Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Feb 1.
Published in final edited form as: Semin Cancer Biol. 2011 Dec 24;22(1):41–49. doi: 10.1016/j.semcancer.2011.12.008

Targeting stroma to treat cancers

Boris Engels a, Donald A Rowley a, Hans Schreiber a
PMCID: PMC3321387  NIHMSID: NIHMS346923  PMID: 22212863

Abstract

All cancers depend on stroma for support of growth. Leukemias, solid tumors, cancer cells causing effusions, metastases as well as micro-disseminated cancer cells release factors that stimulate stromal cells, which in turn produce ligands that stimulate cancer cells. Therefore, elimination of stromal support by destroying the stromal cells or by inhibiting feedback stimulation of cancer growth is in the focus of many evolving therapies. A stringent evaluation of the efficacy of stromal targeting requires testing in animal models. Most current studies emphasize the successes of stromal targeting rather than deciphering its limitations. Here we show that many of the stromal targeting approaches, while often reducing tumor growth rates, are rarely curative. Therefore, we will also discuss conditions where stromal targeting can eradicate large established tumors. Finally, we will examine still unanswered questions of this promising and exciting area of cancer research.

Keywords: tumor stroma, tumor promotion, cancer therapy, relapse, eradication

1. Abuses of terminology

Without adherence to proper terminology, scientific results are easily misread. There is an enormous thirst for high impact factors of journals and for translational bench-to bedside protocols by agencies and by the public. This situation fosters highly misleading titles that have become fashionable to “increase visibility”, i.e., inflate the impact of papers. For example: “Eradication” does not mean just a reduction of tumor growth, it should mean destroying a tumor completely like tearing a tree out with its roots so it cannot re-grow. Experimental microscopic subcutaneous cancers treated on day 6 or day 9 are not “established cancers”. “Tumor” means to lay and professional people a swelling, which is what the word means also in Latin; therefore, you cannot treat a “tumor-bearing mouse” before a tumor becomes visible or palpable. There also is a very strong bias to use the word “therapeutic” for treating small lesions of only a few mm in diameter, which is of unknown relevance. Clinically most tumors are not detected until they are 0.5 to 1 cm in diameter and contain 109 cells [1]. Individuals, mice or people, can be treated, e.g. by vaccination, to prevent cancer, but “therapy” implies treatment of existing disease. Therefore, humans are treated when they have diagnosable cancer, but it is inappropriate in experimental models to call therapy as being effective few days or so after cancer cell inoculation when the tumors are microscopic. Oncogene-transgenic cancer models in animals are widely referred to as “spontaneous” implying these cancers closely mimic human cancers that are mostly sporadic. However, the term “spontaneous cancer” is clearly reserved for cancers arising in the absence of any experimental manipulation [2]. These kinds of abuses mean that the words eventually lose their original meaning, and that loss is no longer even noticed. Abuse of terminology does not serve either scientists or patients well.

2. Tumor promoting stroma

2.1 Tumor formation depends on reciprocal induction between cancer cells and stroma

Rudolf Virchow believed that compression of the growing cancer cells induced a structural fibroblastic framework in which the cancer cells grew. He thought that cancer cells and stroma both developed from the same primitive precursors. This concept changed with Paul Ehrlich stating clearly that the host provided the stroma of solid tumors [3]. But it was Borst in 1924 [4], who was the first to clearly point out the essential mutual relationship between cancer cells and tumor stroma by stating: “With regards to the question of whether the epithelium or the connective tissue has the leading role in carcinogenesis, we think that asking “either/or” is bad.” In other words, stroma of tumors is dependent on the presence of cancer cells, and cancer cells are dependent on stromal cells (reviewed in [5]). Thus, cancer cells release factors that attract stromal precursor cells, and stromal cells in turn produce factors that support cancer cell growth. Once the cancer cells are destroyed, the stroma that supported the tumor will vanish within weeks unless scars have formed during the regression process. Unfortunately, it is usually omitted that the reverse is often not true: destruction of stroma does not necessarily lead to the death of all cancer cells (see section 8).

2.2 Tumor promoting anti-apoptotic stroma

Lack of tumor stroma drastically reduces tumorigenicity [6-8]. Inoculated cancer cells embedded in tumor stroma are 10 to 100-fold more tumorigenic than stroma-free suspensions of cancer cells [6, 7]. It had long been known that certain cancers would only grow in mice when transplanted as tumor fragments. This erroneously was thought to be due to more cancer cells being inoculated with fragments [9]. Later analyses revealed fragment inoculations contained fewer cancer cells than injected cell suspensions [6, 7]. Another erroneous explanation was that the stroma of fragments provided a physical barrier preventing cancer cells from migrating to draining lymph nodes and priming a protective T cell response [7, 10]. It is more likely, however, that cancer cells simply remain more viable when embedded in tumor stroma (by preventing anoikis [11]) and therefore, less antigen is available for release and T cell stimulation. In any case, as long as cancer cells express sufficient levels of antigen, professional antigen presenting cells in the tumor stroma pick up the antigen and travel to the draining lymph nodes where they present the antigen to naïve T cells [12].

Stroma promotes tumor growth by two distinct mechanisms: (i) stimulation of vasculature and connective tissue by inflammation-induced attraction of the various myeloid and lymphoid derived cell types and (ii) the suppression of an immune response. Vasculature provides oxygenation and nutrients; connective tissue and the extracellular matrix (ECM) are needed for adherence, structure, and the binding/release of cytokines, chemokines and growth factors that signal as part of a paracrine loop to the cancer cells [13-16]. The tight link between inflammation, leukocyte infiltration and cancer is being studied intensively. It is the topic of many reviews and therefore is not covered in depth here [17-21]. In short, it has been observed that tumor infiltrating T cells and other leukocytes secrete cytokines and growth factors that activate the stroma and accelerate tumor growth [14, 15, 22, 23]. Angiogenesis is a fundamental necessity for tumor growth. Myeloid cells including mast cells, eosinophils, and tumor-associated macrophages (TAM) have been shown to promote tumor angiogenesis (reviewed in [24]). However, also cancer cells themselves and tumor-associated fibroblasts (TAF) can produce pro-angiogenic as well as growth stimulatory factors such as vascular endothelial growth factor (VEGF) (reviewed in [25]).

2.3 Cancers, solid or leukemic, depend on paracrine stimulatory loops involving stroma

For growth, leukemias, solid tumors, malignant effusions, metastases as well as micro-disseminated cancer cells depend on reciprocal cancer cell-stroma interactions. This loop seems to depend on oncogenic mutations in the cancer cells that cause them to release molecules, which induce non-malignant stromal cells of the host to make factors that promote cancer cell growth [26]. An early example is the oncogenic fusion protein RET/PTC3 (RP3) in thyroid cancer [27, 28]. Stromal cells then produce other growth factors and alter the ECM so that both are conductive to growth and invasion. Two-stage carcinogenesis involves an initiating mutational event followed by repeated applications of a nonmutagenic pro-inflammatory promoter (typically the phorbol ester PMA (phorbol 12-myristate 13-acetate)) [29]. The critical step in cancer development is the acquisition by the cancer cell to release its own stimuli to the stroma, so the neoplastic cells become promoter-independent, and thus truly neoplastic.

2.4 Paracrine loop mismatch persists in “humanized” mice

Many paracrine factors have been identified. Interruption of paracrine loops by antibodies, receptor traps or drugs are important new therapeutic avenues to explore for more effective cancer therapy (see below). However, despite all what we have learned about chemokines, cytokines their receptors and signaling cascades, we do not have adequate procedures for culturing cancer cells from the most common human cancers: breast, prostate, lung, colon and pancreas. Eventually use of the right combination of factors and surfaces for growth will probably be found. The search may be aided by growing human cancer in immuno-incompetent mice and culturing the cancer cells in a three dimensional matrix ([30] and reviewed in [11, 31]). Only some human cancers can be grown in immuno-incompetent mice and can be serially transferred in these mice. But it is unclear at present whether, and for which human cancers “humanizing” immunodeficient mice with human stem cells must be used. Both human and fibroblastic precursors may be critical for success. Generating artificial extracellular matrixes may also mimic cancer-stroma interactions in humans. It must be remembered, however, that there are major hurdles: e.g. human and murine IFN-γ do not work on the reciprocal receptors [32] and thus human IFN-γ will not affect fibroblasts of the non-bone-marrow derived stroma in “humanized mice”. There are many papers claiming growth of human cancer xenografts, which is too often based on transient inflammatory reactions. Reports of “prevention” or “reduced growth” of such transplants after treatment has, therefore, questionable significance and reports on the effects of therapies targeting stroma may be misleading because of lack of interaction between human cancer cells and the non-BM derived stroma of mouse origin.

2.5 “Pseudo-Stroma” resulting from epithelial-to-mesenchymal transition (EMT) cannot replace host-derived stroma

The less differentiated, more fibroblastic appearance of carcinoma cells adjacent to the stroma is associated with the acquisition of genes such as snail, slug and vimentin, prominently expressed by mobile embryonic fibroblastic cells [33, 34]. While these changes may help cancer cells to acquire an increased metastatic and invasive potential, there is no evidence that they make cancer growth self-sufficient and can replace the need of cancer cells to establish a paracrine stimulatory loop with nonmalignant stroma [35].

3. The four components of tumor stroma

Stroma consists of connective tissue and extracellular matrix, vasculature and infiltrating hematopoietic cells. The latter comprise myeloid and lymphoid lineage cells, both derived from hematopoietic stem cells (HSC) in the bone marrow.

3.1 Connective tissue and vasculature

R.A. Willis in a careful survey of his own studies and published literature [36] subdivided tumor stroma into just two major components, connective tissue - which usually represents the bulk of stroma - and vasculature which is usually a smaller fraction. Fibroblasts are a prominent cell type in tumor stroma as well as in healing wounds and embryonic connective tissues. Unfortunately, we still lack reliable fibroblast-specific immunological markers for these cells despite repeated assertions to the contrary [37, 38]. For example, fibroblast–specific protein 1 (FSP-1), also called S100A4, has been found on various cell types including epithelial cancer cells and leukocytes [39-43]. Therefore, fibroblasts are still mostly defined by morphology and function [44-46]. Characteristically, they synthesize, secrete and modulate proteins of the fibrous ECM, particularly collagen [47-49]. ECM is a major reservoir for growth factors and cytokines [50]. Furthermore, the ECM is gaining increased attention due to its role in providing a scaffold but also growth signals, thereby preventing cell death due to anoikis (reviewed in [11, 31]). Interestingly, fibroblastic cells have been reported to present antigen and stimulate lymphocytes under certain experimental conditions [51, 52]. Stromal fibroblasts in cancers are metabolically active and the degree of activation correlates with aggressiveness of the cancer and it correlates inversely with survival of patients [53, 54]. The resting less active fibroblasts that are usually referred to as fibrocytes, are rarely observed in stroma of malignant tumors. Unfortunately the term fibrocyte has also recently been used to designate a cell population in the peripheral blood that can enter sites of inflammation and produce collagen [55]. Fibrocytes are resting cells surrounded by collagen fibers and extracellular matrix and are the prominent cell types in mature adult connective tissues and some malignancies, e.g. breast cancers [48, 49, 56-58].

3.2 Sources of progenitor cells

Mitoses in tumor stromal cells seem too sparse to explain the dense cellularity of stroma, as reported over a century ago [59]. Most likely, the cells in the stroma come from progenitor cells entering the site of tumor growth either via the blood circulation or from adjacent normal tissues. The relative contribution of these two sources is still hotly debated [60]. However, recent studies using non-myeloablative conditions have shown convincingly that precursors for tumor vasculature come primarily from local, i.e., adjacent normal tissues, not from the circulating, bone-marrow-derived precursor pool [61-64]. Indeed, recent and older studies demonstrate major progenitor reservoirs not only in the bone marrow but also in the perivascular spaces of all other organs [65-67]. Thus, tissues adjacent to the neoplastic lesion, wherever they may occur throughout the body, should have these progenitors. It is not entirely clear how these mesenchymal progenitor cells relate to the so-called pericytes [68] that are also thought to be pluripotent and that build sleeves around, and are essential for, endothelial cells to form capillaries. We are lacking conclusive evidence determining whether adjacent tissues are also a major source of fibroblasts in tumor stroma. In wound healing, however, there is clear evidence for adjacent normal tissues being the main source of fibroblast progenitors based on extensive and careful studies using parabiosis and labeling techniques [69-71]. Other studies suggest that precursors circulating in the blood can enter sites of inflammation, injury and tumors to generate stromal fibroblasts. The differentiation of blood leukocytes into fibroblasts has repeatedly been described in vitro and in vivo [72-74] and circulating mononuclear progenitor cells for TAFs have been isolated from venous blood in resected tumors of lung cancer patients [75]. These isolated cells differentiated into fibroblasts in xenografts of human lung cancer in NOD/SCID mice. Similar experiments have shown the differentiation of bone marrow-derived progenitors into TAFs [76, 77]. It remains unclear, however, whether the transferred progenitor cells are of hematopoietic or mesenchymal lineage. A study by LaRue and colleagues does suggest that TAFs and pericytes originate from HSCs [78]. So far, it remains unclear and controversial: (i) what percentage of TAF infiltrate is from bone marrow or surrounding tissue origin [79], (ii) what percentage of bone marrow-derived fibroblasts originate from hematopoietic versus mesenchymal lineage, and (iii) whether the origin of stromal fibroblast results in functionally different subtypes or not.

3.3 Differentiation of MDSC into tumor associated macrophages

Tumor-induced myeloid derived suppressor cells (MDSC) are the focus of a large body of literature (reviewed in [20, 80]). This heterogeneous population of largely immature cells of myeloid origin (Gr-1 and CD11b-positive in mice) suppresses adaptive and innate immunity, which is believed to be an important arm of tumor-induced immune-evasion. The suppression is achieved through several mechanisms including the production of nitric oxide and reactive oxygen species, the nitration of T cell receptors (TCR), arginine and cysteine deprivation, and the induction of regulatory T cells (Tregs). Interestingly, several reports show the same immune suppressive capacities of MDSC from naïve and tumor-bearing mice [81, 82] and splenic and tumor-derived MDSC were recently shown to exert their immunosuppressive activity through different mechanisms and onto different T cell subsets [83]. The same and older reports describe the rapid differentiation of MDSCs into suppressive TAMs in tumors [83-85]. In fact, the majority of hematopoietic cells in the tumor are macrophages [84, 86]. A recent review by Mandruzzato and colleagues argues that under certain conditions in vitro, depending on T cell numbers and their activation status, the suppression by MDSC may be overcome [87]. Taken together, the impact of the extensive literature on MDSCs, mainly studying circulating or splenic cells, on immune suppression within the tumor is unclear, and will need to be analyzed more carefully.

4. Can malignancy be “reversed” by normal and “induced” by cancerous stroma?

The question is old and contentious and not really answerable. The problems are that cancer and stromal cells can have abnormal karyotypes, which may or may not mean the cells are transformed to be malignant. Also, epithelial cells in tissues surrounding a cancer can contain cells that are morphologically abnormal but not certifiable as cancer cells either. In classic models of two-stage carcinogenesis pre-malignant lesions develop before cancer. These are dependent on promoting factors such as present in wound healing or chemicals that result in stromal stimulation (conditional neoplasms). To become promotion-independent, pre-malignant cells may acquire pathways that induce the stroma to provide a favorable environment [29, 88, 89]. Alternatively, the pre-malignant cells may loose their susceptibility to inhibitory effects in normal stroma that counteract cancer development [90-93]. Studies on mammary cancers show a strong cancer promoting effect of cancerous stroma through the expression of ECM-degrading metalloproteinase stromelysin-1 [94, 95]. However, the influences of and requirement for the stromal microenvironment to support an established cancer must be clearly distinguished from the often substantially different requirements of the stromal microenvironment during tumor initiation and development. For the induction of some epithelial cancers, exposure of stroma to carcinogen may be crucial [38, 96, 97] and result in mutations in stromal cells. But exposure of the stroma to non-carcinogenic, non-mutagenic agents such as PMA, usually seem to suffice to promote the development of epithelial cells carrying cancer-initiating mutations into an invasive cancer. Teratocarcinoma cells keep their malignant potential and do not genetically return to “normal” even when surrounded by non-cancerous stroma while completely normal cells are unlikely to develop true invasive serially transplantable tumors even when brought into the vicinity of cancer-induced stroma. Many factors such as diet, microbial flora, chemicals and wounding influence inflammation have strong potential impact on cancer induction and development by acting on the stromal microenvironment; while these factors have usually very little if any effect once cancer has developed. This also means that strategies to prevent or most likely cure cancer should be fundamentally different. Yet recent reviews do not make this distinction [11, 98].

5. Cancers induce local and systemic stromal changes

It is often assumed that the factors released by the cancer cells only act in the immediate vicinity of the cancer cells and this may often be true. However, it is also clear from many studies that cancer cells release factors such as granulocyte-, macrophage- and granulocyte macrophage colony-stimulating factor (G-CSF, M-CSF and GM-CSF) that have systemic effects. Thus in a way, the cancer may use distant organs such as the bone marrow to make its “stromal bed”. Whether “systemic stroma” can substitute for local stroma is unclear but in certain cancers such as leukemias the “systemic stroma” apparently suffices to cause progressive growth. For example, chronic lymphocytic leukemia cells release microvesicles that induce bone marrow stromal cells to release VEGF, a survival factor for chronic lymphocytic leukemia [99].

About 30% of patients with solid tumors have elevated numbers of granulocytes in their peripheral blood (>8000/μl); granulocytosis is also common in tumor-bearing mice [100-108]. Growth of autochthonous or transplanted murine tumors is accompanied by splenic enlargement (Schreiber, K. and Schreiber, H. unpublished results and [109]). Enlargement is mostly caused by increased myeloid hemopoiesis with macrophages and neutrophils usually accounting for more than half of the splenocytes [110]. G-CSF [111], GM-CSF and/or IL-6 [108, 112] released by cancer cells may be of particularly important in inducing these systemic effects.

Certain primary tumors can facilitate the outgrowth of a second tumor at a distant site [113, 114], even if the second tumor would never grow (or only grow very slowly) when injected alone. Using human tumor cell lines in an immune compromised host, it was recently found that an “instigating” tumor not only recruits murine stromal cells very effectively to support its own growth but also helps the incorporation of bone marrow-derived cells into the second “responding” tumor [115]. Osteopontin, which is over-expressed by many human and murine cancers [116, 117], and is required for recruitment and migration of neutrophils [118] appears to be required but not sufficient for instigating the growth of the second tumor at a distant site [115]. Systemic facilitation of stroma formation by mobilization of bone marrow-derived hemopoietic progenitors may also be the basis for the development of “metastatic niches”. These niches are thought to represent focal changes in the stroma of organs such as lungs caused by circulating factors released from cancer cells of the primary tumor; these changes are thought to provide a bed conducive for circulating cancer cells to settle down, vascularize and form a metastatic tumor [119, 120].

Furthermore, many studies have shown that growing tumors incorporate myeloid-derived Gr-1+ CD11b+ cells that are proangiogenic [121, 122] and cause increased TGF-β production in tumors [123]. Proteases released from stromal inflammatory cells such as neutrophils and macrophages are likely to be critical in locally activating latent TGF-β bound to ECM or to cells such as resting Tregs. Active TGF-β will then (i) orchestrate attraction and activation of fibroblasts to become SDF1-expressing myofibroblasts, which attract more stromal progenitors [124-126] and (ii) polarize naïve T cells towards Tregs thereby antagonizing adaptive immune responses [127-130]. Furthermore, neutrophil-derived elastase attenuated T lymphocyte transmigration by deactivating endothelial-bound SDF-1 [131]. In addition, tumor growth can induce systemic T cells to produce high levels of VEGF and matrix metalloproteinase-9 (MMP-9) and these T cells may help tumor growth at distant sites [23]. Another subset of T cells, which is induced by tumors, is the regulatory type [132]. These Tregs alter DC function [133] and convert naïve T cells to become Tregs themselves [127-130] thereby preventing the generation of a destructive T cell response. Despite the systemic effects of tumor growth described above, tumor-bearing individuals usually do not seem to be immunosuppressed systemically since they do not suffer from increased opportunistic infections except at very late stages [110].

6. Aggressive cancers induce stroma more effectively

During the continued growth human and experimental cancers become more malignant measured by rate of growth or capacity to metastasize. This is due to the generation of heritable variants, a process referred to as “tumor progression” [88, 134].

The most dramatic example may well be the progression of UV-induced regressor tumors to progressor variants: regressors are rejected in fully syngeneic naïve mice at any testable dose, while progressors grow. Since tumor stroma increases the viability and growth of cancer cells and has been shown to suppress the immune system, it is not surprising that cancer cell variants capable of inducing tumor stroma escape immune destruction. Evidence for this comes from studying heritable variants that escaped immune destruction in T cell-competent mice inoculated with regressor tumors. Regressor tumors can be induced in T cell-deficient mice [135-139]; when transplanted into naïve immunocompetent syngeneic mice, these tumors are regularly destroyed by a T cell response after transient growth. Occasionally, progressor variants escape this immune destruction, and most of these variants show more rapid and effective stromal induction and activation but no loss of antigen [14, 15]. It is likely that a systemic innate Gr-1+ response is critical. Systemic depletion of neutrophils and possibly other Gr-1+ cells by treatment with anti-Gr-1 antibodies not only reduces the growth rate of malignant cells in naïve T cell-deficient mice [15] but also leads to destruction of inoculated cancer cells in T cell competent mice [13]. While antigen loss can be found in progressor variants evolving from some regressors it is not mandatory. Instead, all progressor variants we have analyzed so far induce stroma much more effectively by more effective paracrine loops and most (but not all) retain the same immunogenicity as regressors [14, 140, 141].

7. Stroma as a target for cancer therapy

Because of the important role of tumor stroma in tumor progression and the suppression of the immune system, it has become a much-investigated target for cancer therapies. For targeting tumor stroma effectively, it is important to know what its components are and what the turnover of these components is (see 3). MDSCs, and more importantly TAMs, are attractive targets, as these cell types have been shown to both promote growth and suppress effective immunotherapy. Several targeting strategies are being explored, often to improve the usual failure of cancer vaccines. One of the approaches is to force differentiation of MDSC to mature myeloid cells such as dendritic cells, macrophages, or granulocytes. A treatment with all trans-retinoic acid (ATRA) and vaccination showed the hypothesized effect on MDSC and T cells and subsequently led to slower tumor growth [142]. A model of autochthonous pancreatic tumor shows regression in 30% of mice after activating macrophages to become tumoricidal [143]: the treatment with the CD40 agonist induced a T cell independent shrinkage of the tumors; it did not however lead to their eradication. In a model of IL-4 over-expression by transplanted tumors, the tumoricidal potency of neutrophils was demonstrated [144]. In an oncogene-transgenic mouse model of autochthonous mammary tumors and transplanted metastasis it was shown that a skewing of TAMs toward a M1 phenotype (in STAT6-/- mice) enhanced the host's anti-cancer immune response [82, 145]. The anti-tumor response was dependent on IFN-γ and CD8+ T cells and was accompanied by a decrease in circulating MDSC. To what extent this finding can be translated to therapy, for example by using STAT6 siRNA, remains to be shown. Nevertheless, the effect might be useful for post-surgical treatment of minimal residual disease after removal of the primary tumor.

Using the same model, it was demonstrated that MDCS were killed by T cells via Fas-FasL interactions [81]. FasL-deficient mice showed higher numbers of circulating MDCS and higher incidence of metastasis. However, as discussed above, MDSC are a small population of myeloid stromal cells compared to TAMs, which do express Fas but could not be killed through this receptor [81]. Therefore, the general therapeutic importance of this latter finding is not clear. Another strategy is to eliminate MDSCs with drugs such as gemcitabine [146]. The numbers of MDSCs were reduced and T and NK cell activity was augmented, which resulted in slight regression and then slower growth of large established tumors. Depletion of MDSCs with anti-Gr-1 antibody reduced but did not prevent growth of UV-induced cancer cells in athymic nude mice [14, 15]. In euthymic mice, T cells were not capable of controlling cancer cell inoculums of progressor variants [14] but treatment of the challenged mice with anti-Gr-1 antibody prevented outgrowth of the tumor [13], indicating a synergistic effect between T cells and the depletion of cancer-promoting stromal cells. Interestingly, the approach of combining MDSC depletion by cyclophosphamide and inducing inflammatory monocytes and neutrophils by IL-12 led to T cell dependent eradication of 70% of established MC38 colon carcinoma tumors [147]. In another model, an immune response against TAMs was induced. Legumain-specific T cells reduced metastasis after immunization and reduced death of mice, which were vaccinated shortly after cancer cell inoculation [148].

An alternative approach to differentiating and depleting MDSCs is to inhibit expansion or function of these cells. For example, the administration of MMP-9 inhibitors led to a delay in outgrowth of autochthonous mammary tumors by inhibiting MDSC expansion and macrophage infiltration into tumor stroma [149]. Also the inhibition of arginase 1 expression induced strengthened T cells responses and slowed tumor growth by inhibiting the suppressive effects exerted by MDSCs [150].

Macrophages and MDSC are not the only stromal cells that are targeted in attempts to cure cancer. In a transgenic mouse model fibroblast activation protein-α (FAP)-expressing cells were depleted in 12 day-old tumors, leading to a significant reduction of tumor growth when the cancer cells expressed a tumor-specific antigen (OVA257) [151]. Again, the effect on tumor growth was dependent on the adaptive immune system, as it was abrogated by ablation of IFN-γ and TNF-α and the effect was not existent in T and B cell-deficient mice. Similar results were obtained in two studies where T cells, which were induced by a FAP-DNA vaccine, targeted FAP-expressing cells [152, 153]. Another stromal target are pericytes. Targeting the pericytes-associated high molecular weight-melanoma associated antigen (HMW-MAA) with antibodies or T cells prevented tumor growth (reviewed in [154]).

Most studies targeting tumor stroma in one way or the other show the hypothesized effects on the stromal cells, such as their depletion and activation of the adaptive and/or innate immune systems. However, the effect on tumor growth is often marginal (slower growth), especially when treatment was initiated at later times. Most studies show the need for T cells for reduced tumor growth, thus the addition of these cells by adoptive transfer may lead to better outcomes (see below).

8. Cancer cells harbor clonal cancer-specific mutations but stroma does not

Stroma recruiting cancer variants arise because of the remarkable genetic instability of cancer cells. By contrast, stromal cells are non-malignant and generally genetically stable. Although chromosomal abnormalities occur [155-162] these changes are rare and do not show the clonality that is characteristic of cancer cells [163]. There are however studies that do describe the same leukemia-associated mutations to be present in a part of the myeloid stem cells and the malignant cells of patients [164]. Epigenetic changes in stromal cells can contribute to their tumor-promoting phenotype [165, 166], but when stromal cells are targeted for destruction by chemo-, radiation-, and/or immunotherapy, there is no escape of mutant stromal cells, and thus there is no loss of the targeted gene(s), antigen(s) or antigen-presenting MHC molecules [35].

9. Loading of stroma with cancer antigens

9.1 Cancer cell derived membrane vesicles as carriers of antigen

Modulation of tumor stroma is not only induced by soluble factors but also by small vesicles released by cancer cells. This includes pro-angiogenic effects by microvesicles released from ovarian cancer [167] and vesicles from B cell leukemia cells that activate bone-marrow stem cells [99]. Most cells release membrane vesicles of various sizes and origin such as microvesicles that have budded from the plasma membrane and exosomes, which are derived from intracellular compartments (reviewed in [168] and [169]). These vesicles contain hydrophilic compartments, and can therefore transport membrane bound proteins and also molecules of cytoplasmatic or luminal origin. Accordingly, microvesicles and exosomes can transfer transmembrane peptide-MHC molecules of cancer cells [170, 171] and antigenic material from the cytoplasm. Exosomes obtained from cancer cells cultured in vitro [172] or from patients' ascites [173] have been shown to contain tumor antigens capable of stimulating T cells. However, these vesicles not only transport antigen, but can also influence the immune system in other ways, for example inhibit [174] or stimulate immune responses [175, 176], the latter after exposing the cancer cells to stress conditions such as a heat shock [177, 178].

9.2 Stroma has no cancer specific-antigens except for those absorbed from adjacent cancer cells

Antigens originating from cancer cells can be cross-presented by stromal cells, thereby making stromal cells targets of cancer-specific cytotoxic T cells. Combined CD8+ T cell attack on cancer and stromal cells leads to tumor eradication [179, 180]. In a model where only stroma can be targeted by CD8+ T cells because on MHC restriction, growth arrest and long term “equilibrium” of tumor is observed [35]. In this model, the T cells cannot target the cancer cells directly, but the cross-presenting tumor stroma is destroyed [35, 179]. These tumors become completely necrotic inside while cancer cells continue to proliferate at the margins of the tumors. This is in accordance with the notion that targeting stroma alone usually does not eradicate tumors (see section 8). In another model, where the stroma cannot be targeted because of low antigen amounts expressed by the cancer, antigen loss variants take over and eventually kill the tumor-bearer [179].

Cross-presented antigen can also stimulate naïve T cells. APCs in the tumor pick up antigen, given that the levels are sufficient, travel to the draining lymph nodes and present the antigen to naïve T cells [12]. A different mechanism of T cell activation is the cross-presentation of antigen released by dead cancer cells and transported to the draining lymph nodes via lymphatic flow where it is taken up and presented by professional antigen presenting cells [181].

9.3 Approaches to load the margins with cancer-specific antigens and destroy it: sensitization or exogenous introduction of antigen

The amount of antigen expressed by the cancer cells seems to influence the degree of cross-presentation by the stroma [179]. We have shown, however, that the release of antigen can be enhanced by locally irradiating tumors or by administering the chemotherapeutical drug gemcitabine [182]. This pretreatment led to the eradication of tumors that would under simple adoptive T cell therapy escape as antigen loss variants. These experiments show the importance of cross-presentation for tumor eradication. Cross-presenting stromal cells become targets and once killed there is no support for the growth of remaining loss variants. Tumors are usually detected at a size of one cm in diameter and 109 cells [1]. Most of the variants have already evolved at this point. Variants can arise by deletion of the epitope or reduced surface expression/presentation. It is unclear whether the remaining loss variants die through T cell-mediated bystander killing, missing paracrine loop or anoikis, death by loss of attachment through destruction of ECM and fibroblasts [11].

10. Cancer cells survive at tumor margins because of pre-existing stroma and its vasculature

Cancer cells survive at tumor margins where they are supported by preexisting stroma and vasculature, which is different from neovasculature. There are several important implications: (i) Agents that block neangiogenesis such as anti-VEGF antibodies or VEGF-receptor traps arrest the growth of tumors but do not affect preexisting vessels at the tumor margins and therefore fail to be curative; (ii) targeting molecules preferentially expressed on neovasculature has the same limitations; (iii) for tumor eradication, antigens released from cancer cells must be abundant enough to be cross-presented by pre-existing stroma at the tumor margin so the T cells “excise” the tumor mass in the healthy margin which is also the aim of curative surgery.

11. Conclusion

Tumor stroma, consisting of connective tissue and ECM, vasculature and an infiltrate of immune cells is critical for tumor growth. Apart from providing the growing tumor with structure and blood supply, stromal cells are part of a paracrine loop necessary for the survival of cancer cells and also induce immune suppression. The importance of the single aspects of the support of tumor growth are not clear and it remains to be answered whether the cancer promotion effect or the immune suppressive effect supplied by the stroma plays the more important role. Because of the crucial role of stroma in tumor growth, however, various strategies are employed to target stroma and thereby treat cancer. Most approaches do not lead to cancer eradication however. This is probably due to redundancies among stromal cell types and the inability to target stroma just outside of the cancer rim. Surviving cancer cells can recruit new stroma and use preexistent vasculature to support their growth.

Only in cases where T cells can target cancer cells, stromal cells and presumably also the extended rim of the cancer (healthy margin), could established tumors be rejected. So far, this has only been shown using model antigens for tumor-specific antigens like SIYRYYGL, LCMV gp33 and OVA257, which are transfected into cancer cells [179, 180, 183]. We do not know how these peptides differ from naturally occurring tumor-specific antigens or which kinds of antigens can be effectively cross-presented. Furthermore, up to the present, only TCR-transgenic T cells have been used for adoptive transfer when targeting single epitopes. The evolving field of TCR gene transfer [184, 185] enables us to generate therapeutic quantities of tumor-specific T cells. The use of this approach to target tumor-specific antigens that are efficiently cross-presented on tumor stroma will hopefully lead to the successful results we now find in animal studies.

Acknowledgments

We thank Ainhoa Arina, David C. Binder and Rebecca B. Liu for critical review of the manuscript. This work was supported by NIH grants P01-CA97296, R01-CA22677 and R01-CA37516 to HS.

Abbreviations

APC

antigen presenting cell

BM

bone marrow

DC

dendritic cell

ECM

extracellular matrix

FAP

fibroblast activation protein-α

HSC

hematopoietic stem cells

MDSC

myeloid-derived suppressor cell

MHC

major histocompatibility complex

MMP-9

matrix metalloproteinase-9

PMA

phorbol 12-myristate 13-acetate

TAF

tumor-associated fibroblast

TAM

tumor-associated macrophage

TCR

T cell receptors

Treg

regulatory T cell

VEGF

vascular endothelial growth factor

Footnotes

Conflict of interest statement: The authors declare that there are no conflicts of interest.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Stricker TP, Kumar V. Robbins and Cotran Pathologic Basis of Disease. 8th. Philadelphia, PA: Saunders/Elsevier; 2010. Neoplasia; pp. 259–330. [Google Scholar]
  • 2.Klein G, Klein E. Immune surveillance against virus-induced tumors and nonrejectability of spontaneous tumors: contrasting consequences of host versus tumor evolution. Proc Natl Acad Sci U S A. 1977;74:2121–5. doi: 10.1073/pnas.74.5.2121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Ehrlich P. Experimentelle Studien an Mäusetumoren. Zeitschrift für Krebsforschung. 1907;5:59–81. [Google Scholar]
  • 4.Borst M. Allgemeine Pathologie der malignen Geschwülste. Leipzig: S. Hirzel; 1924. [Google Scholar]
  • 5.Tlsty TD, Coussens LM. Tumor stroma and regulation of cancer development. Annu Rev Pathol. 2006;1:119–50. doi: 10.1146/annurev.pathol.1.110304.100224. [DOI] [PubMed] [Google Scholar]
  • 6.Singh S, Ross SR, Acena M, Rowley DA, Schreiber H. Stroma is critical for preventing or permitting immunological destruction of antigenic cancer cells. J Exp Med. 1992;175:139–46. doi: 10.1084/jem.175.1.139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Ochsenbein AF, Klenerman P, Karrer U, Ludewig B, Pericin M, Hengartner H, et al. Immune surveillance against a solid tumor fails because of immunological ignorance. Proc Natl Acad Sci U S A. 1999;96:2233–8. doi: 10.1073/pnas.96.5.2233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Lin EY, Nguyen AV, Russell RG, Pollard JW. Colony-stimulating factor 1 promotes progression of mammary tumors to malignancy. J Exp Med. 2001;193:727–40. doi: 10.1084/jem.193.6.727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Old LJ, Boyse EA, Clarke DA, Carswell EA. Antigenic properties of chemically-induced tumors. Ann NY Acad Sci. 1962;101:80–106. [Google Scholar]
  • 10.Ochsenbein AF, Sierro S, Odermatt B, Pericin M, Karrer U, Hermans J, et al. Roles of tumour localization, second signals and cross priming in cytotoxic T-cell induction. Nature. 2001;411:1058–64. doi: 10.1038/35082583. [DOI] [PubMed] [Google Scholar]
  • 11.Ingber DE. Can cancer be reversed by engineering the tumor microenvironment? Semin Cancer Biol. 2008;18:356–64. doi: 10.1016/j.semcancer.2008.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Spiotto MT, Yu P, Rowley DA, Nishimura MI, Meredith SC, Gajewski TF, et al. Increasing tumor antigen expression overcomes “ignorance” to solid tumors via crosspresentation by bone marrow-derived stromal cells. Immunity. 2002;17:737–47. doi: 10.1016/s1074-7613(02)00480-6. [DOI] [PubMed] [Google Scholar]
  • 13.Seung LP, Rowley DA, Dubey P, Schreiber H. Synergy between T-cell immunity and inhibition of paracrine stimulation causes tumor rejection. Proc Natl Acad Sci U S A. 1995;92:6254–8. doi: 10.1073/pnas.92.14.6254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Seung LP, Seung SK, Schreiber H. Antigenic cancer cells that escape immune destruction are stimulated by host cells. Cancer Res. 1995;55:5094–100. [PubMed] [Google Scholar]
  • 15.Pekarek LA, Starr BA, Toledano AY, Schreiber H. Inhibition of tumor growth by elimination of granulocytes. J Exp Med. 1995;181:435–40. doi: 10.1084/jem.181.1.435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Sawyers CL. Where lies the blame for resistance--tumor or host? Nat Med. 2007;13:1144–5. doi: 10.1038/nm1007-1144. [DOI] [PubMed] [Google Scholar]
  • 17.Philip M, Rowley DA, Schreiber H. Inflammation as a tumor promoter in cancer induction. Semin Cancer Biol. 2004;14:433–9. doi: 10.1016/j.semcancer.2004.06.006. [DOI] [PubMed] [Google Scholar]
  • 18.Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420:860–7. doi: 10.1038/nature01322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Bissell MJ, Radisky D. Putting tumours in context. Nat Rev Cancer. 2001;1:46–54. doi: 10.1038/35094059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ostrand-Rosenberg S, Sinha P. Myeloid-derived suppressor cells: linking inflammation and cancer. J Immunol. 2009;182:4499–506. doi: 10.4049/jimmunol.0802740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140:883–99. doi: 10.1016/j.cell.2010.01.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Stuelten CH, Barbul A, Busch JI, Sutton E, Katz R, Sato M, et al. Acute wounds accelerate tumorigenesis by a T cell-dependent mechanism. Cancer Res. 2008;68:7278–82. doi: 10.1158/0008-5472.CAN-08-1842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Owen JL, Iragavarapu-Charyulu V, Gunja-Smith Z, Herbert LM, Grosso JF, Lopez DM. Up-regulation of matrix metalloproteinase-9 in T lymphocytes of mammary tumor bearers: role of vascular endothelial growth factor. J Immunol. 2003;171:4340–51. doi: 10.4049/jimmunol.171.8.4340. [DOI] [PubMed] [Google Scholar]
  • 24.Murdoch C, Muthana M, Coffelt SB, Lewis CE. The role of myeloid cells in the promotion of tumour angiogenesis. Nat Rev Cancer. 2008;8:618–31. doi: 10.1038/nrc2444. [DOI] [PubMed] [Google Scholar]
  • 25.Barnas JL, Simpson-Abelson MR, Yokota SJ, Kelleher RJ, Bankert RB. T cells and stromal fibroblasts in human tumor microenvironments represent potential therapeutic targets. Cancer Microenviron. 2010;3:29–47. doi: 10.1007/s12307-010-0044-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Bhowmick NA, Neilson EG, Moses HL. Stromal fibroblasts in cancer initiation and progression. Nature. 2004;432:332–7. doi: 10.1038/nature03096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Russell JP, Shinohara S, Melillo RM, Castellone MD, Santoro M, Rothstein JL. Tyrosine kinase oncoprotein, RET/PTC3, induces the secretion of myeloid growth and chemotactic factors. Oncogene. 2003;22:4569–77. doi: 10.1038/sj.onc.1206759. [DOI] [PubMed] [Google Scholar]
  • 28.Pufnock JS, Rothstein JL. Oncoprotein signaling mediates tumor-specific inflammation and enhances tumor progression. J Immunol. 2009;182:5498–506. doi: 10.4049/jimmunol.0801284. [DOI] [PubMed] [Google Scholar]
  • 29.Berenblum I. The mechanism of carcinogenesis: A study of the significance of cocarcinogenic action. Cancer Res. 1941;1:807–14. [Google Scholar]
  • 30.Ingber DE, Madri JA, Jamieson JD. Role of basal lamina in neoplastic disorganization of tissue architecture. Proc Natl Acad Sci U S A. 1981;78:3901–5. doi: 10.1073/pnas.78.6.3901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Nelson CM, Bissell MJ. Of extracellular matrix, scaffolds, and signaling: tissue architecture regulates development, homeostasis, and cancer. Annu Rev Cell Dev Biol. 2006;22:287–309. doi: 10.1146/annurev.cellbio.22.010305.104315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Fidler IJ, Fogler WE, Kleinerman ES, Saiki I. Abrogation of species specificity for activation of tumoricidal properties in macrophages by recombinant mouse or human interferon-gamma encapsulated in liposomes. J Immunol. 1985;135:4289–96. [PubMed] [Google Scholar]
  • 33.Thiery JP. Epithelial-mesenchymal transitions in development and pathologies. Curr Opin Cell Biol. 2003;15:740–6. doi: 10.1016/j.ceb.2003.10.006. [DOI] [PubMed] [Google Scholar]
  • 34.Guarino M, Rubino B, Ballabio G. The role of epithelial-mesenchymal transition in cancer pathology. Pathology. 2007;39:305–18. doi: 10.1080/00313020701329914. [DOI] [PubMed] [Google Scholar]
  • 35.Zhang B, Zhang Y, Bowerman NA, Schietinger A, Fu YX, Kranz DM, et al. Equilibrium between host and cancer caused by effector T cells killing tumor stroma. Cancer Res. 2008;68 doi: 10.1158/0008-5472.CAN-07-5324. in press. [DOI] [PubMed] [Google Scholar]
  • 36.Willis RA. Pathology of Tumours. London, England: Butterworth & Co., Ltd.; p. 1948. [Google Scholar]
  • 37.Iwano M, Plieth D, Danoff TM, Xue C, Okada H, Neilson EG. Evidence that fibroblasts derive from epithelium during tissue fibrosis. J Clin Invest. 2002;110:341–50. doi: 10.1172/JCI15518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Bhowmick NA, Chytil A, Plieth D, Gorska AE, Dumont N, Shappell S, et al. TGF-beta signaling in fibroblasts modulates the oncogenic potential of adjacent epithelia. Science. 2004;303:848–51. doi: 10.1126/science.1090922. [DOI] [PubMed] [Google Scholar]
  • 39.Rosty C, Ueki T, Argani P, Jansen M, Yeo CJ, Cameron JL, et al. Overexpression of S100A4 in pancreatic ductal adenocarcinomas is associated with poor differentiation and DNA hypomethylation. Am J Pathol. 2002;160:45–50. doi: 10.1016/S0002-9440(10)64347-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Mazzucchelli L. Protein S100A4: too long overlooked by pathologists? Am J Pathol. 2002;160:7–13. doi: 10.1016/S0002-9440(10)64342-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Ito K, Chen J, El Chaar M, Stern JM, Seshan SV, Khodadadian JJ, et al. Renal damage progresses despite improvement of renal function after relief of unilateral ureteral obstruction in adult rats. Am J Physiol Renal Physiol. 2004;287:F1283–93. doi: 10.1152/ajprenal.00441.2003. [DOI] [PubMed] [Google Scholar]
  • 42.Inoue T, Plieth D, Venkov CD, Xu C, Neilson EG. Antibodies against macrophages that overlap in specificity with fibroblasts. Kidney Int. 2005;67:2488–93. doi: 10.1111/j.1523-1755.2005.00358.x. [DOI] [PubMed] [Google Scholar]
  • 43.Le Hir M, Hegyi I, Cueni-Loffing D, Loffing J, Kaissling B. Characterization of renal interstitial fibroblast-specific protein 1/S100A4-positive cells in healthy and inflamed rodent kidneys. Histochem Cell Biol. 2005;123:335–46. doi: 10.1007/s00418-005-0788-z. [DOI] [PubMed] [Google Scholar]
  • 44.Schwann T. Microscopical researches into the accordance in the structure and growth of animals and plants. London: Sydenham Society; 1947. [DOI] [PubMed] [Google Scholar]
  • 45.Paget J. Lectures on surgical pathology delivered at the Royal College of Surgeons of England, London. London: Longmans & others; p. 1863. [Google Scholar]
  • 46.Dunphy JE. The fibroblast- a ubiquitous ally for the surgeon. New Engl J Med. 1963;268:1368–77. [Google Scholar]
  • 47.Clark JG, Hilderbran JN. Fibroblast procollagen production rates in vitro based on [3H] hydroxyproline production and procollagen hydroxyproline specific activity. Anal Biochem. 1984;140:478–85. doi: 10.1016/0003-2697(84)90197-0. [DOI] [PubMed] [Google Scholar]
  • 48.Bucher O. Histologie und mikroskopische Anatomie des Menschen. 4th. Bern, Switzerland: Medizinischer Verlag Hans Huber; 1965. [Google Scholar]
  • 49.Ham AW, Cormack DH. Histology. 8th. J.B.Lippincott Company; 1979. [Google Scholar]
  • 50.Folkman J, Klagsbrun M, Sasse J, Wadzinski M, Ingber D, Vlodavsky I. A heparin-binding angiogenic protein--basic fibroblast growth factor--is stored within basement membrane. Am J Pathol. 1988;130:393–400. [PMC free article] [PubMed] [Google Scholar]
  • 51.Kündig TM, Bachmann MF, DiPaolo C, Simard JJ, Battegay M, Lother H, et al. Fibroblasts as efficient antigen-presenting cells in lymphoid organs. Science. 1995;268:1343–7. doi: 10.1126/science.7761853. [DOI] [PubMed] [Google Scholar]
  • 52.Smith RS, Smith TJ, Blieden TM, Phipps RP. Fibroblasts as sentinel cells. Synthesis of chemokines and regulation of inflammation. Am J Pathol. 1997;151:317–22. [PMC free article] [PubMed] [Google Scholar]
  • 53.Chang HY, Nuyten DS, Sneddon JB, Hastie T, Tibshirani R, Sorlie T, et al. Robustness, scalability, and integration of a wound-response gene expression signature in predicting breast cancer survival. Proc Natl Acad Sci U S A. 2005;102:3738–43. doi: 10.1073/pnas.0409462102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Chang HY, Sneddon JB, Alizadeh AA, Sood R, West RB, Montgomery K, et al. Gene expression signature of fibroblast serum response predicts human cancer progression: similarities between tumors and wounds. PLoS Biol. 2004;2:E7. doi: 10.1371/journal.pbio.0020007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Strieter RM. What differentiates normal lung repair and fibrosis? Inflammation, resolution of repair, and fibrosis. Proc Am Thorac Soc. 2008;5:305–10. doi: 10.1513/pats.200710-160DR. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Kierszenbaum AL. Histology and cell biology An introduction to pathology. St Louis, MO: Mosby; 2002. Histology and cell biology; pp. 106–8. total pages 619. [Google Scholar]
  • 57.Junqueira LC, Carneiro J. Text and atlas. New York: Lange; 2003. Basic histology; pp. 95–125. [Google Scholar]
  • 58.Gottrup F, Storgård Jensen S, A JO. Wound healing subsequent to injury. In: Andreasen JO, Andreasen FM, Andersson L, editors. Textbook and color atlas of traumatic Injuries to teeth. 4th. Oxford, UK: Blackwell Munksgaard; pp. 2007pp. 1–61. [Google Scholar]
  • 59.Hansemann D. Das Krebsstroma und die Grawitz'sche Theorie der Schlummerzellen. Virchows Arch. 1893;133:147–65. [Google Scholar]
  • 60.Kerbel RS, Benezra R, Lyden DC, Hattori K, Heissig B, Nolan DJ, et al. Endothelial progenitor cells are cellular hubs essential for neoangiogenesis of certain aggressive adenocarcinomas and metastatic transition but not adenomas. Proc Natl Acad Sci U S A. 2008;105:E54. doi: 10.1073/pnas.0804876105. author reply E5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Gothert JR, Gustin SE, Van Eekelen AM, Schmidt U, Hall MA, Jane SM, et al. Genetically tagging endothelial cells in vivo:bone marrow-derived cells do not contribute to tumor endothelium. Blood. 2004;104:1769–77. doi: 10.1182/blood-2003-11-3952. [DOI] [PubMed] [Google Scholar]
  • 62.Larrivee B, Niessen K, Pollet I, Corbel SY, Long M, Rossi FM, et al. Minimal contribution of marrow-derived endothelial precursors to tumor vasculature. J Immunol. 2005;175:2890–9. doi: 10.4049/jimmunol.175.5.2890. [DOI] [PubMed] [Google Scholar]
  • 63.Udagawa T, Puder M, Wood M, Schaefer BC, D'Amato RJ. Analysis of tumor-associated stromal cells using SCID GFP transgenic mice: contribution of local and bone marrow-derived host cells. Faseb J. 2006;20:95–102. doi: 10.1096/fj.04-3669com. [DOI] [PubMed] [Google Scholar]
  • 64.Purhonen S, Palm J, Rossi D, Kaskenpaa N, Rajantie I, Yla-Herttuala S, et al. Bone marrow-derived circulating endothelial precursors do not contribute to vascular endothelium and are not needed for tumor growth. Proc Natl Acad Sci U S A. 2008;105:6620–5. doi: 10.1073/pnas.0710516105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Aschoff L. Das reticulo-endotheliale System. Ergebnisse der Inneren Medizin und Kinderheikunde. 1924;26:1–118. [Google Scholar]
  • 66.Crisan M, Yap S, Casteilla L, Chen CW, Corselli M, Park TS, et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell. 2008;3:301–13. doi: 10.1016/j.stem.2008.07.003. [DOI] [PubMed] [Google Scholar]
  • 67.Caplan AI. All MSCs are pericytes? Cell Stem Cell. 2008;3:229–30. doi: 10.1016/j.stem.2008.08.008. [DOI] [PubMed] [Google Scholar]
  • 68.Rouget CMB. Sur la contractilité capillaires sanguins. Comptes rendus de l'Académie des Sciences. 1879;88:916–8. [Google Scholar]
  • 69.Grillo HC. Origin of fibroblasts in wound healing. An autoradiographic study of inhibition of cellular proliferation by local x-irradiation. Ann Surg. 1963;157:453–67. doi: 10.1097/00000658-196303000-00018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Macdonald RA. Origin of fibroblasts in experimental healing wounds: autoradiographic studies using tritiated thymidine. Surgery. 1959;46:376–82. [PubMed] [Google Scholar]
  • 71.Ross R, Everett NB, Tyler R. Wound healing and collagen formation. VI. The origin of the wound fibroblast studied in parabiosis. J Cell Biol. 1970;44:645–54. doi: 10.1083/jcb.44.3.645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Maximov A. Culture of blood leukocytes. From lymphocytes and monocytes to connective tissue. Arch Exp Zellforsch. 1928;5:169–268. [Google Scholar]
  • 73.Bucala R, Spiegel LA, Chesney J, Hogan M, Cerami A. Circulating fibrocytes define a new leukocyte subpopulation that mediates tissue repair. Mol Med. 1994;1:71–81. [PMC free article] [PubMed] [Google Scholar]
  • 74.Phillips RJ, Burdick MD, Hong K, Lutz MA, Murray LA, Xue YY, et al. Circulating fibrocytes traffic to the lungs in response to CXCL12 and mediate fibrosis. J Clin Invest. 2004;114:438–46. doi: 10.1172/JCI20997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Ishii G, Ito TK, Aoyagi K, Fujimoto H, Chiba H, Hasebe T, et al. Presence of human circulating progenitor cells for cancer stromal fibroblasts in the blood of lung cancer patients. Stem Cells. 2007;25:1469–77. doi: 10.1634/stemcells.2006-0449. [DOI] [PubMed] [Google Scholar]
  • 76.Sangai T, Ishii G, Kodama K, Miyamoto S, Aoyagi Y, Ito T, et al. Effect of differences in cancer cells and tumor growth sites on recruiting bone marrow-derived endothelial cells and myofibroblasts in cancer-induced stroma. Int J Cancer. 2005;115:885–92. doi: 10.1002/ijc.20969. [DOI] [PubMed] [Google Scholar]
  • 77.Direkze NC, Hodivala-Dilke K, Jeffery R, Hunt T, Poulsom R, Oukrif D, et al. Bone marrow contribution to tumor-associated myofibroblasts and fibroblasts. Cancer Res. 2004;64:8492–5. doi: 10.1158/0008-5472.CAN-04-1708. [DOI] [PubMed] [Google Scholar]
  • 78.LaRue AC, Masuya M, Ebihara Y, Fleming PA, Visconti RP, Minamiguchi H, et al. Hematopoietic origins of fibroblasts: I. In vivo studies of fibroblasts associated with solid tumors. Exp Hematol. 2006;34:208–18. doi: 10.1016/j.exphem.2005.10.009. [DOI] [PubMed] [Google Scholar]
  • 79.De Wever O, Demetter P, Mareel M, Bracke M. Stromal myofibroblasts are drivers of invasive cancer growth. Int J Cancer. 2008;123:2229–38. doi: 10.1002/ijc.23925. [DOI] [PubMed] [Google Scholar]
  • 80.Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009 doi: 10.1038/nri2506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Sinha P, Chornoguz O, Clements VK, Artemenko KA, Zubarev RA, Ostrand-Rosenberg S. Myeloid-derived suppressor cells express the death receptor Fas and apoptose in response to T cell-expressed FasL. Blood. 2011;117:5381–90. doi: 10.1182/blood-2010-11-321752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Sinha P, Clements VK, Ostrand-Rosenberg S. Reduction of myeloid-derived suppressor cells and induction of M1 macrophages facilitate the rejection of established metastatic disease. J Immunol. 2005;174:636–45. doi: 10.4049/jimmunol.174.2.636. [DOI] [PubMed] [Google Scholar]
  • 83.Corzo CA, Condamine T, Lu L, Cotter MJ, Youn JI, Cheng P, et al. HIF-1alpha regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment. J Exp Med. 2010;207:2439–53. doi: 10.1084/jem.20100587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Kusmartsev S, Gabrilovich DI. STAT1 signaling regulates tumor-associated macrophage-mediated T cell deletion. J Immunol. 2005;174:4880–91. doi: 10.4049/jimmunol.174.8.4880. [DOI] [PubMed] [Google Scholar]
  • 85.Movahedi K, Guilliams M, Van den Bossche J, Van den Bergh R, Gysemans C, Beschin A, et al. Identification of discrete tumor-induced myeloid-derived suppressor cell subpopulations with distinct T cell-suppressive activity. Blood. 2008;111:4233–44. doi: 10.1182/blood-2007-07-099226. [DOI] [PubMed] [Google Scholar]
  • 86.Movahedi K, Laoui D, Gysemans C, Baeten M, Stange G, Van den Bossche J, et al. Different tumor microenvironments contain functionally distinct subsets of macrophages derived from Ly6C(high) monocytes. Cancer Res. 2010;70:5728–39. doi: 10.1158/0008-5472.CAN-09-4672. [DOI] [PubMed] [Google Scholar]
  • 87.Solito S, Bronte V, Mandruzzato S. Antigen specificity of immune suppression by myeloid-derived suppressor cells. J Leukoc Biol. 2011 doi: 10.1189/jlb.0111021. [DOI] [PubMed] [Google Scholar]
  • 88.Rous P, Kidd JG. Conditional neoplasms and subthreshold neoplastic states. A study of the tar tumors of rabbits. J Exp Med. 1941;73:365–90. doi: 10.1084/jem.73.3.365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Yuspa SH. The pathogenesis of squamous cell cancer: lessons learned from studies of skin carcinogenesis-Thirty-third G.H.A. Clowes Memorial Award Lecture. Cancer Res. 1994;54:1178–89. [PubMed] [Google Scholar]
  • 90.Braun AC. Plant tumors as an experimental model. Harvey Lect. 1960;56:191–210. [PubMed] [Google Scholar]
  • 91.Mintz B, Illmensee K. Normal genetically mosaic mice produced from malignant teratocarcinoma cells. Proc Natl Acad Sci U S A. 1975;72:3585–9. doi: 10.1073/pnas.72.9.3585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Kenny PA, Bissell MJ. Tumor reversion: correction of malignant behavior by microenvironmental cues. International Journal of Cancer. 2003;107:688–95. doi: 10.1002/ijc.11491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Proia DA, Kuperwasser C. Stroma: tumor agonist or antagonist. Cell Cycle. 2005;4:1022–5. doi: 10.4161/cc.4.8.1903. [DOI] [PubMed] [Google Scholar]
  • 94.Sympson CJ, Bissell MJ, Werb Z. Mammary gland tumor formation in transgenic mice overexpressing stromelysin-1. Semin Cancer Biol. 1995;6:159–63. doi: 10.1006/scbi.1995.0022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Sternlicht MD, Bissell MJ, Werb Z. The matrix metalloproteinase stromelysin-1 acts as a natural mammary tumor promoter. Oncogene. 2000;19:1102–13. doi: 10.1038/sj.onc.1203347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Maffini MV, Soto AM, Calabro JM, Ucci AA, Sonnenschein C. The stroma as a crucial target in rat mammary gland carcinogenesis. J Cell Sci. 2004;117:1495–502. doi: 10.1242/jcs.01000. [DOI] [PubMed] [Google Scholar]
  • 97.Medina D, Kittrell F. Stroma is not a major target in DMBA-mediated tumorigenesis of mouse mammary preneoplasia. J Cell Sci. 2005;118:123–7. doi: 10.1242/jcs.01597. [DOI] [PubMed] [Google Scholar]
  • 98.Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454:436–44. doi: 10.1038/nature07205. [DOI] [PubMed] [Google Scholar]
  • 99.Ghosh AK, Secreto CR, Knox TR, Ding W, Mukhopadhyay D, Kay NE. Circulating microvesicles in B-cell chronic lymphocytic leukemia can stimulate marrow stromal cells: implications for disease progression. Blood. 2010;115:1755–64. doi: 10.1182/blood-2009-09-242719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Delmonte L, Liebelt RA. Granulocytosis-Promoting Extract Of Mouse Tumor Tissue: Partial Purification. Science. 1965;148:521–3. doi: 10.1126/science.148.3669.521. [DOI] [PubMed] [Google Scholar]
  • 101.Hibberd AD, Metcalf D. Proliferation of macrophage and granulocyte precursors in response to primary and transplanted tumors. Isr J Med Sci. 1971;7:202–10. [PubMed] [Google Scholar]
  • 102.Milas L, Basic I. Stimulated granulocytopoiesis in mice bearing fibrosarcoma. Eur J Cancer. 1972;8:309–13. doi: 10.1016/0014-2964(72)90026-6. [DOI] [PubMed] [Google Scholar]
  • 103.Balducci L, Hardy C. High proliferation of granulocyte-macrophage progenitors in tumor-bearing mice. Cancer Res. 1983;43:4643–7. [PubMed] [Google Scholar]
  • 104.Lee MY, Kaushansky K, Judkins SA, Lottsfeldt JL, Waheed A, Shadduck RK. Mechanisms of tumor-induced neutrophilia: constitutive production of colony-stimulating factors and their synergistic actions. Blood. 1989;74:115–22. [PubMed] [Google Scholar]
  • 105.Cardiff RD, Leder A, Kuo A, Pattengale PK, Leder P. Multiple tumor types appear in a transgenic mouse with the ras oncogene. American Journal of Pathology. 1993;142:1199–207. [PMC free article] [PubMed] [Google Scholar]
  • 106.Scholl SM, Pallud C, Beuvon F, Hacene K, Stanley ER, Rohrschneider L, et al. Anti-colony-stimulating factor-1 antibody staining in primary breast adenocarcinomas correlates with marked inflammatory cell infiltrates and prognosis. J Natl Cancer Inst. 1994;86:120–6. doi: 10.1093/jnci/86.2.120. [DOI] [PubMed] [Google Scholar]
  • 107.Uemura Y, Kobayashi M, Nakata H, Harada R, Kubota T, Taguchi H. Effect of serum deprivation on constitutive production of granulocyte-colony stimulating factor and granulocyte macrophage-colony stimulating factor in lung cancer cells. Int J Cancer. 2004;109:826–32. doi: 10.1002/ijc.20023. [DOI] [PubMed] [Google Scholar]
  • 108.DuPre SA, Hunter KW., Jr Murine mammary carcinoma 4T1 induces a leukemoid reaction with splenomegaly: association with tumor-derived growth factors. Exp Mol Pathol. 2007;82:12–24. doi: 10.1016/j.yexmp.2006.06.007. [DOI] [PubMed] [Google Scholar]
  • 109.Woglom WH. The size of the spleen in immune mice. Journal of Cancer Research. 1919;4:281–323. [Google Scholar]
  • 110.Radoja S, Rao TD, Hillman D, Frey AB. Mice bearing late-stage tumors have normal functional systemic T cell responses in vitro and in vivo. J Immunol. 2000;164:2619–28. doi: 10.4049/jimmunol.164.5.2619. [DOI] [PubMed] [Google Scholar]
  • 111.Petit I, Szyper-Kravitz M, Nagler A, Lahav M, Peled A, Habler L, et al. G-CSF induces stem cell mobilization by decreasing bone marrow SDF-1 and up-regulating CXCR4. Nat Immunol. 2002;3:687–94. doi: 10.1038/ni813. [DOI] [PubMed] [Google Scholar]
  • 112.Jameson JL, Johnson BE. Paraneoplastic syndromes: endocrinologic/hematologic. In: Fauci AS, editor. Harrison's principles of internal medicine. 17th. New York: McGraw-Hill Medical; pp. 2008pp. 617–23. [Google Scholar]
  • 113.Mullen CA, Urban JL, Van Waes C, Rowley DA, Schreiber H. Multiple cancers. Tumor burden permits the outgrowth of other cancers. J Exp Med. 1985;162:1665–82. doi: 10.1084/jem.162.5.1665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Mullen CA, Rowley DA, Schreiber H. Highly immunogenic regressor tumor cells can prevent development of postsurgical tumor immunity. Cell Immunol. 1989;119:101–13. doi: 10.1016/0008-8749(89)90227-x. [DOI] [PubMed] [Google Scholar]
  • 115.McAllister SS, Gifford AM, Greiner AL, Kelleher SP, Saelzler MP, Ince TA, et al. Systemic endocrine instigation of indolent tumor growth requires osteopontin. Cell. 2008;133:994–1005. doi: 10.1016/j.cell.2008.04.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Adams GB, Scadden DT. The hematopoietic stem cell in its place. Nat Immunol. 2006;7:333–7. doi: 10.1038/ni1331. [DOI] [PubMed] [Google Scholar]
  • 117.Rodrigues LR, Teixeira JA, Schmitt FL, Paulsson M, Lindmark-Mansson H. The role of osteopontin in tumor progression and metastasis in breast cancer. Cancer Epidemiol Biomarkers Prev. 2007;16:1087–97. doi: 10.1158/1055-9965.EPI-06-1008. [DOI] [PubMed] [Google Scholar]
  • 118.Koh A, da Silva AP, Bansal AK, Bansal M, Sun C, Lee H, et al. Role of osteopontin in neutrophil function. Immunology. 2007;122:466–75. doi: 10.1111/j.1365-2567.2007.02682.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Kaplan RN, Riba RD, Zacharoulis S, Bramley AH, Vincent L, Costa C, et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature. 2005;438:820–7. doi: 10.1038/nature04186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Wels J, Kaplan RN, Rafii S, Lyden D. Migratory neighbors and distant invaders: tumor-associated niche cells. Genes Dev. 2008;22:559–74. doi: 10.1101/gad.1636908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Yang L, DeBusk LM, Fukuda K, Fingleton B, Green-Jarvis B, Shyr Y, et al. Expansion of myeloid immune suppressor Gr+CD11b+ cells in tumor-bearing host directly promotes tumor angiogenesis. Cancer Cell. 2004;6:409–21. doi: 10.1016/j.ccr.2004.08.031. [DOI] [PubMed] [Google Scholar]
  • 122.Shojaei F, Wu X, Malik AK, Zhong C, Baldwin ME, Schanz S, et al. Tumor refractoriness to anti-VEGF treatment is mediated by CD11b+Gr1+ myeloid cells. Nat Biotechnol. 2007;25:911–20. doi: 10.1038/nbt1323. [DOI] [PubMed] [Google Scholar]
  • 123.Yang L, Huang J, Ren X, Gorska AE, Chytil A, Aakre M, et al. Abrogation of TGF beta signaling in mammary carcinomas recruits Gr-1+CD11b+ myeloid cells that promote metastasis. Cancer Cell. 2008;13:23–35. doi: 10.1016/j.ccr.2007.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Kryczek I, Wei S, Keller E, Liu R, Zou W. Stroma-derived factor (SDF-1/CXCL12) and human tumor pathogenesis. Am J Physiol Cell Physiol. 2007;292:C987–95. doi: 10.1152/ajpcell.00406.2006. [DOI] [PubMed] [Google Scholar]
  • 125.Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, et al. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 2005;121:335–48. doi: 10.1016/j.cell.2005.02.034. [DOI] [PubMed] [Google Scholar]
  • 126.Orimo A, Weinberg RA. Stromal fibroblasts in cancer: a novel tumor-promoting cell type. Cell Cycle. 2006;5:1597–601. doi: 10.4161/cc.5.15.3112. [DOI] [PubMed] [Google Scholar]
  • 127.Chen W, Jin W, Hardegen N, Lei KJ, Li L, Marinos N, et al. Conversion of peripheral CD4+CD25- naive T cells to CD4+CD25+ regulatory T cells by TGF-beta induction of transcription factor Foxp3. J Exp Med. 2003;198:1875–86. doi: 10.1084/jem.20030152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Faunce DE, Terajewicz A, Stein-Streilein J. Cutting edge: in vitro-generated tolerogenic APC induce CD8+ T regulatory cells that can suppress ongoing experimental autoimmune encephalomyelitis. J Immunol. 2004;172:1991–5. doi: 10.4049/jimmunol.172.4.1991. [DOI] [PubMed] [Google Scholar]
  • 129.Zhang-Hoover J, Stein-Streilein J. Tolerogenic APC generate CD8+ T regulatory cells that modulate pulmonary interstitial fibrosis. J Immunol. 2004;172:178–85. doi: 10.4049/jimmunol.172.1.178. [DOI] [PubMed] [Google Scholar]
  • 130.Andersson J, Tran DQ, Pesu M, Davidson TS, Ramsey H, O'Shea JJ, et al. CD4+ FoxP3+ regulatory T cells confer infectious tolerance in a TGF-beta-dependent manner. J Exp Med. 2008;205:1975–81. doi: 10.1084/jem.20080308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Rao RM, Betz TV, Lamont DJ, Kim MB, Shaw SK, Froio RM, et al. Elastase release by transmigrating neutrophils deactivates endothelial-bound SDF-1alpha and attenuates subsequent T lymphocyte transendothelial migration. J Exp Med. 2004;200:713–24. doi: 10.1084/jem.20040499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Zou L, Barnett B, Safah H, Larussa VF, Evdemon-Hogan M, Mottram P, et al. Bone marrow is a reservoir for CD4+CD25+ regulatory T cells that traffic through CXCL12/CXCR4 signals. Cancer Res. 2004;64:8451–5. doi: 10.1158/0008-5472.CAN-04-1987. [DOI] [PubMed] [Google Scholar]
  • 133.Zou W, Machelon V, Coulomb-L'Hermin A, Borvak J, Nome F, Isaeva T, et al. Stromal-derived factor-1 in human tumors recruits and alters the function of plasmacytoid precursor dendritic cells. Nat Med. 2001;7:1339–46. doi: 10.1038/nm1201-1339. [DOI] [PubMed] [Google Scholar]
  • 134.Foulds L. The experimental study of tumor progression. A review. Cancer Res. 1954;14:327–39. [PubMed] [Google Scholar]
  • 135.Kripke ML. Antigenicity of murine skin tumors induced by ultraviolet light. J Natl Cancer Inst. 1974;53:1333–36. doi: 10.1093/jnci/53.5.1333. [DOI] [PubMed] [Google Scholar]
  • 136.Ward PL, Koeppen H, Hurteau T, Schreiber H. Tumor antigens defined by cloned immunological probes are highly polymorphic and are not detected on autologous normal cells. J Exp Med. 1989;170:217–32. doi: 10.1084/jem.170.1.217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Engel AM, Svane IM, Rygaard J, Werdelin O. MCA sarcomas induced in scid mice are more immunogenic than MCA sarcomas induced in congenic, immunocompetent mice. Scand J Immunol. 1997;45:463–70. doi: 10.1046/j.1365-3083.1997.d01-419.x. [DOI] [PubMed] [Google Scholar]
  • 138.Svane IM, Engel AM, Nielsen MB, Ljunggren HG, Rygaard J, Werdelin O. Chemically induced sarcomas from nude mice are more immunogenic than similar sarcomas from congenic normal mice. Eur J Immunol. 1996;26:1844–50. doi: 10.1002/eji.1830260827. [DOI] [PubMed] [Google Scholar]
  • 139.Shankaran V, Ikeda H, Bruce AT, White JM, Swanson PE, Old LJ, et al. IFN gamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature. 2001;410:1107–11. doi: 10.1038/35074122. [DOI] [PubMed] [Google Scholar]
  • 140.Ward PL, Koeppen HK, Hurteau T, Rowley DA, Schreiber H. Major histocompatibility complex class I and unique antigen expression by murine tumors that escaped from CD8+ T-cell-dependent surveillance. Cancer Res. 1990;50:3851–8. [PubMed] [Google Scholar]
  • 141.Wu TH, Schreiber K, Arina A, Khodarev NN, Efimova EV, Rowley DA, et al. Progression of cancer from indolent to aggressive despite antigen retention and increased expression of interferon-gamma inducible genes. Cancer Immun. 2011;11:2. [PMC free article] [PubMed] [Google Scholar]
  • 142.Kusmartsev S, Cheng F, Yu B, Nefedova Y, Sotomayor E, Lush R, et al. All-trans-retinoic acid eliminates immature myeloid cells from tumor-bearing mice and improves the effect of vaccination. Cancer Res. 2003;63:4441–9. [PubMed] [Google Scholar]
  • 143.Beatty GL, Chiorean EG, Fishman MP, Saboury B, Teitelbaum UR, Sun W, et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science. 2011;331:1612–6. doi: 10.1126/science.1198443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Noffz G, Qin Z, Kopf M, Blankenstein T. Neutrophils but not eosinophils are involved in growth suppression of IL-4-secreting tumors. J Immunol. 1998;160:345–50. [PubMed] [Google Scholar]
  • 145.Sinha P, Clements VK, Miller S, Ostrand-Rosenberg S. Tumor immunity: a balancing act between T cell activation, macrophage activation and tumor-induced immune suppression. Cancer Immunol Immunother. 2005;54:1137–42. doi: 10.1007/s00262-005-0703-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Suzuki E, Kapoor V, Jassar AS, Kaiser LR, Albelda SM. Gemcitabine selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity. Clin Cancer Res. 2005;11:6713–21. doi: 10.1158/1078-0432.CCR-05-0883. [DOI] [PubMed] [Google Scholar]
  • 147.Medina-Echeverz J, Fioravanti J, Zabala M, Ardaiz N, Prieto J, Berraondo P. Successful colon cancer eradication after chemoimmunotherapy is associated with profound phenotypic change of intratumoral myeloid cells. J Immunol. 2011;186:807–15. doi: 10.4049/jimmunol.1001483. [DOI] [PubMed] [Google Scholar]
  • 148.Luo Y, Zhou H, Krueger J, Kaplan C, Lee SH, Dolman C, et al. Targeting tumor-associated macrophages as a novel strategy against breast cancer. J Clin Invest. 2006;116:2132–41. doi: 10.1172/JCI27648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Melani C, Sangaletti S, Barazzetta FM, Werb Z, Colombo MP. Amino-biphosphonate-mediated MMP-9 inhibition breaks the tumor-bone marrow axis responsible for myeloid-derived suppressor cell expansion and macrophage infiltration in tumor stroma. Cancer Res. 2007;67:11438–46. doi: 10.1158/0008-5472.CAN-07-1882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Serafini P, Meckel K, Kelso M, Noonan K, Califano J, Koch W, et al. Phosphodiesterase-5 inhibition augments endogenous antitumor immunity by reducing myeloid-derived suppressor cell function. J Exp Med. 2006;203:2691–702. doi: 10.1084/jem.20061104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Kraman M, Bambrough PJ, Arnold JN, Roberts EW, Magiera L, Jones JO, et al. Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein-alpha. Science. 2010;330:827–30. doi: 10.1126/science.1195300. [DOI] [PubMed] [Google Scholar]
  • 152.Liao D, Luo Y, Markowitz D, Xiang R, Reisfeld RA. Cancer associated fibroblasts promote tumor growth and metastasis by modulating the tumor immune microenvironment in a 4T1 murine breast cancer model. PLoS One. 2009;4:e7965. doi: 10.1371/journal.pone.0007965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Wen Y, Wang CT, Ma TT, Li ZY, Zhou LN, Mu B, et al. Immunotherapy targeting fibroblast activation protein inhibits tumor growth and increases survival in a murine colon cancer model. Cancer Sci. 2010;101:2325–32. doi: 10.1111/j.1349-7006.2010.01695.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Campoli M, Ferrone S, Wang X. Functional and clinical relevance of chondroitin sulfate proteoglycan 4. Adv Cancer Res. 2010;109:73–121. doi: 10.1016/B978-0-12-380890-5.00003-X. [DOI] [PubMed] [Google Scholar]
  • 155.Moinfar F, Man YG, Arnould L, Bratthauer GL, Ratschek M, Tavassoli FA. Concurrent and independent genetic alterations in the stromal and epithelial cells of mammary carcinoma: implications for tumorigenesis. Cancer Res. 2000;60:2562–6. [PubMed] [Google Scholar]
  • 156.Wernert N, Locherbach C, Wellmann A, Behrens P, Hugel A. Presence of genetic alterations in microdissected stroma of human colon and breast cancers. Anticancer Res. 2001;21:2259–64. [PubMed] [Google Scholar]
  • 157.Matsumoto N, Yoshida T, Yamashita K, Numata Y, Okayasu I. Possible alternative carcinogenesis pathway featuring microsatellite instability in colorectal cancer stroma. Br J Cancer. 2003;89:707–12. doi: 10.1038/sj.bjc.6601141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Allinen M, Beroukhim R, Cai L, Brennan C, Lahti-Domenici J, Huang H, et al. Molecular characterization of the tumor microenvironment in breast cancer. Cancer Cell. 2004;6:17–32. doi: 10.1016/j.ccr.2004.06.010. [DOI] [PubMed] [Google Scholar]
  • 159.Fukino K, Shen L, Patocs A, Mutter GL, Eng C. Genomic instability within tumor stroma and clinicopathological characteristics of sporadic primary invasive breast carcinoma. Jama. 2007;297:2103–11. doi: 10.1001/jama.297.19.2103. [DOI] [PubMed] [Google Scholar]
  • 160.Ishiguro K, Yoshida T, Yagishita H, Numata Y, Okayasu T. Epithelial and stromal genetic instability contributes to genesis of colorectal adenomas. Gut. 2006;55:695–702. doi: 10.1136/gut.2005.079459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Bian Y, Knobloch TJ, Sadim M, Kaklamani V, Raji A, Yang GY, et al. Somatic acquisition of TGFBR1*6A by epithelial and stromal cells during head and neck and colon cancer development. Hum Mol Genet. 2007;16:3128–35. doi: 10.1093/hmg/ddm274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Patocs A, Zhang L, Xu Y, Weber F, Caldes T, Mutter GL, et al. Breast-cancer stromal cells with TP53 mutations and nodal metastases. N Engl J Med. 2007;357:2543–51. doi: 10.1056/NEJMoa071825. [DOI] [PubMed] [Google Scholar]
  • 163.Qiu W, Hu M, Sridhar A, Opeskin K, Fox S, Shipitsin M, et al. No evidence of clonal somatic genetic alterations in cancer-associated fibroblasts from human breast and ovarian carcinomas. Nat Genet. 2008 doi: 10.1038/ng.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Shalapour S, Eckert C, Seeger K, Pfau M, Prada J, Henze G, et al. Leukemia-associated genetic aberrations in mesenchymal stem cells of children with acute lymphoblastic leukemia. J Mol Med. 2010;88:249–65. doi: 10.1007/s00109-009-0583-8. [DOI] [PubMed] [Google Scholar]
  • 165.Hu M, Yao J, Cai L, Bachman KE, van den Brule F, Velculescu V, et al. Distinct epigenetic changes in the stromal cells of breast cancers. Nat Genet. 2005;37:899–905. doi: 10.1038/ng1596. [DOI] [PubMed] [Google Scholar]
  • 166.Fiegl H, Millinger S, Goebel G, Muller-Holzner E, Marth C, Laird PW, et al. Breast cancer DNA methylation profiles in cancer cells and tumor stroma: association with HER-2/neu status in primary breast cancer. Cancer Res. 2006;66:29–33. doi: 10.1158/0008-5472.CAN-05-2508. [DOI] [PubMed] [Google Scholar]
  • 167.Janowska-Wieczorek A, Wysoczynski M, Kijowski J, Marquez-Curtis L, Machalinski B, Ratajczak J, et al. Microvesicles derived from activated platelets induce metastasis and angiogenesis in lung cancer. Int J Cancer. 2005;113:752–60. doi: 10.1002/ijc.20657. [DOI] [PubMed] [Google Scholar]
  • 168.Cocucci E, Racchetti G, Meldolesi J. Shedding microvesicles: artefacts no more. Trends Cell Biol. 2009;19:43–51. doi: 10.1016/j.tcb.2008.11.003. [DOI] [PubMed] [Google Scholar]
  • 169.Thery C, Ostrowski M, Segura E. Membrane vesicles as conveyors of immune responses. Nat Rev Immunol. 2009;9:581–93. doi: 10.1038/nri2567. [DOI] [PubMed] [Google Scholar]
  • 170.Utsugi-Kobukai S, Fujimaki H, Hotta C, Nakazawa M, Minami M. MHC class I-mediated exogenous antigen presentation by exosomes secreted from immature and mature bone marrow derived dendritic cells. Immunol Lett. 2003;89:125–31. doi: 10.1016/s0165-2478(03)00128-7. [DOI] [PubMed] [Google Scholar]
  • 171.Obregon C, Rothen-Rutishauser B, Gitahi SK, Gehr P, Nicod LP. Exovesicles from human activated dendritic cells fuse with resting dendritic cells, allowing them to present alloantigens. Am J Pathol. 2006;169:2127–36. doi: 10.2353/ajpath.2006.060453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Wolfers J, Lozier A, Raposo G, Regnault A, Thery C, Masurier C, et al. Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming. Nature Medicine. 2001;7:297–303. doi: 10.1038/85438. [DOI] [PubMed] [Google Scholar]
  • 173.Andre F, Schartz NE, Movassagh M, Flament C, Pautier P, Morice P, et al. Malignant effusions and immunogenic tumour-derived exosomes. Lancet. 2002;360:295–305. doi: 10.1016/S0140-6736(02)09552-1. [DOI] [PubMed] [Google Scholar]
  • 174.Poutsiaka DD, Schroder EW, Taylor DD, Levy EM, Black PH. Membrane vesicles shed by murine melanoma cells selectively inhibit the expression of Ia antigen by macrophages. J Immunol. 1985;134:138–44. [PubMed] [Google Scholar]
  • 175.Vega VL, Rodriguez-Silva M, Frey T, Gehrmann M, Diaz JC, Steinem C, et al. Hsp70 translocates into the plasma membrane after stress and is released into the extracellular environment in a membrane-associated form that activates macrophages. J Immunol. 2008;180:4299–307. doi: 10.4049/jimmunol.180.6.4299. [DOI] [PubMed] [Google Scholar]
  • 176.Gastpar R, Gehrmann M, Bausero MA, Asea A, Gross C, Schroeder JA, et al. Heat shock protein 70 surface-positive tumor exosomes stimulate migratory and cytolytic activity of natural killer cells. Cancer Res. 2005;65:5238–47. doi: 10.1158/0008-5472.CAN-04-3804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Skitzki JJ, Repasky EA, Evans SS. Hyperthermia as an immunotherapy strategy for cancer. Curr Opin Investig Drugs. 2009;10:550–8. [PMC free article] [PubMed] [Google Scholar]
  • 178.Wang XY, Sun X, Chen X, Facciponte J, Repasky EA, Kane J, et al. Superior antitumor response induced by large stress protein chaperoned protein antigen compared with peptide antigen. J Immunol. 2010;184:6309–19. doi: 10.4049/jimmunol.0903891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Spiotto MT, Rowley DA, Schreiber H. Bystander elimination of antigen loss variants in established tumors. Nat Med. 2004;10:294–8. doi: 10.1038/nm999. [DOI] [PubMed] [Google Scholar]
  • 180.Spiotto MT, Schreiber H. Rapid destruction of the tumor microenvironment by CTLs recognizing cancer-specific antigens cross-presented by stromal cells. Cancer Immun. 2005;5:8. [PubMed] [Google Scholar]
  • 181.Asano K, Nabeyama A, Miyake Y, Qiu CH, Kurita A, Tomura M, et al. CD169-positive macrophages dominate antitumor immunity by crosspresenting dead cell-associated antigens. Immunity. 2011;34:85–95. doi: 10.1016/j.immuni.2010.12.011. [DOI] [PubMed] [Google Scholar]
  • 182.Zhang B, Bowerman NA, Salama JK, Schmidt H, Spiotto MT, Schietinger A, et al. Induced sensitization of tumor stroma leads to eradication of established cancer by T cells. J Exp Med. 2007;204:49–55. doi: 10.1084/jem.20062056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Garcia-Hernandez Mde L, Hamada H, Reome JB, Misra SK, Tighe MP, Dutton RW. Adoptive transfer of tumor-specific Tc17 effector T cells controls the growth of B16 melanoma in mice. J Immunol. 2010;184:4215–27. doi: 10.4049/jimmunol.0902995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Engels B, Uckert W. Redirecting T lymphocyte specificity by T cell receptor gene transfer--a new era for immunotherapy. Mol Aspects Med. 2007;28:115–42. doi: 10.1016/j.mam.2006.12.006. [DOI] [PubMed] [Google Scholar]
  • 185.Bendle GM, Haanen JB, Schumacher TN. Preclinical development of T cell receptor gene therapy. Curr Opin Immunol. 2009;21:209–14. doi: 10.1016/j.coi.2009.02.007. [DOI] [PubMed] [Google Scholar]

RESOURCES