% of patients whose tumors expressed low, basal levels of both HER2/Neu and VEGF were alive eight years after diagnosis. In contrast, only about 35% of the patients whose tumors expressed elevated levels of both HER2/Neu and VEGF were still alive at this time . (A. from R. Heimann and S. Hellman. J. Clin. Onco/. 162686-2692, 1998; B, from GE. Konecny, YG. Meng, M . Untch et ai, Clin . Cancer Res. 101706- 1716, 2004)

Figure 13,43 Heterogeneous degrees of vascularization within a tumor cell population (A) W hen a population of cells in a human liposarcoma cel l line is subjected to single-cell cloning (in w hich the cells in each resulting ce ll clone all derive from a common ancestor), the various cloned cell popu lations show greatly differing abilities to form tumors (individual curves) when implanted into immuno-compromised mice. (B) When the resulting tumors are analyzed for microvessel density (microvessels per microscopic field) and tumor volume, plotted logarithmically here, it becomes clear that they have greatly differing angiogenic capabilities and that angiogenesis is likely to be the rate­limiting determinant of the ability to form tumors . (From E.G. Achilles, A Fernandez, EN Allred et aI. , J. Natl.

days post transplantation tumor volume (mm 3) Cancer Insf. 931075- 1081 , 2001.)

569

Page 44: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

Figure 13.44 Defective tumor angiogenesis in Id1+1- Id3-i- mice Mutant mice (Id MUT), w hich lack three of four Id gene copies (and have an IdJ+l- Id3-1- genotype), show impaired mobilization of endothelial precursor cells (EPCs) from the bone marrow and thus impaired recruitment of circulating EPCs into tumors that they may carry. (A) Wild-type (wt) mice bearing Lewis lung carcinoma (LLC) cells develop rapidly growing tumors (red curve), w hile those of the mutant strain (Id MUT) are unable to support vigorous tumor growth (brown curve) This defect is essentially reversed if the bone marrow (BM) of the mutant mice is eliminated by irradiation and replaced with transplanted wild-type bone marrow (blue curve, wt BM Irr Id MUT), indicating that the recruitment of bone marrow-derived cells is defective in the mutant mice and is responsible for the inability of the tumor to grow in them. (B) This defective recruitment can be further localized by the use of plugs of Matrigel (an extracellular matrix material) that have been impregnated with VEGF and are then implanted subcutaneously. These plugs are able to recruit neovasculature when implanted in wild­type mice (left) but not in the mutant mice (center). However, if the mutant mice receive a graft of wild-type bone marrow cells, the defect is cured and now angiogenesis within these plugs is comparable to that seen in w ild-type mice (right). (From D. Lyden, K. Hattori, S. Dias et aI., Nat. fVled. 71194-1201,2001)

(A) 600

wt~ 500 wt 8M Irr Id MUTEo

S 400

'" ~ '" 300OJ u

't'" 200 ~

Id MUT 100

o 2 4 6 8 10 12 14 16 18 20 22 24

days

Id MUT wt 8M Irr Id MUTwt

The angiogenic switch may have effects that extend far beyond the immediate vicinity of a tumor. At first, the tumor-associated blood vessels that are formed may sprout directly from existing vessels in adjacent normal tissues. At this stage, it is likely that the endothelial cells forming these new capillaries arise through the proliferation of endothelial cell precursors residing in these already­formed vessels. However, as tumor progression proceeds, neovascularization is likely to rely increasingly on the recruitment of endothelial precursor cells (EPCs) that originate in the marrow and travel via the circulation to the tumor mass.

VEGF released into the circulation by a tumor stimulates the production ofEPCs in the bone marrow and helps to attract EPCs to the tumor mass. As described earlier (Section 13.4), stroma-derived factor-1 (SDF-1, also called CXCLl2), which is liberated by stromal myofibroblasts, can also help in this recruitment. Once they have arrived in the tumor mass, the EPCs are induced to differentiate into functional endothelial cells and construct the tumor-associated vascula­ture. Detailed analyses have shown that the proportion of endothelial cells derived from circulating EPCs (versus those arising from nearby capillaries) varies greatly from one type of tumor to another.

These dynamics ofEPC recruitment are dramatically illustrated by the behavior of mice that lack one copy of the Jdl gene and both copies of the Jd3 gene. Recall that these Jd genes encode transcription factors regulating differentiation of a variety of cell types (Section 8.11). Many of these mutant mice survive to adult­hood and show a very peculiar phenotype: they are defective in neoangiogene­sis and will not permit several types of engrafted tumors to grow (Figure 13.44A).

The defect in these Idl+ l - Jd;j/- mice can be cured by transplanting wild-type stem cells into their bone marrow (see Figure 13.44A) . Alternatively, the subset of bone marrow stem cells whose production is normally spurred by high levels of circulatingVEGF can be harvested from a wild-type mouse and transplanted into the bone marrow of Jdl+ l - Jd;)I- mice. These mice soon generate large numbers of endothelial precursor cells (EPCs) in their circulation and, following implantation of cancer cells, develop rapidly growing tumors. Taken together, these observations indicate that these tumors rely on VEGF to mobilize EPCs

570

Page 45: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Angiogenesis can be blocked by specific inhibitors

from the bone marrow (Figure 13.44B) and to recruit the resulting circulating cells into the tumor stroma. These findings, as well as others demonstrating the recruitment of myofibroblast precursors into tumor stroma (Sidebar l3.3), show that primary tumors can extend their reach throughout the body long before they metastasize.

13.9 Angiogenesis is normally suppressed by physiologic inhibitors

In finely tuned physiologic processes, the actions of positive effectors must be counterbalanced by negative regulators. We have read much about the positive effectors of angiogenesis, such as VEGF and bFGF, but their antagonists have remajned offstage until now. They turn out to be as interesting and important as the angiogenic factors whose actions they antagonize.

During the process of wound healing, for instance, the burst of angiogenesis that is required to repair the wound site must be shut down once the newly formed capillaries have reached a density that suffices to support normal tissue function. This shutdown is achieved, at least in part, by suppressing formation of the HIF-1 transcription factor. Its assembly is induced under hypoxic condi­tions and is reversed once normal oxygenation in the wound site has been restored (see Section 7.12).

In addition, a number of the components of the extracellular matrix are used by tissues to actively block excessive angiogenesis (Table 13.3). The best character­ized of these is the thrombospondin-1 (Tsp-lJ protein, which is secreted by many cell types into the surrounding extracellular space, where it forms homotrimers and carries out several distinct functions. Most important for our discussion, Tsp-1 associates with a receptor (termed CD36) that is displayed on the surfaces of endothelial cells and halts their proliferation. In addition, research indicates that Tsp-1 treatment of endothelial cells causes them to release Fas ligand (FasL), the pro-apoptotic signaling protein that acts by bind­ing to the Fas death receptor. Recall that the latter, once it has bound its ligand, activates an intracellular caspase cascade that triggers apoptosis (Section 9.14) . Therefore, once Tsp-1 causes endothelial cells to release FasL, the latter may act in an autocrine fashion to trigger the death of these cells in the event that they also display the Fas receptor.

Interestingly, the Fas receptor is displayed on endothelial cells that are actively proliferating or have recently ceased proliferation, but its display is suppressed once these cells have successfully formed mature capillaries and retreated into quiescence (Figure 13.45A). This seems to explain a most intriguing aspect of Tsp-1 behavior: it selectively inhibits and causes regression of newly formed and still-growing capillaries but has little if any effect 011 already-formed, mature capillaries. In fact, a number of other natural anti-angiogenic factors also seem to exhibit such selectivity. The actions of these other anti-angiogenic agents seem, once again, to depend upon activation of the pro-apoptotic caspase cas­cade in endothelial cells, since compounds that inhibit the caspase enzymes also protect endothelial cells from the anti -angiogenic effects of Tsp-1 and the other natural blockers of angiogenesis.

Transcription of the TSP1 gene is strongly induced by p53, ostensibly as part of the p53-mediated emergency response that leads to a generalized shutdown of cell proliferation and tissue growth. Conversely, the loss of p53 function, which is seen in almost all human tumors (Chapter 9), leads to a substantial decrease in Tsp-1 levels. This permits angiogenesis to be induced by cells that normally would have been prevented from doing so by the high Tsp-1 concentrations in the surrounding extracellular matrix.

571

Page 46: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

Table 13.3 Endogenous inhibitors of angiogenesis

Inhibitor Description

A. Derived from extracellular matrix Arresten fragment of type IV collagen U1 chain of vascular

basement membrane Canstatin fragment of type IV collagen U2 chain of vascLilar

. basement membrane EFC-XV fragment of type XV collagen Endorepellin fragment of perlecan Endostatin fragment of collagen type XVIII Anastetlin fragment of fibron.ectin .. FibLilin fragment of basement membrane protein · Thrombospondin-l and -2 ECM glycoproteins Tumstatin fragment of type IV collagen U3 chain .Chondrornodulrn-I component of cartilage ECM Troponin I cornponent of cartilage ECM

B.Non-matrix-'CIerived (;irowth factors and cytokines Interferon-u (IFN-u) cytokine Interleukins (lL-1P, -12, -18) cytokines Pigment epithelium-growth factor

derived factor (PEDF) Platelet factor-4 released by platelets during degranulation Other types Angiostatin fragment of plasminogen

. Antithrombin III fragment of antithrombin III 2~Methoxyestradiol endogenous metabolite of estrogen PEX fragment of MMP~2 Plasminogenkringle 5 fragment of angiostatin Prolactin fragments specific cleavage fragment Prothrombin kringle 2 fragment of prothrombin sFlt-' soluble form of VEGF-R1(= Flt-1}

.. TIMP-2 inhibitor ofmetalloproteinase-2 TrpRS fragment of tryptophan yl-tRNA synthetase Vasostatin fragment of calreticulin

Adapted from P Nyberg , L. Xie and R. Kalluri, Cancer Res. 65:3967-3979, 2005.

The Ras oncoprotein, acting through a complex signaling cascade, acts in the opposite fashion, since it causes shutdown of TSP1 gene expression. The result­ing absence of significant levels ofTsp-l can also contribute substantially to the elevated angiogenic powers of ras-transformed cells compared with their nor­mal neighbors. In one study of melanomas arising in transgenic mice, expres­sion of the H-ras oncogene, which had been used to initiate these tumors, was shut down after they had grown to a considerable size. These tumors collapsed rapidly thereafter, and this collapse was directly traceable to their loss of func­tional vasculature, which began to disintegrate within 6 hours after the loss of Ras function (Figure 13.4SB). The observed rapid entrance into apoptosis of endothelial cells forming the tumor vasculature preceded any decline in the lev­els of VEGF (which provides survival signals for endothelial cells). While not demonstrated directly in these experiments, it seems likely that this apoptosis derived from the rapid re-expression ofTsp-l that follows close on the heels of Ras shutdown.

Tsp-l is surely a major governor of angiogenesis, but as hinted above, it is only one of a large cohort of natural inhibitors of angiogenesis that are found in the spaces between cells (see Table 13.3). The existence of several other anti-angio­genic molecules was first suggested by observations of the behavior of certain primary tumors and their derived metastases. In some mouse models of tumori­genesis, metastases were found to remain small in size as long as the primary

572

Page 47: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

1 FasL

~

....,

Angiogenesis can be blocked by specific inhibitors

(A) (B) pericyte

r:::: \i"2 ; If?? e~

I:;w ~- J!sh ~ «~ ~

O'L- endothelial cells

vascu lar basement ,, membrane

angiogenic ""'- ( .... . ]\

agent ORY recently formed mature endothelial cell endothelial cell .,

[ ~ ) Fas Figure 13.45 Thrombospondin, endothelial cell

survival, and tumorigenesis (A) The stimulation of resting endothelial cells with angiogenic agents, such as VEGF orl~j bFGF, creates activated, growing endothelial ce lls (left branch), w hich soon express the Fas death receptor on their surface. Subsequent treatment of mature and recently formed endothelial cells with Tsp-1 (blue) causes both groups of cells to secrete FasL (green), the ligand of Fas . However, because only the activated endothelial cell displays the Fas receptor, it is preferentially induced to enter into

FasL apoptosis by Tsp-1 . This seems to explain w hy Tsp-1 can block new angiogenesis but has relatively minimal effects on

cell already-constructed vasculature, whose endothelial cells are survival

only rarely involved in active growth . (B) The loss of ras oncogene expression can lead to a rapid increase in Tsp-1 expression. This may explain why, in thi s transgenic mouse model of melanoma development, shutdown of ras oncogene expression in already-formed tumors leads to

! rapid collapse of the tumor-associated vascu lature and tumor regression . Here, the TUNEL assay (Figure 3B 0 ) for apoptosis reveals apoptotic endothelial ce lls within a capillary (arrow) of a regressing melanoma. (A, adapted from o.v. Vol pert T. Zaichuk, W. Zhou et aI., Nat. Med. 8:349-357, 2002; B, from L. Chin, A. Tam , J. Pomerantz et

apoptosis aI., Nature 400468-472, 1999 )

tumor that spawned them continued to grow. However, the moment the pri­mary tumor was surgically removed, the metastases began to grow vigorously. This behavior is echoed by anecdotal reports of cancer surgeons, who have observed that after the successful surgical removal of a primary tumor, substan­tial numbers of metastases may suddenly sprout and flourish, doing so within several months' time.

Such observations suggested that some type of inhibitory substance released by the primary tumor acted, via the circulation, to suppress the proliferation of dis­tant nests of metastatic cells. More specifically, these inhibitory factors, what­ever their nature, seemed to block angiogenesis in these secondary growths, which failed to expand to a diameter of more than several tenths of a millimeter. Once the primary tumors were excised, the hypothetical inhibitory factor(s) dis­appeared from the circulation, removing some constraint on the growth of already-seeded metastases.

The subsequent isolation of these circulating factors, initially from the urine of tumor-bearing animals, yielded the intensively studied angiostatin and endo­statin molecules. Determination of the amino acid sequences of these two pro­tein species indicated that they arise as cleavage products of familiar proteins of

573

Page 48: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

Sidebar 13.8 TIMPs suppress angiogenesis in multiple ways Accumulating evidence indi­cates that the TIMPs can act on other molecular targets besides the MMPs. For example, the VEGF and fibroblast growth fac­tor (FGF) receptors, which are req uired for endothelial cell responses to these two critical angiogenic growth factors, are also inhibited by TIMP-2. Evidence indicates that this receptor antagonism proceeds through a quite indirect route: TIMP-2 binds to a cell surface integrin, and the latter, acting via its cytoplasmic tail, activates an intracellular phosphatase that proceeds to dephosphory­late and thereby shut down sig­naling by the VEGF and FGF receptors. These diverse anti­neoplastic activities of TIMPs have made them attractive agents for development as anti­cancer therapeutics. However, in spite of numerous research-and­development projects, none of the TIMPs or TIMP analogs has yet proven to be a useful antago­nist of tumor development when lIsed in the clinic.

the extracellular matrix (ECM) or plasma. With the passage of time, yet other anti-angiogenic substances have been isolated (see Table 13.3), many of which are also formed by the proteolysis of extracellular proteins. Taken together, these discoveries suggest that as angiogenesis proceeds in normal tissues during development and wound healing, this process is eventually curtailed by the accumulation of anti-angiogenic protein fragments in the extracellular space. These naturally occurring anti-angiogenic substances can therefore be depicted as important components of negative-feedback loops operating to ensure that excessive vascularization of a tissue does not occur.

In more detail, the specialized basement membrane surrounding most capillar­ies is the source of several proteins that have potent anti-angiogenic powers. Proteolytic cleavage of collagen XVIII yields the 20-kD C-terminal fragment that was discovered as endostatin. Yet another, tumstatin, derives from cleavage of one of the chains of collagen IV (which constitutes as much as half of the capil­lary basement membrane), yielding a 28-kD fragment. Collagen IV cleavage also yields another anti-angiogenic fragment termed arresten. And cleavage of plas­minogen (the precursor of the plasmin protease involved in activating coagula­tion) generates the 38-kD internal fragment known as angiostatin.

Another important class of natural anti-angiogenic proteins function as antag­onists of the matrix metalloproteinases (MMPs). VEGF stimulates the localized production of MMPs 1 to 4, which enable elongating capillaries to invade through the extracellular matrix between cells. A class of secreted proteins, termed tissue inhibitors of metalloproteinases (TIMPs), can prevent this elon­gation by blocking the actions of these and other MMPs. For example, by forcing the ectopic expression of TIMP-2 in tumor cells, researchers have blocked the angiogenic and thus tumorigenic powers of these cells. However, the precise mechanisms by which TIMPs block angiogenesis are still not clearly resolved (Sidebar 13.8).

Arguably the most bizarre natural angiogenesis inhibitor is a variant form of tryptophanyl-tRNA synthetase. This enzyme is normally responsible for charg­ing a tRNA with the amino acid tryptophan, and therefore is one of the core components of the protein synthesis apparatus. However, an alternatively spliced version of the mRNA encoding this enzyme leads to a truncated form of this protein, called mini TrpRS, which is secreted from cells; its production is inducible by interferon treatment of cells. This protein, which can specifically trigger the apoptosis of endothelial cells, is an example of the opportunistic use by evolution of any substance at hand to solve certain biological problems.

When integrated, these disparate observations about the physiologic regulators of angiogenesis reinforce the idea, cited earlier, that angiogenesis is not a binary state-either on or off. Instead, different types of tumor cells acquire greater or lesser angiogenic powers, and even within a given tumor, the tumor cells are likely to show differing abilities to attract vasculature. Such behavior is likely to be explained by a scheme (Figure 13.46) in which the balance between pro- and anti-angiogenic factors determines whether neoangiogenesis will proceed, and if so, how intensively regions within tumors will become vascularized.

13.10 Certain anti-angiogenesis therapies hold great promise for treating cancer

The more complex a system becomes, the more vulnerable it is to various types of disruption. The process of angiogenesis, as described here, clearly falls in the class of highly complex systems, as shown by its dependence on multiple cell types and signaling molecules. Indeed, while we have featured several impor­tant angiogenic factors, by some counts there are at least a dozen involved in regulating various steps of vascular morphogenesis (see Table 13.2). 574

Page 49: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Anti-angiogenesis therapies hold great promise

_on

_off

activators • inhibitors VEGF-A thrombospondin-l, -2 VEGF-8, -c interferon ClJ~ FGFl (aFGF) angiostatin FGF2 (bFGF) endostatin other FGFs collagen IV fragments etc. etc.

For those researchers intent on developing new types of anti-cancer therapeu­tics, this complexity offers mUltiple targets for intervention. In particular, highly targeted therapies may be devised to inhibit the several cell types that partici­pate in angiogenesis as well as the multiple signaling channels through which they intercommunicate. Since tumors depend absolutely on angiogenesis to grow above a certain size (-0.2 mm diameter), any successes in blocking angio­genesis or in undoing the products of angiogenesis should represent a highly effective strategy for treating cancer. Microscopic tumors should be prevented from growing larger, while larger tumors should collapse once their already­established blood supply disintegrates.

In principle, anti-angiogenesis therapies have a major advantage over those directed at the neoplastic cells within tumors. As we will learn in Chapter 16, one of the great frustrations of anti-cancer drug development comes from the fact that, sooner or later, tumors that have responded initially to a drug treatment will relapse and become refractory (resistant) to further treatment by the drug. Almost always, these relapses can be traced to the emergence of drug-resistant variants within tumor cell populations; these variants arise at an almost-pre­dictable frequency and proceed to proliferate and regenerate aggressively grow­ing tumor masses. The emergence of these drug-resistant variants seems to be one of the consequences of the highly unstable genomes of cancer cells (see Sections 12.11 and 12.12) and their resulting ability to spawn mutants at high frequency.

Many anti-angiogenesis therapies, in stark contrast, are directed at killing the genetically normal cells that have been recruited into tumor masses and co­opted by the cancer cells to do their bidding. There is every reason to believe that the endothelial cells vvithin tumors possess normal, stable genomes and are therefore stable phenotypically. Hence, drug therapies directed against these cells are not likely to select for the outgrovvth of drug-resistant variants, and tumors should not, in theory, become refractory to anti-angiogenic drug ther­apy (see, however, Sidebar 13.9).

This interest in treating tumor-associated endothelial cells is further heightened by the peculiar biology of these cells. They are continually being formed and lost within tumor masses, with lifetimes measured as short as a week, while their counterparts that line normal blood vessels elsewhere in the body rarely divide and have lifetimes that are measured in hundreds of days, some being as long as seven years. Cycling cells (Le., cells racing around the active cell cycle) are, almost always, far more sensitive to drug-induced killing than are quiescent cells. For this reason, cytotoxic therapies directed against endothelial cells should have drastic effects on the tumor-associated vasculature, while leaving blood vessels located elsewhere in the body unscathed (but for the caveat cited in Sidebar 13.9).

Figure 13.46 Balancing the angio­genic switch This diagram presents the major physiologic regulators that work to promote or inhibit angiogenesis within tissues and indicates that it is the balance between these two countervailing groups of regulators that determine whether or not angiogenesis proceeds. While the angiogenic switch IS

depicted here as an essentially on-off binary decision, in fact the vasculature that grows within a tumor mass after the switch has been activated shows various gradations of development. (From D. Hanahan and J Folkman, Cell 86353-364, 1996, and courtesy of D. Hanahan.)

575

Page 50: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

Sidebar 13.9 Tumors may outsmart even the best anti­angiogenic therapies Much of the allure of anti-angiogenic therapies comes from the likelihood that the cells being tar­geted, notably the endothelial cells, are unlikely to generate drug-resistant variants. The complexity of heterotypic sig­naling may, however, allow tumors to circumvent even the cleverest anti-angiogenic therapies. Imagine, for example, that we devise a strategy to block the VEGF signaling that is so critical for the formation of new vessels in a tumor and their subsequent maintenance (since VEGF is required for endothelial . cell survival) . A tumor treated in: th)s way should rapidly collapse, since its capillary beds will disinte­grate. Indeed, just such a response has been observed in Rip-Tag mice that developed pancreatic islet tumors (see Figure 13.37): When treated with an anti-VEGF-R2 anti­body, their tumors regressed by more than 50%. However, the residual, surviving tumor cells responded by spawning variants that acquired the ability to produce elevated levels of fibroblast growth factors (FGFs), which are also potent angiogenic factors. These growth factors then supplanted VEGF as the main conveyor of signals from the islet tumor to the endothelial cells and succeeded in triggering the

regeneration of vasculature and, in turn, the rebirth of a vigorously growing tumor. Accordingly, truly effective anti­angiogenesis strategies will require the inhibition of multi­ple angiogenic pathways.

Anti-angiogenesis therapies are also attractive because of their potential selectivity for killing the proliferating endothelial cells within tumors, since many blood vessels in these growths seem to be in a constant state of formation and collapse, and are therefore susceptible to cytotoxic drugs that may leave the quiescent endothelial cells in nor- . mal tissues untouched. For example, dramatically contrast­ing groWth states of normal and tumor-associated blood vessels can be seen iIi many mouse models of cancer, in which erigrafted tumors expand rapidly. However, it is pos­sible that many of the vessels in slowly growing human tumors may have existed for years and therefore inayhave had ample time to consolidate and mature into robust, well-structured vessels in which the endothelial cells turn over slowly. These tumor-associated vessels may then ·be as resistant to anti-angiogenic drugs as the normal blood ves- . sels elsewhere in the body.

We will learn in great detail about the general principles of anti-cancer therapy later in Chapter 16 but will take the opportunity here to describe the details of emerging therapies that are directed specifically against the tumor-associated vasculature. Some of the first efforts in this area have come from experiments in which the natural anti-angiogenesis inhibitors mentioned above have been used to treat tumors borne by mice.

Since these agents, such as angiostatin and endostatin, are native to the body, they have the advantage of being reasonably well tolerated without toxic side effects. They have virtually no effect on the in vitro proliferation of a variety of cells, and instead are biologically active only on endothelial cells that are actively participating in neoangiogenesis in vivo. Moreover, these proteins can persist for some time in the circulation, thereby increasing the exposure of tumor vasculature to their anti-angiogenic effects. On the negative side, their manufacture in large quantities, like that of all proteins, is costly and challeng­ing, and their precise mechanisms of action have been elusive.

Both angiostatin and endostatin had only modest effects in blocking the angio­genic switch in the pancreas of Rip-Tag mice (Section 13.7). But when applied to already-vascularized small tumors, endostatin reduced by more than 80% their subsequent growth, while angiostatin reduced this size by 50%. When large, well-established tumors were treated with either of these agents, relatively little effect was seen, but the two introduced together caused a 75% reduction in the mass of such tumors. Long-term endostatin treatment of mice bearing tumors formed from a lung carcinoma cell line led to regression ofthe tumors. This suc­cess might suggest that endostatin may be a highly effective therapeutic in the oncology clinic. Ho"vever, responses like these in mice are rarely predictive of similar efficacy in the oncology clinic. Some dramatic responses have been reported following endostatin treatment in a small number of patients treated in early clinical trials.

Yet another class of natural angiogenesis antagonists (see Table 13.3) are the interferons, which are usually studied in the context of their ability to modulate the activities of various cell types operating in the immune system. Interferon­a and -~ have proven to be potent suppressors of the synthesis of basic fibrob­last growth factor (bFGF) and of interleukin-8 (IL-8) , both of which are strong

576

Page 51: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Anti-angiogenesis therapies hold great promise

angiogenic agents. And administration of interferon-a has proven to be useful in causing the regression of some hemangiomas (endothelial cell tumors) as well as Kaposi's sarcomas, which are also of endothelial cell origin (possibly the endothelial cells forming lymph ducts). In both cases, the tumor regressions have been attributed to the anti-angiogenic effects of the interferon .

An experiment reported in 1991 provided one of the earliest indications of the promise of another type of anti-angiogenic therapy. In this work, cancer cells were engineered to release a modified basic fibroblast growth factor (bFGF), which greatly increased their tumorigenicity in mice, simply because this bFGF strongly enhanced the angiogenic powers of these cells. Use of a monoclonal antibody that specifically bound and neutralized this bFGF (but had no effect on the endogenous bFGF of the mouse) blocked the angiogenicity of these tumor cells and led to a dramatic reduction in tumor volume. Within two years, a sim­ilar experiment was performed with an anti-human-VEGF monoclonal anti­body. It succeeded in blocking the proliferation of two human sarcoma cell lines as well as a glioblastoma in Nude (immunocompromised) mouse hosts.

A more modern version of this therapy came a decade later with the use of a monoclonal antibody that binds and neutralizes VEGF-A. This antibody, termed variously Avastin and bevacizumab, showed significant efficacy in large-scale clinical trials. For example, patients with metastatic colon carcinoma who were treated with this antibody plus chemotherapy (the drug 5-fluorouracil) sur­vived, on average, four months longer than patients treated with chemotherapy alone, and the addition of Avastin to conventional chemotherapy extended the survival of patients with non-small-cell lung carcinoma (NSCLC) by about two months. Similarly, Avastin could retard the progression of renal cell carcinomas in patients, but in the end had no effect on their long-term survival. It is plausi­ble that the synergistic effects of Avastin with conventional chemotherapeutic drugs derive directly from the ability of this VEGF-A inhibitor to normalize tumor-associated vasculature (see Figure 14 0 ), thereby greatly facilitating the delivery of drugs to the tumor parenchyma.

Some synthetic low-molecular-weight anti-angiogenic compounds have been discovered or developed that are directed against various molecular targets in the angiogenic program. The first of these was fumagillin, an anti-angiogenic compound of fungal origin, and its chemical derivative, TNP-470 (Table 13.4C) . These two compounds were found to inhibit the proliferation of endothelial cells both in vitro and in vivo; this suggests that their anti-angiogenic effects in vivo derive from their ability to prevent the growth of new capillaries, which depends on endothelial cell proliferation. Importantly, TNP-470 had no effect on the proliferation of tumor cells in vitro but strongly blocked their tumori­genicity in mice. For example, this drug reduced by 70 to 80% the sizes of pan­creatic islet tumors arising in the Rip-Tag mice. The mechanism of action of TNP-470 remains obscure, but is under active investigation. Its proven ability to inhibit the methionine aminopeptidase-2 enzyme in endothelial cells may one day help to explain its preferential effects on these cells while it has almost no effects on other cultured cell types.

The most informative studies of angiogenic inhibitors have come using syn­thetic receptor inhibitors (see Table 13.4B) in the transgenic Rip-Tag model of pancreatic islet tumorigenesis (see Section 13.7). Two types of low-molecular­weight synthetic compounds have been utilized in attempts to block various stages of islet tumor progression. One of these drugs is directed against VEGF­R2, which is the main receptor for angiogenesis in this tumor model. By inhibit­ing the tyrosine kinase of this receptor, the agent termed SU5416 should mimic the effects of Avastin (the anti-VEGF monoclonal antibody described earlier), that is, both should be able to shut down VEGF signaling. Drugs of a second class, such as an agent termed SU6668, are directed primarily against the tyro­sine kinase of the PDGF receptor. Our discussions here about the role of PDGF

577

Page 52: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

Table 13.4 Angiogenesis inhibitors and their development and use in clinical trials

Name Status Responses

A. Endogeno;:'-s inhibitors of angiogenesis -Endostatin in clinical trial scattered responses Interferonsc<x and -~ effective in treating hemangioblastomas Kaposi's sarcomas; limited efficacy against

most other types of tumors

B. Agents that block VEGF and VEGF-R signaling Avastin anti-VEGF MoAb in clintcal trial delayed progression 1-3 months in lung,

3~months in colon SU5416 inhibitor of trial abandoned severe vascular toxicities

VEGF-R2 (Flk-1) ZD6474 inhibitor of under clinical test

VEGF-R2 CP547,632 inhibitor ii". trial -

of VEGF-R2

C.Miscelh:lneous other drugs Thalidomide - in trial inhibits bFGF- and VEGF-dependent angiogenesis

--- Squalamine sterol in trial strong anti-angiogenic activity from shark liver

Celecoxib anti­ in trial mUltiple anti-neoplastic effects inflammatory drug

ZD6126 in trial antagonist of tubulin in endothelial cell cytoskeleton

Fumagillin and TNP-470 in trial;. slowed tumor growth antagonist of methionine aminopeptidase in endothelial cells

D. Inhibitors of ECM breakdown-MMP inhibitors Marimastat in clinical trial no delay of tumor progression Prinomastat in clinical trial no slowing of tumor progression ­BMS275291 in clinical trial BAY12-9566 in clinical trial Neovastat (shark in clinical trial

cartilage MMPI)

in recruiting pericytes and smooth muscle cells to growing capillaries (Sidebar 13.1) indicated that these "mural" cells are highly important for consolidating and strengthening recently formed capillaries.

SU5416, the anti-VEGF-R agent, was able to block 90% of the early-stage, dys­plastic islets from undergoing the angiogenic switch, thereby holding them to a small size and a noninvasive state (Figure 13.47C). However, SU5416 had no effect on late-stage, well-established tumors, which continued to progress in spite of its presence in equally mgh concentrations. Hence, in the early stages of angiogenesis, VEGF signaling plays a critical role, while later on, this process seems to become increasingly independent ofVEGF.

The anti-PDGF receptor agent, SU6668, had a far weaker effect on preventing dysplastic islets from undergoing the angiogenic switch, reducing by about half the islets that did so (see Figure 13.4 7C). But it was far more potent than the anti­VEGF-R drug in treating the late-stage, advanced twnors, reducing their size by about half and substantially reducing their vascularity (Figure 13.47D). Importantly, the only cells expressing PDGF-R in and near these tumors were the capillary-associated pericytes and related smooth muscle cells (Figure 13.47B) , indicating that these mural cells were the targets of SU6668 action. Indeed, microscopic examination confirmed that SU6668 had prevented these mural cells from associating with and reinforcing the capillary tubes formed by endothelial cells. Together, these experiments showed that the initial steps of angiogenesis could proceed reasonably well without PDGF receptor function, but that later in tumor progression, PDGF signaling, and thus the involvement of mural cells, became increasingly important to angiogenesis and growth of the

578 tumor masses.

Page 53: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Anti-angiogenesis therapies hold great promise

Combination therapy using the two agents proved to be a highly potent way of intervening at various stages of tumor formation (see Figure 13.47C and D). Thus, simultaneous inhibition of both VEGF and PDGF receptors prevented

(A) (B)

untreated control

(C) prevention60

E E 45 Q)

E ~ 30 >

~ 15 :::l , +' ob i .

~'-> to~ ,to :0' ~ totO ~t>< o<f"

,->-0 ,->-0 c

(F). 'd' Iperlcytes provi e survlva functions to endothelial cells

endothelial cells are partially resistant to VEGF-R inhibition and a.re less sensitive to chemotherapy

SU6668 + SU5416 treatment

(D) (E)regression

200 N'>

E E 150 Q)

E :::l '0100 > oE 50 :::l +' I •o ~'-> to~ ,to ,:::-'0' tumors at 13.5 weeks +3.5 weeks treatment ~ totO ~t>< 0

,->-0 ,->-0 c

~ impaired support or protection by pericytes

targeting pericytes e.g., via PDGF receptor

inhibitors

endothelial cells are very sensitive to VEGF-R inhibition and chemotherapy

Figure 13.47 Angiogenesis inhibitors as treatments of islet cell carcinogenesis The Rip-Tag transgenic mouse model of pancreatic islet cell carcinogenesis makes possible the testing of anti-angiogenic pharmacologic inhibitors. (A) The treatment of mice bearing established angiogenic islet tumors (left) for 4 weeks with the PDGF receptor inhibitor SU6668 plus the VEGF-R inhibitor SU5416 results in the regression of the vasculature (right). The lumina of the capillaries are labeled yel/ow w hile the associated pericytes are labeled red. (B) The actions of the SU6668 PDGF-R inhibitor could be traced specifically to its effects on the pericytes that act as support cells for the endothelial cells. In this image, an antibody reactive with the PDGF-R reveals that the only cells within the islets that express this receptor are the pericytes (pes, green) that are closely associated with the endothelial cells (Ees, red). Nuclei are stained blue. (e) While neither the SU6668 PDGF-R inhibitor (blue bar) nor the SU5416 VEGF receptor inhibitor (red bar) was able, on its own, to fully prevent the formation of such tumors, the two applied in concert (combined, purple bar) succeeded in doing so. PBS, phosphate-buffered saline control (green) (D) In an attempt to cause regression of already-formed tumors, SU5416 and SU6668 were introduced at 12 weeks of age into Rip-Tag mice either singly or in combination and the size of tumors was measured 4 weeks later. While the SU5416 VEGF-R

antagonist on its own (red bar) showed only minimal reduction of tumor volume, the SU6668 PDGF-R antagonist showed greater effects (blue bar), and the two together (brown bar) worked synergistically to reduce overall tumor volume by approximately 85%. This indicates, once again, that antagonists of the endothelial cells (w hich depend on the VEGF-R) together w ith antagonists of the supporting pericytes (which depend on the PDGF-R) can act synergistically in anti-angiogenic therapy. (E) The combined effects of these two drugs can be seen: when 13.5-week-old mice that have developed substantial pancreatic islet tumors (red growths, left) are treated for 3.5 weeks with this combination therapy, their tumors largely regress (right) (F) A schematic summary of these and other observations indicates that endothelial cells depend on closely apposed pericytes for various types of biological support. Inhibition of PDGF signaling, as achieved by certain inhibitors, causes dissociation of pericytes from endothelial cells and renders the latter sensitive to various types of subsequent therapy, including inhibition of VEGF-R function . This emphasizes the fact that anti-angiogenesis therapy is most effective w hen two synergistically acting treatments are applied . (A, B, and E, from G. Bergers, S. Song, N. Meyer-Morse et aI., I ~in . Invest. 111: 1287-1295, 2003; e and D, courtesy of D. Hanahan; F, from K. Pietras and D. Hanahan, I Oin. Onco/. 23939-952, 2005.) 579

Page 54: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

angiogenic switching, holding virtually all (>98%) islets at a pre-angiogenic stage. This drug combination blocked the further expansion of small, already­established angiogenic tumors by 90% and caused an approximately 80% regression of large tumors, as did another PDGF-R inhibitor, the drug Gleevec, which we will read about in great detail in Chapter 16. In some mice, these com­bination drug therapies held already-formed tumors to a small size for as long as two months.

Significantly, these combinations of drugs had virtually no toxic effects on nor­mal pancreatic tissue adjacent to neoplastic islets, confirming that recently formed capillaries within a tumor are far more vulnerable to disruption than the well-established vessels v.rithin normal tissues. In addition, it seemed that PDGF played a major role in initially attracting pericytes to the capillary tubes formed by endothelial cells, but in normal tissues, continued PDGF signaling was not required to maintain this association. These observations indicate that the most effective ways of inhibiting angiogenesis and thus blocking tumor progression are likely to depend on targeting several of the cell types that construct the tumor-associated vasculature.

The results of these targeted anti-angiogenesis treatments illustrate the highly promising nature of this general therapeutic strategy. In fact, this type of ther­apy may have been in use for many years: unbeknownst to the community of clinical oncologists, much of the efficacy of some widely used, traditional anti­tumor therapies may also derive from their effects on tumor-associated microvasculature. One striking example of this, which sheds light on how radi­ation therapy succeeds in destroying certain tumors, has come from studies of genetically altered mice that lack the genes encoding either the Bax protein or the acid sphingomyelinase (asmase) enzyme. Both of these proteins are impor­tant pro-apoptotic regulators in a variety of cell types including, importantly, endothelial cells. Consequently, the endothelial cells from these genetically altered mice are far more resistant to toxic agents, including X-rays, than their wild -type counterparts.

In the experiments shown here, mouse tumor cells-from either a fibrosarcoma or a melanoma cell line-were introduced into both wild-type and asmase-I­

mice. After tumors had grown to a substantial size, these animals were exposed to a dose of 15 grays (Gy) of X-rays (Figure 13.48A and B). This dose of radiation usually causes a significant reduction in the tumor burden, and indeed it suc­ceeded in doing so in the wild-type mice. However, the identically sized tumors grown in the asmase- I - host mice responded quite differently, in that their growth continued unabated. When, as a further experiment, the bone marrow of asmase+ l +mice was replaced with transplanted mutant bone marrow cells, the tumors became highly resistant to X-ray-induced killing (Figure 13.48C) .

These experiments demonstrate that the radiosensitivity of these tumors was not intrinsic to the tumor cells themselves. Instead, it was governed by host cells, specifically by cells recruited into engrafted tumors from the host bone marrow. These recruited host cells were indeed endothelial cells, as evidenced by the fact that (1) the apoptosis of the endothelial cells in tumor fragments irra­diated in vitro directly paralleled and predicted the in vivo behavior of the tumors, and (2) the only cells that showed significant apoptosis in the tumors shortly after irradiation were associated with capillaries (Figure 13.48B).

Observations like these indicate that anti-angiogenesis therapy has played a far greater role in a conventional anti-tumor radiotherapy than anyone dared to imagine; similarly, clinical responses to certain types of conventional chemo­therapy may also be strongly influenced by the sensitivity of the tumor-associ­ated microvasculature to these agents. This suggests that in the future, treat­ments with many anti-cancer chemotherapeutics may be optimized by gauging their effects on the tumor-associated microvessels rather than on the tumor cells themselves. 580

Page 55: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

13.11 Synopsis and prospects

Metazoan tissues are organized as condominiums of various cell types that are continuously communicating with one another. To the developmental biologist, the need for this organizational plan is self-evident: only through such interac­tions can the proper numbers and locations of each of these cell types be ensured. These heterotypic interactions continue to operate after embryogene­sis has been completed in order to support the maintenance and repair of already-formed tissues.

As we have learned in this chapter, this organizational plan confers another, quite distinct benefit on the organism. By making its cells so interdependent, none is easily able to extricate itself from its complex web of interactions and go off on its own. Interdependence imposes the regimentation that wards off the chaos of neoplasia.

(A) (8)

500

400

E .s 300 (]J

E :::J

0 > o 200 E :::J +-'

100

0

(C)

200

no 150 E .s

(]J

E :::J

1000 > 0 E :::J +-'

50

o

II 10 15 20 25 30

days after tumor implantation

_ asmase+l+(10)

_ asmase-I-(10)

Oil 10 15 20 25 30

days after tumor implantation

donor recipient _ asmase+1+ (10) asmase+l+ (10) _ asmase-I- (10) asmase+1+ (10)

Synopsis and prospects

Figure 13.48 Survival of wild-type and asmase-negative tumor-bearing mice The responses of tumors to radiotherapy may often be determined by the radiosensitivity of the endothelial cells that form their vasculature . Mice were bred to be either w ild type or homozygous null for the gene encoding the pro-apoptotic enzyme acid sphingomyelinase (asmase). When mouse fibrosarcoma cells were implanted in the wild-type and the asmase-l- hosts, the tumors grew twice as fast in the mutant mice as in the w ild­type mice, suggesting that a host factor, such as recruited endothelial cells, was governing the rate of tumor growth . (A) When these tumors were given a therapeutic dose (15 Gy) of radiation (arrow), the tumors in wild-type mice regressed (red filled circles) and, after a while, began to grow again. In contrast, the tumors in the asmase-I- hosts (blue filled circles) continued to grow unabated. (B) When these tumors were examined microscopically follow ing irradiation, the endothelial cells in tumors carried by w ild-type mice (below), identified by an endothelial cell-specific immunostain, were apoptotic, as indicated by the TUNEL staining. (brown spots), while the endothelial cells in tumors borne by the asmase-I- hosts showed no signs of apoptosis (above) (C) When the bone marrow of wild-type hosts was replaced through transplantation of either wild­type or asmase-l- donor marrow cells, the tumors implanted in mouse hosts w ith engrafted mutant marrow (blue curve) continued to grow following 15 Gy of radiation (arrow), while tumors implanted in mouse hosts eng rafted with wild-type marrow were stopped by the radiation (red curve). This demonstrated that the radiosensitivity of the tumor was determined by cells of host bone marrow Origin, not by th e tumor cells themselves. (From M. Garcia­Barros, F Paris, C. Cordon-Carda et aI., Science 300 1155-11 59, 2003) 581

Page 56: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

Sidebar 13.10 Kaposi's sarcoma cells hold the record for the number of documented het­erotypic signals they receive Kaposi's sarcoma (KS) seems to derive from the cells related to those generating the endothe­liallinings of lymphatic vessels. The causal agent of KS-human herpesvirus-8 (HHV-H)-flour­ishes when immune defenses have been compromised; ex­plaining the high frequency of this tumor in AIDS patients (see Sidebar 3.10). Gene expression analyses from several laborato­ries have documented the increased expression of mRNAs encoding 15 distinct receptors in KS cells; the respective lig­ands for these receptors appear not to be produced by the KS cells, This suggests that the ele­vated expression of these recep­tors allows KS cells to take advantage of ligands that are supplied, via paracrine signal­ing, by other nearby cell types. The list of receptors includes those binding the following lig­ands: TNF-a, INF-y, oncostatin M, bFGF, VEGF-C and -D, in­sulin, thromboxane A2, IL-2, IL­6, IL-I0, IL-13, thrombospo­ndin, VEGF 165, CXCLl2, and CSF-I. (The dependence of KS cells on all of these signals has not yet been demonstrated experimentally.) This complex­ity hints that other histologically complex tumors, like KS, may rely on multiple heterotypic sig­nals to sustain their prolifera­tion and survival.

Earlier, we viewed multi-step cancer progression as the hurdling of successive barriers placed in the path of developing cancer cells (Chapter 11). Each suc­cessfully completed step, whether achieved by genetic or epigenetic changes, removes one of these obstacles and places the cancer cell incrementally closer to full -fledged malignancy. Now we can conceptualize multi-step tumor pro­gression in a quite different way: as pre-malignant cells evolve toward malig­nancy, they progressively sever their ties with their neighbors and their depend­ence on neighborly support.

Perhaps the biggest surprise is how dependent most cancer cells remain on stro­mal support in spite of having completed multiple steps of tumor progression. The epithelial cells residing in many carcinomas continue to rely on many ofthe physiologic signals that sustained their precursors in normal tissues (see, for example, Sidebar 13.10). This conservatism is evident under the microscope. Thus, a well-trained pathologist can recognize the origins of perhaps 95% of the tumor samples viewed under the microscope, because most of the heterotypic interactions that govern normal morphology are still operative in the great majority of cancers.

The remaining approximately 5% of tumors present a challenge, because they are anaplastic and therefore have lost most of the histologic traits that make it possible to identify their tissue of origin. The cells in these anaplastic tumors have shed most forms of dependence that tied their precursors to normal neigh­boring cells. However, most anaplastic tumor cells have not progressed all the way to total independence, because they still assemble to form solid tumors. The ultimate independence is achieved only by the cells in those cancers that have advanced so far that they can grow as pleural effusions or ascites (see Sidebar 25 0 ) and therefore have no direct contact with supporting cells and, apparently, with an extracellular matrix (ECM).

Even without detailed knowledge of heterotypic interactions, the dependence of most carcinoma cells on at least one form of stromal support could have been predicted from the known physiology of mammalian tissues: virtually all of them depend on a functional blood supply. Less predictable was the mechanism by which tumors acquire their vasculature. Rather than invading normal tissue and expropriating existing capillary beds, tumors actively recruit endothelial cells that proceed to construct capillaries and larger vessels within the tumor masses.

The sources of these endothelial cells could also not be deduced from first prin­ciples. The proliferation of endothelial cells in neighboring tissues seems to be the main mechanism for acquiring neovasculature soon after the angiogenic switch has been tripped. However, in many tumors, the bulk of the vessels made thereafter seem to be constructed by cells that originate in the bone marrow and then differentiate in the tumor stroma into functional endothelial cells. The contribution of these circulating endothelial precursor cells (EPCs) to the tumor-associated vasculature seems to vary from one tumor to another, but clearly represents a significant source in many neoplasias.

Major aspects of angiogenesis are still poorly understood, even paradoxical. For example, it seems apparent that once tumors become angiogenic, they can launch into a prolonged phase of growth and expansion, and that tumors that are more angiogenic can grow even more rapidly than those that are less able to attract new vasculature. Indeed, as we read, measurements of microvessel den­sity-the number of capillaries per microscope field-correlate quite well ..vith the likelihood that a primary tumor, such as a breast cancer, will progress to a highly malignant endpoint.

The paradox comes from the frequent observations that patients bearing highly hypoxic tumors also confront a poor prognosis. This makes no sense, given that hypoxia starves tumor cells and often leads to their death through apoptosis 582

Page 57: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Synopsis and prospect

and, on a larger scale, to extensive necrotic regions within tumors. The paradox may one day be resolved by invoking the actions of the HIF-1 transcription fac­tor, which becomes activated in hypoxic cells (Section 7.12) and induces pro­duction of a large number of other proteins besides VEGF. Indeed, a high level of HIF-l expression is also an indicator of poor clinical prognosis. Included among the genes activated by HIF-1 are those specifying PDGF, TGF-a, TGF-~, and sev­eral matrix metalloproteinases (MMPs) that are responsible for remodeling the extracellular matrix (ECM). As we have learned, several of these secreted pro­teins act as potent mitogens that drive the proliferation of both epithelial cells and their stromal neighbors.

An additional HIF-l-induced gene encodes the Met protein, which functions as the receptor for hepatocyte growth factor (HGF), also known as scatter factor (SF). Once the Met receptor is activated by binding its HGF ligand, it activates a diverse set of responses in epithelial cells, including the epithelial-mesenchy­mal transition (EMT)' increased motility, invasiveness, and proliferation. HGF seems to be widely available in many human tumors of both epithelial and mes­enchymal origin. Consequently, HIF-l-mediated increases in Met expression can sensitize tumor cells to HGF molecules that are present in their surround­ings, such as the HGF that has been released by stromal cells. These diverse products of HIF-l action may therefore help explain the poor prognosis attached to hypoxic tumors, which thrive in spite of great adversity and actually turn out to be more aggressive than their well-oxygenated counterparts.

The recently discovered dynamic interactions between tumors and the bone marrow have been a surprise to most cancer researchers. In the absence of metastasis, most tumors have traditionally been considered to be localized dis­eases confined to one or another corner of the body. But as we learn more about the cells of the stromal compartment, the more we come to realize that even localized tumors extend their reach far and wide throughout the body in order to recruit the cells that they need to support their own survival and proliferation programs. Besides endothelial precursor celis (EPCs), cited above, carcinomas recruit mast cells and monocytes from the marrow, the latter differentiating on­site into macrophages. Even some stromal fibroblasts and myofibroblasts may have bone marrow origins, since they may often arise from circulating precur­sor celis termed fibrocytes.

The physiology of the carcinomas, on which we have focused in this chapter, is far more complex than bidirectional exchanges of signals between epithelium and stroma. The latter is composed of more than half a dozen distinct cell types (fibroblasts, myofibroblasts, endothelial celis, pericytes, smooth muscle cells, mast cells, macrophages, lymphocytes, and, in some tissues, adipocytes), each of which signals to the neoplastic epithelial cells as well as to the other compo­nents of the stromal compartment. Physicists have struggled, so far unsuccess­fully, .'lith solutions to the three-body problem. Here, we confront a world of as many as eight or more distinct cell types, each of which is sending a complex mixture of signals to other types of cells within tumors. We have only begun to scratch the surface of the complex epithelial-stromal interactions operating in neoplastic tissues and in their normal counterparts.

For more than half a century, the mindset of oncologists has been focused on eradicating the bulk of the cancer cells within solid tumors, >vith the hope of achieving the impossible-cures of common carcinomas that have traditionally been incurable. As we learned, first in Chapter 11 and now in this one, this focus needs to be radically redirected. First, the true targets of anti-cancer therapies can no longer be the bulk of neoplastic celis in a tumor, because they have lim­ited self-renewal capacity; instead, we are likely to find that truly durable cures can come only from eradicating the still-elusive tumor stem cells that hide out, here and there, throughout tumor masses and represent their engines of self­renewal. Second, many of the useful anti-cancer therapies to be developed in the future will not come from targeting the cancer cells themselves. Instead, it 583

Page 58: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Q

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

ENDOTHELIAL CELL

Inhibitors of VEGF, FGF, etc., signaling, e.g., anti-VEGF and anti ­VEGF-R antibodies, small-molecule VEGF-R inhibitors, VEGF-Trap, Ang2lTie2 blocking antibodies. Endogenous angiogenesis inhibitors, e.g., endostatin, tumstatin . Inhibitors of EPC recruitment.

PERICYTE

Inhibitors of PDGF signaling, e.g., anti ­PDGF antibodies, PDGF-R inhibitors. Inhibitors of Ang­1lTie2 signaling.

FIBROBLAST

Inhibitors of HGF or its receptor c-Met, inhibitors of CXCL121 SDF-1, PDGF/PDGF-R, of fibroblast activation protein, e.g., sibrotuzumab.

Figure 13,49 Heterotypic interactions as targets for therapeutic intervention As described in this chapter, cancer cells are dependent on the nearby stromal microenvi ronment for a variety of cell-physiologic supports. This dependence on heterotypic interactions has inspired a new type of cancer therapy, some of w hich has been featured in this chapter. Instead of focusing on the intracellular signaling defects within cancer cells, this new type of therapy is directed toward interrupting heterotypic signaling, thereby depriving cancer cells of essential stromal support. This scheme indicates some of the anti-tumor therapies that are being developed or under con sideration. (From J.A. Joyce, Cancer CeI/7 :513-520, 2005)

BASEMENT MEMBRANE EXTRACELLULAR MATRIX

Inhibitors of matrix turnover, e.g. suramin, dalteparin and matrix­degrading enzymes, e.g., proteases (cathepsins, MMPs, uPA etc.), endoglycosidases (e.g., heparanase). Inhibitors of ECM contact, e.g., integrin av~3, av~5, a5~1, or a6~4 antibodies.

NEUTROPHIL MACROPHAGE MAST CELL

Anti-inflammatory inhibitors, e.g., cytokine and chemokine inhibitors, NF-ICB, IKK, TNF-a inhibitors.

Anti-lymphatic targeting: inhibitors of VEGF-C, VEGF-D, VEGF-R3, or PDGF/PDGF-R.

may often be far more profitable to attack the cells that provide them with vital physiologic support; by undermining the elaborate stromal support network on which most cancer cells depend, truly dramatic regressions of solid tumors may one day be achieved (Figure 13.49).

The insight that tumors are "wounds that do not heal" extends and echoes our earlier discussions (Section 11.15), in which the critical role of chronic inflamma­tion in promoting tumor formation was discussed in great depth. Inflammation and wound healing are intertwined processes, and the mechanisms of inflamma­tion-driven tumor promotion, which lead to the initial formation of cancers, are extended and elaborated by the chronic wound healing that seems to best char­acterize the biology of the stroma of well-established tumors.

Here, too, there has been another surprise. Inflammatory cells, notably macro phages, have traditionally been depicted as the front-line soldiers of the immune response that deal effectively with infectious agents, such as bacteria, by phagocytosing them and help guide the long-term immune response through antigen presentation, as we will see later in Chapter 15. Now we learn that macro phages are also critical agents in producing mitogenic growth factors, lib­erating angiogenic factors, and remodeling the extracellular matrix (ECM); the latter process is also critical for tumor invasion and metastasis (Chapter 14).

So, the traditional job assignments of various differentiated cell types are being extended and blurred. Cells of the immune system, which are purportedly dis­patched to protect us from infection and even cancer, are often active collabora­tors in tumor development. And the deletion of one or another cell type from the immune system, achieved in mice through germ-line re-engineering, often cre­ates a host organism that is, paradoxically, less able to support tumorigenesis.

Normal, quite elaborate morphogenetic programs, such as wound healing and the epithelial-mesenchymal transition (EMT), are likely to explain how carci­noma cells are clever enough to acquire the complex cell phenotypes that they 584

Page 59: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Key concepts

need in order to execute the later stages of malignant progression. In hindsight, this notion is not so surprising, since the more we learn about cancer cells, the more we realize how opportunistic they are in co-opting and exploiting normal biological processes in order to further their own ends. This leaves us with the last question of this chapter: Have we begun to truly understand the mechanis­tic complexity of heterotypic interactions, or is there an entire universe of undis­covered signaling pathways and behavioral programs lurking within tumors, waiting, like intergalactic dark matter, to surprise us?

Key concepts • Tumors are complex tissues that depend on intercommunication between

various cell types. Indeed, most tumors are as complex histologically as the normal tissues in which they arise.

• In carcinomas, these cell types can be separated into the neoplastic epithelial celis and recruited stromal cells, which include fibroblasts, myofibroblasts, and macro phages, as well as the various cell types that participate in the con­struction of the tumor-associated vasculature, specifically endothelial cells, pericytes, and smooth muscle cells.

• Most carcinomas depend absolutely on recruited stromal cells for various types of physiologic support. This dependence is lost only in the small subset of tumors that progress to an extremely malignant state, notably the tumor cells growing in ascites and pleural fluid.

• At the biochemical level, this interdependence is manifested by the exchange of various types of mitogenic and trophic factors. For example, carcinoma cells may release PDGF to recruit and activate stromal cells, while the latter respond by releasing IGFs that sustain the carcinoma cells.

• The formation of tumor-associated vasculature, formed by the process of neoangiogenesis, is a critical, rate-limiting determinant of the growth of all tumors larger in size than approximately 0.2 mm.

• In the case of carcinomas, the acquisition of tumor-associated stroma closely resembles the process of healing in wounded epithelial tissues. The genesis of stroma therefore relies on the same gene expression programs that are acti­vated during wound healing.

• As tumor progression proceeds, the fibroblast-rich stroma is increasingly replaced by myofibroblasts, which eventually generate collagen-rich, desmo­plastic stroma.

• The recruitment of the cells that participate directly in the construction of the neovasculature of tumors involves the release of factors, such as VEGF, by both the tumor celis and inflammatory celis, notably macrophages.

• Neoangiogenesis represents an attractive target for the development of novel anti-cancer agents, in that the targeted cells are the various normal stromal cell types participating in angiogenesis rather than the ever-changing cancer cells.

Thought questions

1. \I\lhat diverse lines of evidence prove directly that most car­ 4. Which lines of evidence persuade you that the generation of cinoma cells depend on stromal cell types for various types tumor-associated stroma depends on the same biological of physiologic support? programs that are activated during wound healing?

2. How might anti-angiogenesis therapies improve (in some 5. Which biological forces cause the tumor-associated vascu­cases) or neutralize (in other cases) the efficacy of conven­ lature to be defective in so many respects? tional chemotherapeutic agents? 6. Which types of anti-cancer therapeutic agents would you

3. How might macrophages facilitate or antagonize tumorige­ deploy in order to encourage the collapse of an established nesis? tumor by depriving it of vasculature support? 585

Page 60: The Biology of Cancer (2007) - Robert a. Weinberg - Ch. 13

Chapter 13: Dialogue Replaces Monologue: Heterotypic Interactions and the Biology of Angiogenesis

7. What biochemical strategies can tumor cells use to lessen their dependence on stromal support?

8. Can you cite examples of how oncoproteins perturb the interactions between carcinoma cells and the nearby tumor-associated stroma?

Additional reading

Balkwill F & Mantovani A (2001) Inflammation and cancer back to Virchow 7 Lancet 357, 539-545 .

Bergers G & Benjamin LE (2003) Tumorigenesis and the angiogenic switch. Nat. Rev. Cancer 3,401-410 .

Bhowmick NA, Neilson EG & Moses HL (2004) Stromal fibroblasts in cancer initiation and progression. Nature 432, 332-337.

Bingle L, Brow n NJ & Lew is CE (2002) The role of tumour-associated macrophages in tumour progression: implications for new anticancer therapies. 1. Pathol. 196, 254-265.

Bissell MJ & Radisky D (2001) Putting tumours in context. Nat. Rev. Cancer 1,46-54.

Bouck N, Stellmach V & Hsu sc (1996) How tumors become angiogeni c. Adv. Cancer Res. 69, 135-174.

Carmeliet P (2000) Mechanisms of angiogenesis and arteriogenesis. Nat. Med 6, 389-395 .

Conway EM & Carmel iet P (2003) Cardiovascular biology: signalling silenced. Nature 425, 139-141.

Coussens LM &Werb Z (2002) Inflammation and cancer. Nature 420,860-867.

Doljanski F (2004) The sculpturing role of fibroblast-like cells in morphogenesis. Perspect. Bioi. Med. 47, 339-356.

Dvorak HF (1986) Tumors wounds that do not heal . N. Engl. 1. Med. 315,1650-1689.

Dvorak HF (2002) Vascular permeability factor/vascular endothelial growth factor: a critical cytokine in tumor angiogenesis and a potential target for diagnosis and therapy. 1. Clin. Onco/. 20, 4368-4380.

Dvorak HF (2003) How tumors make bad blood vessels and stroma. Am. 1. Pathol. 162,1747-1757.

Elenbaas B & Weinberg RA (2001) Heterotypic signaling between epithe lial tumor cells and fibroblasts in ca rcinoma formation. Exp. Cell Res. 264, 169-184.

Ferrara N (2002) VEGF and the quest for tumour angiogenesis factors. Nat. Rev. Cancer 2, 795-803.

Ferrara N, Gerber HP & LeCouter J (2003) The biology of VEGF and its receptors . Nat. Med. 9, 669-676.

Folkman J & Kalluri R (2003) Tumor angiogenesis. In Cancer Medicine, 6th ed (DW Kufe, RE Pollock, RR Weichselbaum et al. eds), pp. 161-194. Hamilton, Ontario: BC Decker.

Gold LI (1999) The role for transforming growth factor-beta (TGF­beta) in human cancer. Crit. Rev. Oneal. 10, 303-360

Hanahan D & Folkman J (1996) Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell 86, 353-364.

Harris AL (2002) Hypoxia-a key regulatory factor in tumour grow th . Nat. Rev. Cancer 2, 38-47.

Jain RK (1994) Barriers to drug delivery in solid tumors . Sci. Am. 271(1), 58-65.

Jain RK (2003) Molecular regulation of vessel maturation . Nat. Med 9,685-693.

Jain RK (2005) Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science 307, 58-62.

Jain RK & Booth MF (2003) What brings pericytes to tumor vessels? 586 1. C/in. Invest. 112, 1134-1136.

9. How might you determine what proportion of endothelial cells in the vasculature of a tumor derive from the expan­sion of adjacent vasculature and what proportion of these cells arise from circulating endothelial precursor cells?

Kalluri R (2003) Basement membranes: structure, assembly and role in tumour angiogenesis. Nat. Rev. Cancer 3, 422-433.

Kalluri R & Neilson EG (2003) Epithelial-mesenchymal transition and its implications for fibrosis. 1. Clin. Invest. 112, 1776-1784.

Kerbel RS (2000) Tumor angiogenesis: past, present and the near future . Carcinogenesis 21,505-515 .

Kerbel R & Folkman J (2002) Clinical translation of angiogenesis inhibitors. Nat. Rev. Cancer 2, 727-739.

Leek RD & Harris AL (2002) Tumor-associated macrophages in breast cancer. 1. Mammary Gland Bioi. Neoplasia 7, 177-189.

Littlepage LE, Egeblad M and Werb Z (2005) Evolution of cancer and stromal cellular responses . Cancer Cell 7,499-500.

McCawley LJ and Matrisian LM (2001) Matrix metalloproteinases: they' re not Just for the matrix anymore! Curr. Opin. Cell Bioi. 13, 534- 540.

McDonald DM & Baluk P. (2002) Significance of blood vessel leakiness in cancer. Cancer Res. 62, 5381-5385.

McDonald DM & Foss AJ (2001 ) Endothelial cells of tumor vessels: abnormal but not absent. Cancer Metastasis Rev. 19, 109-120.

Mueller MM & Fusenig N (2004) Friends or foes-bipolar effects of the tumour stroma in cancer. Nat. Rev. Cancer 4, 839-849.

Nyberg P, Xie L & Kalluri R (2005) Endogenous inhibitors of angiogenesis. Cancer Res. 65, 3967-3979.

Pepper MS (2001) Lymphangiogenesis and tumor metastasis: myth or reality! Clin. Cancer Res. 7,462--468.

Rabbani SY, Heissig B, Hattori K & Rafii S (2003) Molecular pathways regulating mobilization of marrow-derived stem cells for tissue revascularization. Trends Mol. Med. 9, 109-117.

Radisky D, Hagios C & Bissell MJ (2001) Tumors are unique organs defined by abnormal signaling and context. Semin. Cancer Bioi. 11,87-95.

Rafii S, Lyden D, Benezra R et al. (2002) Vascular and hematopoietic stem cells: novel targets for anti-angiogenesis therapy! Nat. Rev. Cancer 2, 826-835.

Savagner P (200 1) Leaving the neighborhood: molecular mechanisms involved during epithelial-mesenchymal transition. BioEssays 23, 912-923 .

Serini G & Gabbiani G (1999) Mechanisms of myofibroblast activity and phenotypic modulation. Exp. Cell Res. 250, 273-283.

Sleeman JP (2000) The lymph node as a bridgehead in the metastatic dissemination of tumors Recent Results Cancer Res. 157, 55-81.

Thiery JP (2003) Epithelial -mesenchymal transitions in development and pathologies . Curr. Opin. Cell. BioI. 15, 740-746.

Tlsty T (2001) Stromal cells can contribute oncogenic signals. Semin. Cancer BioI. 11, 97-104.

Walker RA (2001) The complexities of breast cancer desmoplasia . Breast Cancer Res. 3, 143-145.

W iseman BS & Werb Z (2002) Stromal effects on mammary gland development and breast cancer. Science 296, 1046-1049.