Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Nov 7.
Published in final edited form as: Nat Cell Biol. 2014 Aug;16(8):717–727. doi: 10.1038/ncb3015

The tumour-induced systemic environment as a critical regulator of cancer progression and metastasis

Sandra S McAllister 1, Robert A Weinberg 1
PMCID: PMC6220424  NIHMSID: NIHMS993924  PMID: 25082194

Abstract

Recent pre-clinical and clinical research has provided evidence that cancer progression is driven not only by a tumour’s underlying genetic alterations and paracrine interactions within the tumour microenvironment, but also by complex systemic processes. We review these emerging paradigms of cancer pathophysiology and discuss how a clearer understanding of systemic regulation of cancer progression could guide development of new therapeutic modalities and efforts to prevent disease relapse following initial diagnosis and treatment.


Solid tumours are composed of heterogeneous populations of neoplastic cells and an elaborate array of recruited mesenchymal and inflammatory cells of host origin that form the tumour-associated stroma, and are collectively known as the tumour microenvironment (TME)1. Traditionally, the biology of solid tumours has been portrayed as being governed by the genetic and epigenetic alterations that neoplastic cells undergo during the course of multi-step tumour pathogenesis. However, over the past two decades, the TME has also emerged as an equally important determinant of tumour behaviour. Paracrine interactions between cancer cells and the TME have been shown to regulate overall tumour growth, homeostasis and progression2. Moreover, molecular profiling of stromal cells from a variety of different human tumour specimens has yielded information of prognostic value, further highlighting the critical role that the TME plays in directing tumour development37.

Investigations into the role of the TME and the mechanisms of stromal cell recruitment have also provided insights into a distinct and intriguing aspect of tumour biology: cancer progression may also be directed by the body’s systemic responses to malignancy and by the involvement of organ systems located at sites distant from the site of primary tumour growth. Some of the most compelling evidence that cancers exert unique and specific systemic effects comes from studies showing that tissues can be altered at the structural, histopathological and molecular levels in the presence of a distant malignancy, as we discuss in this Review. Thus, the view of cancer as a systemic disease, which was once a matter of speculation, is now emerging into the research spotlight8,9.

In some cases, the systemic interactions between tumour and host mimic and co-opt normal physiological processes, such as inflammation and wound healing. Indeed, the histopathological appearance of the TME of most carcinomas closely resembles that of inflamed and wounded tissues, a similarity that was noted a quarter of a century ago when tumours were described as “wounds that do not heal”10. These similarities explain why studies of other pathological processes, such as wound healing11, inflammation1216 and organ fibrosis17,18, have shed light on carcinoma pathogenesis. Importantly, however, the transient activation of stromal cells observed during wound healing contrasts with the behaviours of these cells in tumours, where stromal cell recruitment and activity persist throughout the course of tumour development2,19,20.

In this Review, we discuss what is known about tumour–host interactions that reach beyond the boundaries of individual tumours to evoke systemic responses. So far, a handful of tumour-derived cytokines have been proven to act in a systemic fashion to affect distant tissues and, thereafter, to foster tumour growth. As this research field expands, this list is likely to grow longer. Here we examine how these tumour-derived factors and underlying pathophysiological processes play a part in nearly all aspects of cancer progression, starting with primary tumour growth and focusing particular attention on the growing body of evidence that tumour-driven systemic perturbations can also influence disease recurrence by governing some of the critical, rate-limiting steps in the metastatic process. Finally, we discuss how these emerging paradigms of systemic interactions may guide new areas of research on cancer pathogenetic mechanisms and therapeutic strategies designed to prevent disease relapse.

Tumour-driven systemic processes that affect primary tumour growth

Tumour cells secrete chemokines that act by recruiting various peripheral blood cells from the circulation into the TME. The repertoire of host circulating cells and the release of tumour-derived chemokines that recruit them into the primary TME remain areas of active investigation (reviewed in refs 1,2,2125). These findings have given rise to the notion that the recruitment of many of the stromal cell types that are found within the TME of most primary adenocarcinomas (Fig. 1a) involves the release of tumour-derived factors that may also mobilize host cells from distant tissues, notably the bone marrow and spleen26. Ultimately, these systemic perturbations impinge on cancer progression when resulting circulating cells are recruited into the tumour-associated stroma (Fig. 1b and Table 1).

Figure 1.

Figure 1

Composition of local and systemic tumour environments. (a) Cell types that comprise tumour tissues and play a role in cancer pathogenesis are depicted, including cancer cells, normal host cells and tumour-derived and/or -entrained cells. Some of the cells that operate within the tumour microenvironment originate from distant sources, including bone marrow and spleen; these cells are represented in areas of overlap between the indicated tissues. Also represented are other normal tissue cells, circulating cells and cell-free particles that are known to be modulated in response to malignancy. Some groups of cells (for example, bone-marrow-derived cells) are illustrated as a collection of differently coloured objects to represent their heterogeneity. This illustration also serves as a key to Figs 2 and 3. (b) Some of the tumour-derived cytokines (green text and differently coloured small particles) and microvesicles that are known to play a necessary role in mobilizing tumour-supportive host cells from tissues at distant anatomical sites. In some cases, these host cells are rendered pro-tumorigenic even prior to their mobilization. In most cases, the precise mechanism of action of these tumour-derived cytokines is not completely understood. Ultimately, these tumour-driven systemic events facilitate the growth of the initiating tumour. Solid arrows indicate activation or translocation of cell types and molecules; dashed arrows represent secretion of molecules.

Table 1.

Summary of known tumour-derived factors that act systemically

Initiating source Systemic factor Distant target tissue Effect on initiating tumour Effect on pre-metastatic tissues Effect on disseminated tumours
T G-CSF Bone marrow: myeloid cells34,122; endothelial progenitor cells122 Promote angiogenesis34,122; Confer resistance to angiogenesis inhibitors31 Promote92 or inhibit93 metastatic seeding in lung Promote94 or inhibit93 metastatic colonization
T VEGF-A Bone marrow: haemangiogenic cells38; myeloid cells38,71; VEGFR1+ cells7173 Lung: tissue parenchyma70 Promote angiogenesis38 Upregulate chemotactic proteins and activate MMPs in lung; BMDCs form pre-metastatic niches7073
T PIGF Bone marrow: haemangiogenic cells38; VEGFR1+ cells72 Lung: tissue parenchyma72 Promote angiogenesis38 Upregulate chemotactic proteins; activate MMPs; BMDCs form pre-metastatic niches72
S (CAFs) SDF-1 Bone marrow: endothelial progenitor cells3941; haematopoietic progenitor cells41 Promote angiogenesis40
C, MV OPN Bone marrow: HPCs36,37; endothelial progenitor cells and myeloid cells65 Promote proliferation, invasion and angiogenesis65 Promote metastatic colonization36; Aid formation of metastatic TME36,37,95
T ANG-II Spleen: myeloid cells50; monocytes, granulocytes51; HSCs, monocytes, neutrophils52 Promote tumour growth and metastasis5053
T, U TGF-β Bone marrow: myeloid cells71 Lung: tissue parenchyma70,71 Remodel lung parenchyma to promote pre-metastatic niche formation71
T, U TNF-α Bone marrow: myeloid cells71 Lung: tissue parenchyma70,71 Remodel lung parenchyma to promote pre-metastatic niche formation71
C LOX Lung: tissue parenchyma75 Recruit myeloid cells to form pre-metastatic niche75
C MCP-1 Lung: tissue parenchyma76 Recruit BMDCs; form niche; suppress NK cells76
C Versican Bone marrow: myeloid cells77 Promote niche formation77
C Prosaposin Lung: tissue parenchyma80 Inhibit angiogenesis to form anti-metastatic niche80
C Endostatin Disseminated tumours: TME102 Inhibit angiogenesis102
C Angiostatin Disseminated tumours: TME102 Inhibit angiogenesis102
S (CAFs) GDF15 Disseminated tumours: tumour cells Promote migration and invasion106 Promote survival, proliferation106
C IGF-II Disseminated tumours: tumour cells Promote proliferation and survival108 Promote survival, proliferation108
T Various Circulating platelets: granule content9698 Release contents to aid vascularization and tumour growth96,97
MV, P Various Bone marrow: HPCs63,65 Lung: tissue parenchyma63,64 Recruit BMDCs to mediate tumour growth, angiogenesis and chemotherapeutic resistence65 Promote vascular leakiness in lung; ‘educate’ pro-angiogenic BMDCs63,64 Promote vascularization65
T Unidentified Bone marrow: VEGFR2+ pro-angiogenic cells96 Promote vascularization96

Shown are tumour-derived factors that act to affect: (1) the initiating tumour from which they were derived; (2) distant tissues before metastatic dissemination; or (3) colonization and progression of disseminated micrometastases at distant anatomical sites. C, cancer cells; S, stromal cells; CAF, cancer-associated fibroblast; T, tumour tissue (specific cellular source unspecified); MV, tumour-derived microvesicles; P, tumour-modulated platelets; U, detected in circulation in cancer-specific manner but source undetermined; BMDC, bone-marrow-derived cell; HSC, haematopoietic stem cell; HPC, haematopoietic progenitor cell; TME, tumour microenvironment; ND, not determined.

Tumour-derived factors that affect the bone marrow.

In the bone marrow, the binding of stromal-cell-derived factor-1α (SDF-1α, also known as CXCL12; expressed by bone marrow endothelium27) to its receptor CXCR4 (expressed by both human and mouse haematopoietic stem cells (HSCs)28) represents a critical axis in bone marrow retention and homing of haematopoietic stem and progenitor cells29. The inflammatory cytokine G-CSF (granulocyte-colony stimulating factor) is known to antagonize this interaction by acting to either mobilize cells into the circulation or to prevent their homing back to the bone marrow30. Hence, G-CSF is a critical modulator of bone marrow haematopoietic progenitor cell mobilization and homing as a part of both normal homeostasis and inflammation29,3133.

Studies using mouse models of melanoma, lymphoma, lung carcinoma and mammary carcinoma have shown that tumour-derived G-CSF induces mobilization of tumour-supporting cells from the bone marrow into the circulation34,35. In one of these studies, certain haematopoietic progenitor cells, defined as CD11b+/Gr1+ myeloid cells, were altered in the bone marrow before their mobilization into the circulation35, and when harvested directly from the bone marrow of tumour-bearing mice, expressed distinct sets of genes — including the G-CSF responsive gene, Bv8 prokineticin — known to regulate myeloid cell mobilization and angiogenesis34. Following their recruitment from the circulation into the TME, CD11b+/Gr1+ myeloid cells were found to facilitate tumour-associated angiogenesis and to render tumours refractory to inhibition by anti-VEGF (vascular endothelial growth factor) treatment34.

Other work has demonstrated that tumour-derived, systemically acting osteopontin (OPN), an inflammatory cytokine, is necessary for the pro-tumorigenic function of bone marrow cells (BMCs) that are recruited into the TME of certain breast tumour xenografts36,37. In mice bearing the OPN-secreting tumours, but not in those with tumours that lack OPN expression, Sca1+/cKit/CD45+ haematopoietic BMCs are rendered pro-tumorigenic at the functional and molecular levels in the bone marrow before their mobilization into the circulation37. The tumour-supportive functions of Sca1+/cKit/CD45+ BMCs were discovered when they were mixed with otherwise-indolent breast tumour cells and injected into mice. It was observed that the BMCs from mice carrying OPN-secreting xenografts facilitated the growth of the indolent breast tumours, whereas the corresponding BMCs from cancer-free mice or mice bearing OPN-deficient xenografts did not33.

Several other studies have revealed tumour-derived factors that are important systemic mediators of tumour angiogenesis through their effects on BMCs. Specifically, vascular endothelial growth factor-A (VEGF-A) and placental growth factor (PlGF) trigger release of haemangiogenic BMCs (a collection of endothelial progenitor cells (EPCs) and VEGFR-1-positive haematopoietic cells) from the bone marrow into the circulation23. Following their recruitment to tumour sites, these haemangiogenic populations may promote tumour neovascularization38, although the precise function of EPCs and whether they are necessary for promoting tumour blood vessel formation remain unresolved39. Cancer-associated fibroblasts (CAFs), which are often abundant in the TME of most carcinomas, have also been linked to tumour angiogenesis through systemic effects. CAFs were shown to release CXCL12 (SDF-1α) into the circulation in a breast tumour xenograft model40. Together with earlier results demonstrating that circulating CXCL12 mobilizes haematopoietic progenitor and stem cells from the bone marrow41, this work led to the notion that CAF-derived circulating CXCL12 triggers the release of progenitor cells into the circulation, thus leading to their subsequent recruitment into the TME to promote angiogenesis.

Several other bone-marrow-derived cell types, including a complex array of leukocytes, have been observed in tumour specimens from mice and human patients42, and many of the mechanisms that control their recruitment from the circulation into the TME by tumour-derived chemokines have been uncovered43. Nevertheless, it is not yet clear whether these various immune cell types (or their precursors) are directly mobilized from distant reservoirs into the circulation by tumour-derived cytokines, or whether certain tumours may merely take advantage of the host cells that travel through the peripheral circulation as a part of normal physiologic homeostasis. Similarly, certain mouse models have revealed that some TME cells, such as mesenchymal stem cells44,45, can have origins in the bone marrow, but whether cancers directly elicit their mobilization from that site is not known.

Tumour-derived factors that affect the spleen.

Populations of CD11b+/Gr1+ myeloid cells are significantly expanded in the bone marrow and spleens of cancer-bearing mice relative to cancer-free counterparts46,47, and their numbers are also increased in the circulation of cancer patients compared with cancer-free subjects48,49. Experiments using mouse models of mammary carcinoma expanded on these observations by demonstrating that when CD11b+/Gr1+ myeloid cells were isolated from the spleens of mice bearing mammary tumours and were mixed with tumour cells before implantation, tumour growth and metastasis were enhanced50. In contrast, equal numbers of antigenically identical myeloid cells from the spleens of tumour-free mice failed to alter tumour progression50. In these experiments, the CD11b+/Gr1+ myeloid cells helped to promote metastasis, at least in part, by expressing functional matrix metalloproteinases. However, although the pro-metastatic myeloid cells were specifically recruited to mammary carcinomas, it was unclear how these tumours communicated systemically with the spleen to induce their mobilization into the circulation.

Later studies using a mouse model of lung adenocarcinoma development suggest that tumour-derived angiotensinogen, the precursor molecule to the circulating peptide hormone angiotensin II, can function as a modulator of tumour-supporting cells in the spleen. One of these studies revealed that the majority of tumour-associated macrophages and neutrophils were derived from progenitor CD11b+/Ly-6CHigh monocytes and CD11b+/Ly-6GHigh granulocytic cells respectively, and that both populations were increased in the spleens of tumour-bearing mice51. Subsequent work showed that elevated levels of circulating angiotensin II were necessary for amplifying self-renewing HSCs and macrophage progenitors in the spleen, but not in the bone marrow, of lung tumour-bearing mice52,53. These studies suggested that angiotensin II was tumour-derived, as expression of angiotensinogen was readily detectable in the lung tumour cells.

In support of the pre-clinical findings discussed above, elevated levels of many of these cytokines have been detected in primary tumour samples as well as in the plasma of patients bearing various cancer types, including colon carcinoma, melanoma, hepatocellular carcinoma and breast cancer, and have been positively correlated with metastatic progression36,5460. As we discuss later, primary tumour-derived secreted factors may also have a profound impact on the outgrowth of disseminated metastatic colonies.

Circulating microvesicles.

The release of soluble cytokines into the circulation is not the only means by which primary tumours communicate with distant tissues. Tumour cells are known to extrude microvesicles, which are membrane-bound particles (including microparticles and exosomes ranging in size from 0.1–1 μm) that carry lipids, proteins, mRNAs and miRNAs61. Recent discoveries indicate that these tumour-derived microvesicles can modulate cancer progression by influencing the behaviour of host cells in distant tissues62. For example, exosomes derived from melanomas were proposed to ‘educate’ pro-metastatic progenitor cells in the bone marrow63. Likewise, renal-carcinoma-derived exosomes were found to promote angiogenesis in lung tumour metastases64. Additionally, a study of murine mammary carcinoma demonstrated that osteopontin, carried through the circulation by tumour-derived microparticles, was necessary to mobilize pro-angiogenic cells from the bone marrow65. Interestingly, in these experiments, the mammary tumours produced microparticles as a result of paclitaxel chemotherapy, which was consistent with the authors’ clinical observations that circulating microparticle numbers were elevated in some cancer patients following chemotherapy.

Although microvesicles have attracted much attention and have even been suggested as targets for cancer therapy, their cellular sources, target tissues and various roles in tumour pathogenesis are only beginning to be uncovered. The importance of these particles in systemic signalling is supported by the finding that their circulating levels in tumour-bearing hosts seem to be higher than the levels observed in cell culture experiments where microvesicle-induced effects have been clearly observed65.

Tumour-driven systemic processes that alter distant tissues before metastasis

Tumour metastasis can be portrayed as the end-product of a multi-step succession of events termed the invasion–metastasis cascade, which has been extensively reviewed elsewhere6669. To disseminate, cancer cells from the primary tumour must first invade the local tissue parenchyma of the primary tumour and intravasate into nearby microvessels to enter into the circulation. Following haematogenous dissemination, tumour cells can be trapped in the microvessels of distant tissues, where they may exit the circulation via the process termed extravasation, to enter into the tissue parenchyma. The final step in this cascade, which we discuss in more detail later, is defined operationally as the acquired ability of micrometastatic deposits to form macroscopic metastases — a process often termed colonization. In some cases, primary-tumour-driven systemic processes that occur before metastasis have been found to dictate the site where subsequently disseminated cancer cells extravasate into tissue parenchyma (Fig. 2a and Table 1).

Figure 2.

Figure 2

Mechanisms of systemic regulation of metastatic tumour cell extravasation and colonization. (a) ‘Pre-metastatic niches’ are thought to form as a consequence of tumour-derived systemic factors that alter the tissue parenchyma of organs at distant sites, for example by extracellular matrix remodelling and activating resident tissue fibroblasts, as well as mobilizing bone-marrow-derived cells that subsequently incorporate into the parenchyma of these tissues. These niches are thus amenable to recruitment and extravasation of circulating metastatic tumour cells. Importantly, some tumour models have also revealed inhibitory factors that accumulate at pre-metastatic sites. (b) Circulating factors and bone-marrow-derived cells can affect disseminated micrometastases that would otherwise remain indolent, thus promoting their subsequent colonization into macroscopic metastases. For example, some reports have demonstrated promotion of vascularization by recruitment of pro-angiogenic bone-marrow-derived cells and platelets. Conversely, primary-tumour-derived anti-angiogenic factors have been reported to inhibit outgrowth of distant metastases. Other reports have revealed molecular mechanisms by which metastatic colonization is promoted by bone-marrow-derived cells that directly promote tumourcell proliferation and/or activation of tissue fibroblasts. Processes that affect colonization are represented in blue text, pro-tumorigenic factorsare represented in green text, and tumour inhibitory factors are shown in red text. Solid arrows indicate activation or translocation of cell types and molecules and dashed arrows represent secretion of molecules. For labelling key of cell types, see Fig. 1a.

Initial evidence that the pre-metastatic tissues of tumour-bearing animals are altered in the presence of distantly located tumours came from studies using subcutaneously implanted murine melanoma and lung carcinomas70,71. This work indicated that tumour-derived VEGF-A, TGF-β and TNF-α induced changes in the lung microenvironment that subsequently made the lung tissue more competent at recruiting circulating CD11b+ myeloid cells in a VEGFR1-dependent manner. The same group showed that the extracellular matrix-remodelling metalloproteinase, MMP9, is expressed in the lungs both by resident endothelial cells and by recruited CD11b+ cells, resulting in significantly more metastatic tumour cells being recruited to the lungs of tumour-bearing mice; they also showed that the metastasis-free lungs from cancer patients exhibit enhanced MMP9 expression70.

A study using the same mouse models of subcutaneously implanted metastatic melanoma and lung carcinoma showed that tumour-derived VEGF-A and PlGF could mobilize VEGFR1+ BMCs, which subsequently localized not only to primary tumour sites, but also to the lungs before dissemination of tumour cells from the primary site72. At the same time, lung fibroblasts upregulated expression of fibronectin at these sites, thus facilitating recruitment of VEGFR1+4β1-integrin+ 72, These sites within the lung tissue were termed pre-metastatic niches72, and were locations where disseminated cancer cells could eventually be detected. However, it was unclear whether other sites within the lung could also provide microenvironments favouring the foundation and outgrowth of metastases, or whether VEGFR1+ cells are necessary for the formation of these specific niches73. Results from more recent studies have uncovered additional tumour-derived factors and mechanisms by which myeloid cells accumulate in pre-metastatic lung tissue to influence the subsequent recruitment of disseminated metastatic cells from a variety of primary tumour types (reviewed in ref. 74). For example, a study of breast cancer human xenografts and syngeneic mouse models indicated that accumulation of tumour-secreted lysyl oxidase (LOX) recruits bone-marrow-derived myeloid cells that establish a pre-metastatic niche in the lungs75.

Since the first descriptions of pre-metastatic niches, more circulating tumour-derived factors that alter lung tissue have been identified, including the inflammatory cytokine MCP1 (monocyte chemotactic protein 1; also known as CCL2)76 and the proetoglycan versican (VCAN)77. At present, the direct targets of these tumour-derived factors (for example, bone marrow cells or host cells in the distant tissue that is the eventual site of metastasis) are not clear78. The role of these factors in normal physiological processes might provide clues to this unanswered question. For example, mobilization of inflammatory monocytes is thought to be modulated by MCP-1 and its receptor, CCR2 (ref. 79).

In contrast to the tumour-supportive lung microenvironments described above, one study demonstrated that certain poorly metastatic primary breast tumours inhibit the growth of future metastases by establishing ‘anti-metastatic niches’ in the lungs80. In this study, primary breast tumours secreted the cytokine prosaposin into the circulation, resulting in enhanced expression of thrombospondin, an anti-angiogenic factor, in distant lung fibroblasts.

The currently available information about pre-metastatic niches leaves open two important questions. First, although experimental metastasis models have demonstrated pre-metastatic niches in organs such as adrenal glands77, liver81 and lymph nodes82, it is not yet clear whether such perturbations are most frequently observed in the lung because it is simply the most-studied site in experimental metastasis models, or whether it is an organ that is particularly predisposed to inflammation. The potentially unique status of the lungs is suggested by a recent report demonstrating that allergy-induced inflammation caused the lungs to serve as a target tissue for metastasis83. Second, as many cancer patients experience bone metastases (particularly those with metastatic breast or prostate cancer84), and as most patients display disseminated tumour cells in the bone marrow even at early stages of disease85,86, it is plausible that similar tumour-supportive niches are also formed in the bone marrow. The fact that various cancer types can elicit tumour-specific systemic effects in the bone microenvironment from a distance (as discussed previously) lends some support to this notion.

A recent study suggests that these considerations are worth further exploration: in a mouse model of arthritis, the systemic inflammatory effects significantly increased breast-cancer-associated metastases to both lungs and bones in arthritic compared to non-arthritic mice87. The pre-metastatic lungs of arthritic mice were highly inflamed, and use of an anti-interleukin (IL)-17 antibody significantly reduced inflammation and metastatic burden in both lungs and bone.

Tumour-driven systemic processes that affect metastatic colonization

The final step in the metastatic cascade involves metastatic colonization — the outgrowth of micrometastatic deposits into overt tumours. Colonization in itself is often viewed as a series of obstacles that disseminated tumour cells must overcome to form macroscopic metastases, such as: remodelling the extracellular microenvironment, gaining access to local trophic growth factors, avoiding immune attack, acquiring a blood supply and forming a tumour-supportive TME (ref. 66). The vast majority of extravasated tumour cells are unable to form a successful tumour and thus remain dormant and eventually disappear88,89. Hence, metastatic colonization is thought to be the rate-limiting step in the successful generation of metastases; indeed, some studies have demonstrated that far fewer than 1% of disseminated tumour cells succeed in forming a macrometastatic growth85,86.

In certain cases, as we discuss in detail below, disseminated tumour cells that would otherwise remain dormant can succeed in colonizing distant tissues in response to changes in the host systemic environment triggered by a distantly located tumour (Fig. 2b and Table 1). Thus, the critical determinants of whether or not a population of disseminated tumour cells succeeds in generating clinically significant metastases may include not only the intrinsic properties of the cancer cells themselves but also the state of the host environment, including both the local tissue microenvironment and the systemic physiological environment (Fig. 3).

Figure 3.

Figure 3

Tumour-driven pathophysiological processes underlying cancer progression. Tumour-mediated events are depicted in space and time in a counter-clockwise fashion beginning with the initiating tumour. Although this cascade of events is represented in a unified fashion, it is clear that tumours from different cancer types employ different systemic tumour-promoting mechanisms that thus far seem to be context- and tissue-specific. In some cases, initiating tumours secrete cytokines into the circulation that cause mobilization of haematopoietic cells from distant organs; these cells can then contribute to pre-metastatic niche formation and/or promote metastatic colonization. In other cases, initiating-tumour-derived cytokines seem to directly affect growth of metastatic colonies at distant anatomical sites. In yet other cases, initiating-tumour-derived cytokines can be carried by circulating microvesicles or platelets, which serve as transporters of these cytokines to distant tissues or metastatic sites. Processes are represented in blue text, pro-tumorigenic factors are represented in green text, and tumour inhibitory factors are shown in red text. Solid arrows indicate activation or translocation of cell types and molecules and dashed arrows represent secretion of molecules. For labelling key of cell types see Fig. 1a.

Impact of systemic regulation of immune function on metastatic colonization

Early experimental models of fibrosarcoma suggested that certain aggressively growing tumours attenuate the anti-tumour immune response of the host, thereby creating a tumour-permissive systemic environment90. In these studies, certain fibrosarcoma cells were found to grow well in immune-deficient mice but were frequently rejected in immune-competent mice. However, these fibrosarcoma cells could grow aggressively and form tumours with short latency when implanted into immune-competent mice bearing already-established fibrosarcomas at distant sites, suggesting immune tolerance in this class of tumour-bearing hosts. A more recent example of this mechanism of systemically induced immunological tolerance comes from work using the transgenic HER2/neu model of mouse mammary carcinogenesis91.

Research using the 4T1 mouse model of murine mammary adeno-carcinoma revealed that tumour-derived G-CSF mobilized neutrophils into the circulation, which played a pro-metastatic role by creating an immunosuppressive microenvironment at metastatic sites92. However, the influence of this pro-metastatic mechanism on the success of lung colonization by 4T1 mouse breast cancer cells remains unresolved in light of other work that used the same mouse model to show that although tumour-derived G-CSF induced neutrophil mobilization into the circulation, these neutrophils instead played an anti-meta-static role93. Specifically, mammary-tumour-derived CCL2 rendered G-CSF-mobilized neutrophils cytotoxic, such that after accumulating in the lungs, they inhibited metastatic colonization. Such cytotoxic tumour-entrained neutrophils were also found in the peripheral blood of breast cancer patients before surgical resection, but not in cancer-free individuals93. It is possible that the particular cell-surface markers used to characterize and isolate neutrophil populations in these studies account for the differences in observed outcomes. Insights into these considerations are provided by experiments showing that among the Gr1+ cells that were recruited to the premetastatic lungs, the monocytic and not the neutrophil subpopulation contributed to metastatic colonization94. These findings point to the fact that the heterogeneity and plasticity of certain haematopoietic populations, particularly neutrophils, are an important consideration when studying tumour-modulating systemic events.

Systemic effects on angiogenesis at metastatic sites

Studies of a form of pro-angiogenic signalling that takes place between pairs of distantly located tumours (a process that was termed systemic instigation36) revealed different mechanisms of systemic influence on metastatic colonization23,89,95. One set of experiments using xenografted cell lines and primary tumour specimens from patients with luminal breast cancer or clear cell renal cell carcinoma showed that platelets act as a long-range delivery system between aggressively growing tumours and otherwise-indolent clusters of tumour cells at distant sites96. In this study, certain human luminal breast cancers (LBCs) mobilized proangiogenic VEGFR2+ BMCs from the bone marrow and secreted proangiogenic cytokines that were taken up by circulating platelets. When otherwise-indolent breast tumour cells or primary renal cell carcinoma specimens, which were unable to undergo angiogenesis or proliferate on their own, were implanted at sites distant from these LBC tumours, pro-angiogenic BMCs and platelets converged on them, promoting their growth into highly vascularized, rapidly proliferating tumours. In these experiments, circulating platelets produced by mice bearing the human LBC tumour xenografts were found to carry bioactive human cytokines, which included VEGF, TGF-β1, PDGF-BB, PlGF, IL-6 and CXCL1, among others96. This finding is in agreement with earlier studies demonstrating that platelets are capable of taking up factors secreted by breast tumour cells in vitro97 and glioma and prostate tumour cells in vivo98. Although this work showed that the cognate receptors for some of these signalling molecules were activated within the TME (ref. 96), it has not yet been established conclusively that the bioactive cytokines acquired in primary tumours by the platelets are delivered to distantly located nests of tumour cells. Thus, although a large body of literature has described the importance of platelets during tissue remodelling and wound healing99, the selective release of platelet bioactive cargo100 and the ability of platelets to help cancer cells to metastasize101, their role as conveyors of systemic signals is only beginning to be appreciated. In contrast to these pro-angiogenic systemic mechanisms, early studies using mouse cell lines and human tumour cell line xenografts indicated that certain primary tumours can suppress the outgrowth of distant metastases by secreting anti-angiogenic factors, such as endostatin and angiostatin102.

Systemic promotion of the metastatic tumour microenvironment

As described, certain triple-negative breast cancers release OPN into the circulation to render haematopoietic cells in the bone marrow protumorigenic36. These activated BMCs were capable of travelling not only to primary tumours, but also to otherwise-indolent tumours at distant sites, and to weakly metastatic disseminated tumour cells in the lungs, thereby acting as the primary mediators of systemic instigation. Subsequent research demonstrated that these pro-tumorigenic BMCs secreted elevated levels of the growth factor granulin (GRN)103 in the marrow before their mobilization as well as within the tumour microenvironment following their recruitment to microscopic nests of tumour cells37,104. After entering the TME, GRN activated resident mam-mary fibroblasts, causing them to adopt a CAF phenotype associated with expression of pro-inflammatory and matrix-remodelling genes that further support tumour progression37. A complementary study using a mouse model of squamous skin carcinogenesis demonstrated that CAFs expressing many of the same factors (including CXCL2, IL-6, IL-1β, IL-8, Cox-2, CCL2 and CXCL3) promoted the recruitment of circulating inflammatory cells to the tumour site105. Ultimately this sequence of events results in the outgrowth of malignant tumours with highly desmoplastic stromata.

Alterations in the TME by distant tumours may also be reflected in the spectrum of mitogenic and trophic factors that neoplastic cells harvest from their nearby environment. Specifically, systemic signals released by aggressively growing breast tumour xenografts caused the formation of a tumour-supportive microenvironment enriched in epidermal growth factor (EGF) and insulin-like growth factor-1 (IGF-1) at distant anatomical sites where clusters of otherwise-indolent tumour cells already resided104. These indolent tumour cells were only able to form proliferating tumours when both EGF and IGF-1 were made available to them as a result of systemic instigation, which was prevented by treating the mice with a combination of EGFR and IGF-1R inhibitors.

A recent report has exposed CAFs as a previously unrecognized source of systemic instigation of disease progression through gene expression analyses of micro-dissected normal and cancer human prostate tissue that revealed significant upregulation of growth/differentiation factor 15 (GDF15; also known as macrophage inhibitory cytokine-1, MIC-1), a member of the TGF-β family of growth factors, in the cancer-specific TME relative to the normal tissue stroma106. CAFs derived from a mouse model of prostate carcinoma, as well as normal fibroblasts in which GDF15 was ectopically expressed, promoted tumour growth when directly admixed with otherwise-indolent prostate carcinoma cells and implanted into mice106. Elevated plasma levels of GDF15 that had been previously observed in certain cancer patients107 and in the experimental mice being studied106 prompted further experiments that demonstrated GDF15-expressing CAFs implanted into one site in the mice promoted the outgrowth of distantly implanted, indolent prostate cancer cells. Although the underlying assumption of these findings is that the disseminated prostate tumour cells benefit from the effects of circulating GDF15 due to its effect on cell proliferation and migration, the direct target(s) of GDF15 in this systemic promotion of metastatic colonization remain unknown.

Metastatic colonization of the lung was also recently reported to occur as a result of systemic instigation in an experimental xenograft model of oesophageal cancer, for which IGF-II (insulin-like growth factor-II) is necessary108. In this study, an IGF-II-secreting oesophageal tumour xenograft could significantly enhance the growth of a distantly implanted tumour, as well as promote metastatic colonization of cancer cells that had disseminated to the lungs. Treating the mice with the IGF-I/IGF-II receptor inhibitor cixutumumab had significant suppressive effects on tumour growth and metastasis. Although the direct target of primary-tumour-derived systemic IGF-II is assumed to be the disseminated tumour cells due to the effects of IGF-II on AKT-dependent tumour cell survival, this hypothesis was not unequivocally proven.

Other systemic processes for consideration

Early considerations of cancer as a systemic disease raised the question of whether the observed manifestations of systemic involvement were an integral part of tumour development or whether they were nothing more than epiphenomena of malignancy — paraneoplastic conditions that had little, if any, effect on tumour growth. Recent results however, suggest that conditions that were once thought to be paraneoplastic syndromes may, in ways that are still unclear, directly benefit the tumours that generated them. The evidence in these cases is still equivocal. The frequently observed dysfunctional immune responses109 and neuropathies110 in cancer patients may well support the expansion of established tumours through mechanisms that remain poorly resolved. More detailed mechanistic understanding has emerged from studies of the pathologic elevation of circulating platelet counts, termed thrombocytosis, which is observed in patients and is associated with reduced patient survival111,112. A recent study of an ovarian cancer experimental model revealed certain mechanisms by which cancer-associated thrombocytosis occurs: hepatic thrombopoietin (TPO) synthesis was enhanced in response to elevated ovarian carcinoma-derived IL-6 in the circulation. This circulating TPO impinged on bone marrow megakaryocytes, resulting in acceleration of the rate of platelet production112 (Fig. 1b). Moreover, therapeutic targeting of platelets using an anti-platelet antibody resulted in reduced tumour growth and angiogenesis in experimental mice, and tumour-derived IL-6 and hepatic TPO were linked to thrombocytosis in a large cohort of ovarian cancer patients.

The various systemic mechanisms by which incipient tumours can progress to a malignant state in response to systemic instigation raise the question of whether other systemic states exist in which neoplastic disease progression can be modulated in the absence of an instigating tumour. For example, epidemiological evidence indicates that obesity contributes to heightened incidence of multiple cancer types113,114. Pre-clinical evidence exists in support of the idea that systemic changes associated with pregnancy can affect the outgrowth of incipient breast tumours115. Finally, a number of studies suggest that the as-yet-undefined systemic effects of surgery can trigger outgrowth of dormant metastases116,117.

Clinical implications and future perspectives

The realization that cancer is often, if not invariably, a systemic disease suggests that a variety of therapeutic interventions could be designed to inhibit or interdict the signalling mechanisms that underlie systemic changes in cancer patients. Although strategies targeting primary malignancies have improved markedly, there are no cures for metastatic disease, which remains the underlying cause of death for the majority of cancer patients. If a broad spectrum of metastases are eventually found to depend on systemic signalling, a new avenue of therapy development would be opened, as, in theory, the relevant systemic signals travelling through the circulation should be more susceptible to interdiction (for example, by a small-molecule inhibitor or neutralizing antibody) than the paracrine and juxtacrine signals that operate within the interstices of solid tumours.

Independently of their vulnerability to intervention, these systemic signalling mechanisms hold implications for various types of existing cancer therapies and treatment modalities. For example, recombinant G-CSF is often administered to bone marrow donors, transplant patients and some cancer patients undergoing myelosuppressive chemotherapy, with the intention of mobilizing their haematopoietic stem and progenitor cells into the circulation118. However, as discussed earlier, pre-clinical evidence indicates that G-CSF mobilizes into the circulation BMCs that are capable of promoting disease progression both at the primary tumour site and in the metastatic setting. Active investigation into the clinical use of haematopoietic growth factors, such as G-CSF and a related haematopoietic stimulating factor, GM-CSF, is ongoing.

Anti-angiogenic therapies, which are used in the oncology clinic, serve as a second example. A compelling pre-clinical study showed that when tumour-free mice were treated with anti-angiogenic therapy, plasma levels of a number of the systemic tumour-promoting factors enumerated earlier (including VEGF, SDF-1α, G-CSF and OPN, among others) were elevated in a dose-dependent fashion compared with those of untreated mice, implying that these treatments used to inhibit tumour growth might actually promote it119. Similarly, other pre-clinical studies have shown that certain chemotherapies and anti-VEGF treatments promote mobilization of cells from the bone marrow that are capable of enhancing tumour growth120122.

Findings such as these have caused some to propose that host ‘systemic conditioning’ provides an explanation for why certain patients are predisposed to the outgrowth of certain cancers, as in the case of radiation treatment123, or to disease relapse, as in the case of certain chemotherapies124. At present it is difficult to know whether or not these treatments, which are designed to benefit cancer patients, may instead stimulate tumour growth.

The ability to identify tumours that are capable of eliciting a host systemic response as well as tumours that can respond to changes in the host systemic environment will carry significant implications for future risk stratification strategies. For example, testing tumour tissues for their ability to alter the systemic environment in pre-clinical models may allow for more accurate identification of patients with a high likelihood of future relapse. Ultimately, the goal of such studies is to devise tests that will allow oncologists to more accurately identify patients who may benefit from specific adjuvant therapies at a time when the progression to overt metastatic disease can still be prevented or at least significantly delayed.

The potential for defining a tumour-specific systemic environment is also leading to a search for biomarkers of malignancy. The studies described above suggest that systemic cellular and molecular signals play a role in driving malignancy, and that a better understanding of these signals may lead to the development of sensitive prognostic tests. For example, recent evidence supports the idea that an increased ratio of circulating neutrophils versus lymphocytes, previously known to be an indicator of systemic inflammation, seems to function as an outcome determinant in patients with various types of cancer, particularly advanced colon and pancreatic cancers, and that this ratio can be used to predict survival and stratify treatment125127. Recently, the neutrophil-tolymphocyte ratio used in combination with elevated platelet count was found to be predictive of the future clinical course of colorectal cancer128. The Glasgow Prognostic Score, which measures circulating C-reactive protein and albumin, is another measure of systemic inflammation that has been in use for over a decade and has independent prognostic value for cancer patients in a variety of clinical scenarios129.

These types of measurements would seem to represent the tip of a far larger iceberg: as the list of tumour-derived cytokines, unique circulating pro-tumorigenic haematopoietic cells and platelets and tumour-derived cell-free particles such as microvesicles expands, so will the opportunities to discover novel biomarkers that are highly useful for patient diagnosis and prognosis.

ACKNOWLEDGEMENTS

All figures were conceptualized and created by Victor Fanjul (Universidad de Oviedo, Spain; former summer intern in the McAllister lab). We are grateful for the helpful discussions and/or editorial comments of Zvika Granot, Mikael Pittet and Yuval Shaked. S.S.M. is an American Cancer Society Scholar, an AACR Gertrude B. Elion Cancer Research Scholar and a Presidential Early Career Award for Scientists and Engineers scholar. R.A.W. is a Daniel K. Ludwig Professor for Cancer Research at MIT and an American Cancer Society Research Professor. This work was supported in part by grants from the National Institutes of Health (NCI) RO1 CA166284 and the American Cancer Society (S.S.M.); the Breast Cancer Research Foundation (BCRF), National Institutes of Health (NIH), R01 CA078461, P01 CA080111, and the Ludwig Center for Molecular Oncology at MIT (R.A.W.)

Footnotes

COMPETING FINANCIAL INTERESTS

The authors declare no competing financial interests.

References

  • 1.Joyce JA & Pollard JW Microenvironmental regulation of metastasis. Nat. Rev. Cancer 9, 239–252 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Pietras K & Ostman A Hallmarks of cancer: interactions with the tumor stroma. Exp. Cell Res 316, 1324–1331 (2010). [DOI] [PubMed] [Google Scholar]
  • 3.Allinen M et al. Molecular characterization of the tumor microenvironment in breast cancer. Cancer Cell 6, 17–32 (2004). [DOI] [PubMed] [Google Scholar]
  • 4.Chang HY et al. Gene expression signature of fibroblast serum response predicts human cancer progression: similarities between tumors and wounds. PLoS Biol. 2, E7 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Finak G et al. Gene expression signatures of morphologically normal breast tissue identify basal-like tumors. Breast Cancer Res. 8, R58 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Casey T et al. Molecular signatures suggest a major role for stromal cells in development of invasive breast cancer. Breast Cancer Res. Treat 114, 47–62 (2009). [DOI] [PubMed] [Google Scholar]
  • 7.Ma XJ, Dahiya S, Richardson E, Erlander M & Sgroi DC Gene expression profiling of the tumor microenvironment during breast cancer progression. Breast Cancer Res. 11, R7 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Zajicek G Cancer as a systemic disease. Med. Hypotheses 4, 193–207 (1978). [DOI] [PubMed] [Google Scholar]
  • 9.Redig AJ & McAllister SS Breast cancer as a systemic disease: a view of metastasis. J. Intern. Med 274, 113–126 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Dvorak HF Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N. Engl. J. Med 315, 1650–1659 (1986). [DOI] [PubMed] [Google Scholar]
  • 11.Arwert EN, Hoste E & Watt FM Epithelial stem cells, wound healing and cancer. Nat. Rev. Cancer 12, 170–180 (2012). [DOI] [PubMed] [Google Scholar]
  • 12.Coussens LM & Werb Z Inflammation and cancer. Nature 420, 860–867 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.De Visser KE, Eichten A & Coussens LM Paradoxical roles of the immune system during cancer development. Nat. Rev. Cancer 6, 24–37 (2006). [DOI] [PubMed] [Google Scholar]
  • 14.Mantovani A, Allavena P, Sica A & Balkwill F Cancer-related inflammation. Nature 454, 436–444 (2008). [DOI] [PubMed] [Google Scholar]
  • 15.Grivennikov SI, Greten FR & Karin M Immunity, inflammation, and cancer. Cell 140, 883–899 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Trinchieri G Cancer and inflammation: an old intuition with rapidly evolving new concepts. Annu. Rev. Immunol 30, 677–706 (2012). [DOI] [PubMed] [Google Scholar]
  • 17.Kalluri R Basement membranes: structure, assembly and role in tumour angiogenesis. Nat. Rev. Cancer 3, 422–433 (2003). [DOI] [PubMed] [Google Scholar]
  • 18.Lech M & Anders HJ Macrophages and fibrosis: How resident and infiltrating mononuclear phagocytes orchestrate all phases of tissue injury and repair. Biochim. Biophys. Acta 1832, 989–997 (2013). [DOI] [PubMed] [Google Scholar]
  • 19.Polyak K, Haviv I & Campbell IG Co-evolution of tumor cells and their microenvironment. Trends Genet. 25, 30–38 (2009). [DOI] [PubMed] [Google Scholar]
  • 20.Marsh T, Pietras K & McAllister SS Fibroblasts as architects of cancer pathogenesis. Biochim. Biophys. Acta 1832, 1070–1078 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Bissell MJ & Radisky D Putting tumours in context. Nat. Rev. Cancer 1, 46–54 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Seruga B, Zhang H, Bernstein LJ & Tannock IF Cytokines and their relationship to the symptoms and outcome of cancer. Nat. Rev. Cancer 8, 887–899 (2008). [DOI] [PubMed] [Google Scholar]
  • 23.Gao D & Mittal V The role of bone-marrow-derived cells in tumor growth, metastasis initiation and progression. Trends Mol. Med 15, 333–343 (2009). [DOI] [PubMed] [Google Scholar]
  • 24.Hanahan D & Coussens LM Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell 21, 309–322 (2012). [DOI] [PubMed] [Google Scholar]
  • 25.Cuiffo BG & Karnoub AE Mesenchymal stem cells in tumor development: emerging roles and concepts. Cell Adh. Migr 6, 220–230 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Shaked Y, McAllister S, Fainaru O & Almog N Tumor dormancy and the angiogenic switch: possible implications of bone marrow-derived cells. Curr. Pharm. Des 19, 1–14 (2014). [DOI] [PubMed] [Google Scholar]
  • 27.Eash KJ, Greenbaum AM, Gopalan PK & Link DC CXCR2 and CXCR4 antagonistically regulate neutrophil trafficking from murine bone marrow. J. Clin. Invest 120, 2423–2431 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Martin C et al. Chemokines acting via CXCR2 and CXCR4 control the release of neutrophils from the bone marrow and their return following senescence. Immunity 19, 583–593 (2003). [DOI] [PubMed] [Google Scholar]
  • 29.Wright DE, Wagers AJ, Gulati AP, Johnson FL & Weissman IL Physiological migration of hematopoietic stem and progenitor cells. Science 294, 1933–1936 (2001). [DOI] [PubMed] [Google Scholar]
  • 30.Kondo M et al. Biology of hematopoietic stem cells and progenitors: implications for clinical application. Annu. Rev. Immunol 21, 759–806 (2003). [DOI] [PubMed] [Google Scholar]
  • 31.Christopher MJ, Rao M, Liu F, Woloszynek JR & Link DC Expression of the G-CSF receptor in monocytic cells is sufficient to mediate hematopoietic progenitor mobilization by G-CSF in mice. J. Exp. Med 208, 251–260 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Chow A et al. Bone marrow CD169+ macrophages promote the retention of hematopoietic stem and progenitor cells in the mesenchymal stem cell niche. J. Exp. Med 208, 261–271 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Winkler IG et al. Bone marrow macrophages maintain hematopoietic stem cell (HSC) niches and their depletion mobilizes HSCs. Blood 116, 4815–4828 (2010). [DOI] [PubMed] [Google Scholar]
  • 34.Shojaei F et al. Tumor refractoriness to anti-VEGF treatment is mediated by CD11b+Gr1+ myeloid cells. Nat. Biotechnol 25, 911–920 (2007). [DOI] [PubMed] [Google Scholar]
  • 35.Shojaei F et al. Bv8 regulates myeloid-cell-dependent tumour angiogenesis. Nature 450, 825–831 (2007). [DOI] [PubMed] [Google Scholar]
  • 36.McAllister SS et al. Systemic endocrine instigation of indolent tumor growth requires osteopontin. Cell 133, 994–1005 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Elkabets M et al. Human tumors instigate granulin-expressing hematopoietic cells that promote malignancy by activating stromal fibroblasts in mice. J. Clin. Invest 121, 784–799 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Kopp HG, Ramos CA & Rafii S Contribution of endothelial progenitors and proangiogenic hematopoietic cells to vascularization of tumor and ischemic tissue. Curr. Opin. Hematol 13, 175–181 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Purhonen 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. USA 105, 6620–6625 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Orimo A et al. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell 121, 335–348 (2005). [DOI] [PubMed] [Google Scholar]
  • 41.Hattori K et al. Plasma elevation of stromal cell-derived factor-1 induces mobilization of mature and immature hematopoietic progenitor and stem cells. Blood 97, 3354–3360 (2001). [DOI] [PubMed] [Google Scholar]
  • 42.DeNardo DG et al. Leukocyte complexity predicts breast cancer survival and functionally regulates response to chemotherapy. Cancer Discov. 1, 54–67 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Anders HJ, Romagnani P & Mantovani A Pathomechanisms: homeostatic chemokines in health, tissue regeneration, and progressive diseases. Trends Mol. Med 20, 154–165 (2014). [DOI] [PubMed] [Google Scholar]
  • 44.Karnoub AE et al. Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature 449, 557–563 (2007). [DOI] [PubMed] [Google Scholar]
  • 45.Kidd S et al. Direct evidence of mesenchymal stem cell tropism for tumor and wounding microenvironments using in vivo bioluminescent imaging. Stem Cells 27, 2614–2623 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Melani C, Chiodoni C, Forni G & Colombo MP Myeloid cell expansion elicited by the progression of spontaneous mammary carcinomas in c-erbB-2 transgenic BALB/c mice suppresses immune reactivity. Blood 102, 2138–2145 (2003). [DOI] [PubMed] [Google Scholar]
  • 47.Serafini P, Borrello I & Bronte V Myeloid suppressor cells in cancer: recruitment, phenotype, properties, and mechanisms of immune suppression. Semin. Cancer Biol 16, 53–65 (2006). [DOI] [PubMed] [Google Scholar]
  • 48.Almand B et al. Increased production of immature myeloid cells in cancer patients: a mechanism of immunosuppression in cancer. J. Immunol 166, 678–689 (2001). [DOI] [PubMed] [Google Scholar]
  • 49.Young MR, Wright MA, Vellody K & Lathers DM Skewed differentiation of bone marrow CD34+ cells of tumor bearers from dendritic toward monocytic cells, and the redirection of differentiation toward dendritic cells by 1alpha, 25-dihydroxyvitamin D3. Int. J. Immunopharmacol 21, 675–688 (1999). [DOI] [PubMed] [Google Scholar]
  • 50.Yang L et al. Abrogation of TGF beta signaling in mammary carcinomas recruits Gr-1+CD11b+ myeloid cells that promote metastasis. Cancer Cell 13, 23–35 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Swirski FK et al. Identification of splenic reservoir monocytes and their deployment to inflammatory sites. Science 325, 612–616 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Cortez-Retamozo V et al. Origins of tumor-associated macrophages and neutrophils. Proc. Natl Acad. Sci. USA 109, 2491–2496 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Cortez-Retamozo V et al. Angiotensin II drives the production of tumor-promoting macrophages. Immunity 38, 296–308 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Ugurel S, Rappl G, Tilgen W & Reinhold U Increased serum concentration of angiogenic factors in malignant melanoma patients correlates with tumor progression and survival. J. Clin. Oncol 19, 577–583 (2001). [DOI] [PubMed] [Google Scholar]
  • 55.Holzer G et al. Concentration of vascular endothelial growth factor (VEGF) in the serum of patients with malignant bone tumors. Med. Pediatr. Oncol 36, 601–604 (2001). [DOI] [PubMed] [Google Scholar]
  • 56.Poon RT et al. Serum vascular endothelial growth factor predicts venous invasion in hepatocellular carcinoma: a prospective study. Ann. Surg 233, 227–235 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Van de Vijver MJ et al. A gene-expression signature as a predictor of survival in breast cancer. N. Engl. J. Med 347, 1999–2009 (2002). [DOI] [PubMed] [Google Scholar]
  • 58.Rudland PS et al. Prognostic significance of the metastasis-associated protein osteopontin in human breast cancer. Cancer Res. 62, 3417–3427 (2002). [PubMed] [Google Scholar]
  • 59.Mor G et al. Serum protein markers for early detection of ovarian cancer. Proc. Natl Acad. Sci. USA 102, 7677–7682 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Tuck AB, Chambers AF & Allan AL Osteopontin overexpression in breast cancer: knowledge gained and possible implications for clinical management. J. Cell. Biochem 102, 859–868 (2007). [DOI] [PubMed] [Google Scholar]
  • 61.Azmi AS, Bao B & Sarkar FH Exosomes in cancer development, metastasis, and drug resistance: a comprehensive review. Cancer Metastasis Rev. 32, 623–642 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Pucci F & Pittet MJ Molecular pathways: tumor-derived microvesicles and their interactions with immune cells in vivo. Clin. Cancer Res 19, 2598–2604 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Peinado H et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med 18, 883–891 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Grange C et al. Microvesicles released from human renal cancer stem cells stimulate angiogenesis and formation of lung premetastatic niche. Cancer Res. 71, 5346–5356 (2011). [DOI] [PubMed] [Google Scholar]
  • 65.Fremder E et al. Tumor-derived microparticles induce bone marrow-derived cell mobilization and tumor homing: A process regulated by osteopontin. Int. J. Cancer 135, 270–281 (2014). [DOI] [PubMed] [Google Scholar]
  • 66.Zetter BR Angiogenesis and tumor metastasis. Annu. Rev. Med 49, 407–424 (1998). [DOI] [PubMed] [Google Scholar]
  • 67.Chambers AF, Groom AC & MacDonald IC Dissemination and growth of cancer cells in metastatic sites. Nat. Rev. Cancer 2, 563–572 (2002). [DOI] [PubMed] [Google Scholar]
  • 68.Fidler IJ The pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisited. Nat. Rev. Cancer 3, 453–458 (2003). [DOI] [PubMed] [Google Scholar]
  • 69.Gupta GP & Massague J Cancer metastasis: building a framework. Cell 127, 679–695 (2006). [DOI] [PubMed] [Google Scholar]
  • 70.Hiratsuka S et al. MMP9 induction by vascular endothelial growth factor receptor-1 is involved in lung-specific metastasis. Cancer Cell 2, 289–300 (2002). [DOI] [PubMed] [Google Scholar]
  • 71.Hiratsuka S, Watanabe A, Aburatani H & Maru Y Tumour-mediated upregulation of chemoattractants and recruitment of myeloid cells predetermines lung metastasis. Nat. Cell Biol 8, 1369–1375 (2006). [DOI] [PubMed] [Google Scholar]
  • 72.Kaplan RN et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature 438, 820–827 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Dawson MR, Duda DG, Chae SS, Fukumura D & Jain RK VEGFR1 activity modulates myeloid cell infiltration in growing lung metastases but is not required for spontaneous metastasis formation. PLoS ONE 4, e6525 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Peinado H, Lavotshkin S & Lyden D The secreted factors responsible for premetastatic niche formation: old sayings and new thoughts. Semin. Cancer Biol 21, 139–146 (2011). [DOI] [PubMed] [Google Scholar]
  • 75.Erler JT et al. Hypoxia-induced lysyl oxidase is a critical mediator of bone marrow cell recruitment to form the premetastatic niche. Cancer Cell 15, 35–44 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Sceneay J et al. Primary tumor hypoxia recruits CD11b+/Ly6Cmed/Ly6G+ immune suppressor cells and compromises NK cell cytotoxicity in the premetastatic niche. Cancer Res. 72, 3906–3911 (2012). [DOI] [PubMed] [Google Scholar]
  • 77.Kim S et al. Carcinoma-produced factors activate myeloid cells through TLR2 to stimulate metastasis. Nature 457, 102–106 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Sceneay J, Smyth MJ & Moller A The pre-metastatic niche: finding common ground. Cancer Metastasis Rev. 32, 449–464 (2013). [DOI] [PubMed] [Google Scholar]
  • 79.Tsou CL et al. Critical roles for CCR2 and MCP-3 in monocyte mobilization from bone marrow and recruitment to inflammatory sites. J. Clin. Invest 117, 902–909 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Kang SY et al. Prosaposin inhibits tumor metastasis via paracrine and endocrine stimulation of stromal p53 and Tsp-1. Proc. Natl Acad. Sci. USA 106, 12115–12120 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Schelter F et al. Tumor cell-derived Timp-1 is necessary for maintaining metastasis-promoting Met-signaling via inhibition of Adam-10. Clin. Exp. Metastasis 28, 793–802 (2011). [DOI] [PubMed] [Google Scholar]
  • 82.Jung T et al. CD44v6 dependence of premetastatic niche preparation by exosomes. Neoplasia 11, 1093–1105 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Taranova AG et al. Allergic pulmonary inflammation promotes the recruitment of circulating tumor cells to the lung. Cancer Res. 68, 8582–8589 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Esposito M & Kang Y Targeting tumor-stromal interactions in bone metastasis. Pharmacol. Ther 141, 222–233 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Pantel K, Brakenhoff RH & Brandt B Detection, clinical relevance and specific biological properties of disseminating tumour cells. Nat. Rev. Cancer 8, 329–340 (2008). [DOI] [PubMed] [Google Scholar]
  • 86.Klein CA Parallel progression of primary tumours and metastases. Nat. Rev. Cancer 9, 302–312 (2009). [DOI] [PubMed] [Google Scholar]
  • 87.Roy LD et al. Collagen induced arthritis increases secondary metastasis in MMTV-PyV MT mouse model of mammary cancer. BMC Cancer 11, 365 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Naumov GN et al. Persistence of solitary mammary carcinoma cells in a secondary site: a possible contributor to dormancy. Cancer Res. 62, 2162–2168 (2002). [PubMed] [Google Scholar]
  • 89.Almog N Molecular mechanisms underlying tumor dormancy. Cancer Lett. 294, 139–146 (2010). [DOI] [PubMed] [Google Scholar]
  • 90.Mullen CA, Urban JL, Van Waes C, Rowley DA & Schreiber H Multiple cancers. Tumor burden permits the outgrowth of other cancers. J. Exp. Med 162, 1665–1682 (1985). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Reilly RT et al. HER-2/neu is a tumor rejection target in tolerized HER-2/neu transgenic mice. Cancer Res. 60, 3569–3576 (2000). [PubMed] [Google Scholar]
  • 92.Yan HH et al. Gr-1+CD11b+ myeloid cells tip the balance of immune protection to tumor promotion in the premetastatic lung. Cancer Res. 70, 6139–6149 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Granot Z et al. Tumor entrained neutrophils inhibit seeding in the premetastatic lung. Cancer Cell 20, 300–314 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Gao D et al. Myeloid progenitor cells in the premetastatic lung promote metastases by inducing mesenchymal to epithelial transition. Cancer Res. 72, 1384–1394 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Castano Z, Tracy K & McAllister SS The tumor macroenvironment and systemic regulation of breast cancer progression. Int. J. Dev. Biol 55, 889–897 (2011). [DOI] [PubMed] [Google Scholar]
  • 96.Kuznetsov HS et al. Identification of luminal breast cancers that establish a tumor-supportive macroenvironment defined by proangiogenic platelets and bone marrow-derived cells. Cancer Discov. 2, 1150–1165 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Battinelli EM, Markens BA & Italiano JE Jr Release of angiogenesis regulatory proteins from platelet alpha granules: modulation of physiologic and pathologic angiogenesis. Blood 118, 1359–1369 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Nilsson RJ et al. Blood platelets contain tumor-derived RNA biomarkers. Blood 118, 3680–3683 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Gay LJ & Felding-Habermann B Contribution of platelets to tumour metastasis. Nat. Rev. Cancer 11, 123–134 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Italiano JE Jr et al. Angiogenesis is regulated by a novel mechanism: pro-and antiangiogenic proteins are organized into separate platelet alpha granules and differentially released. Blood 111, 1227–1233 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Labelle M & Hynes RO The initial hours of metastasis: the importance of cooperative host-tumor cell interactions during hematogenous dissemination. Cancer Discov. 2, 1091–1099 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Folkman J & Kalluri R Cancer without disease. Nature 427, 787 (2004). [DOI] [PubMed] [Google Scholar]
  • 103.Bateman A & Bennett HP The granulin gene family: from cancer to dementia. Bioessays 31, 1245–1254 (2009). [DOI] [PubMed] [Google Scholar]
  • 104.Castano Z et al. Stromal EGF and IGF1 together modulate plasticity of disseminated triple negative breast tumors. Cancer Discov. 3, 922–935 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Erez N, Truitt M, Olson P & Hanahan D Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-κB-dependent manner. Cancer Cell 17, 135–147 (2010). [DOI] [PubMed] [Google Scholar]
  • 106.Bruzzese F et al. Local and systemic pro-tumorigenic effects of fibroblast-derived GDF15/MIC-1. Cancer Res. 74, 3408–3417 (2014). [DOI] [PubMed] [Google Scholar]
  • 107.Breit SN et al. The TGF-beta superfamily cytokine, MIC-1/GDF15: a pleotrophic cytokine with roles in inflammation, cancer and metabolism. Growth Factors 29, 187–195 (2011). [DOI] [PubMed] [Google Scholar]
  • 108.Li B et al. Id1-induced IGF-II and its autocrine/endocrine promotion of esophageal cancer progression and chemoresistance-implications for IGF-II and IGF-IR-targeted therapy. Clin. Cancer Res 20, 2651–2662 (2014). [DOI] [PubMed] [Google Scholar]
  • 109.Lippitz BE Cytokine patterns in patients with cancer: a systematic review. Lancet Oncol. 14, e218–228 (2013). [DOI] [PubMed] [Google Scholar]
  • 110.Graus F & Dalmau J Paraneoplastic neuropathies. Curr. Opin. Neurol 26, 489–495 (2013). [DOI] [PubMed] [Google Scholar]
  • 111.Taucher S et al. Impact of pretreatment thrombocytosis on survival in primary breast cancer. Thromb. Haemost 89, 1098–1106 (2003). [PubMed] [Google Scholar]
  • 112.Stone RL et al. Paraneoplastic thrombocytosis in ovarian cancer. N. Engl. J. Med 366, 610–618 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Calle EE & Kaaks R Overweight, obesity and cancer: epidemiological evidence and proposed mechanisms. Nat. Rev. Cancer 4, 579–591 (2004). [DOI] [PubMed] [Google Scholar]
  • 114.Baserga R, Peruzzi F & Reiss K The IGF-1 receptor in cancer biology. Int. J. Cancer 107, 873–877 (2003). [DOI] [PubMed] [Google Scholar]
  • 115.Gupta PB et al. Systemic stromal effects of estrogen promote the growth of estrogen receptor-negative cancers. Cancer Res. 67, 2062–2071 (2007). [DOI] [PubMed] [Google Scholar]
  • 116.Demicheli R, Biganzoli E, Boracchi P, Greco M & Retsky MW Recurrence dynamics does not depend on the recurrence site. Breast Cancer Res. 10, R83 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Retsky MW, Demicheli R, Hrushesky WJ, Baum M & Gukas ID Dormancy and surgery-driven escape from dormancy help explain some clinical features of breast cancer. APMIS 116, 730–741 (2008). [DOI] [PubMed] [Google Scholar]
  • 118.Gertz MA Current status of stem cell mobilization. Br. J. Haematol 150, 647–662 (2010). [DOI] [PubMed] [Google Scholar]
  • 119.Ebos JM, Lee CR, Christensen JG, Mutsaers AJ & Kerbel RS Multiple circulating proangiogenic factors induced by sunitinib malate are tumor-independent and correlate with antitumor efficacy. Proc. Natl Acad. Sci. USA 104, 17069–17074 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Okazaki T et al. Granulocyte colony-stimulating factor promotes tumor angiogenesis via increasing circulating endothelial progenitor cells and Gr1+CD11b+ cells in cancer animal models. Int. Immunol 18, 1–9 (2006). [DOI] [PubMed] [Google Scholar]
  • 121.Shaked Y et al. Therapy-induced acute recruitment of circulating endothelial progenitor cells to tumors. Science 313, 1785–1787 (2006). [DOI] [PubMed] [Google Scholar]
  • 122.Shaked Y et al. Rapid chemotherapy-induced acute endothelial progenitor cell mobilization: implications for antiangiogenic drugs as chemosensitizing agents. Cancer Cell 14, 263–273 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Barcellos-Hoff MH Does microenvironment contribute to the etiology of estrogen receptor-negative breast cancer? Clin. Cancer Res 19, 541–548 (2013). [DOI] [PubMed] [Google Scholar]
  • 124.Ebos JM, Lee CR & Kerbel RS Tumor and host-mediated pathways of resistance and disease progression in response to antiangiogenic therapy. Clin. Cancer Res 15, 5020–5025 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Guthrie GJ et al. The systemic inflammation-based neutrophil-lymphocyte ratio: Experience in patients with cancer. Crit. Rev. Oncol. Hematol 88, 218–230 (2013). [DOI] [PubMed] [Google Scholar]
  • 126.Absenger G et al. A derived neutrophil to lymphocyte ratio predicts clinical outcome in stage II and III colon cancer patients. Br. J. Cancer 109, 395–400 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Stotz M et al. Increased neutrophil–lymphocyte ratio is a poor prognostic factor in patients with primary operable and inoperable pancreatic cancer. Br. J. Cancer 109, 416–421 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Ishizuka M, Nagata H, Takagi K, Iwasaki Y & Kubota K Combination of platelet count and neutrophil to lymphocyte ratio is a useful predictor of postoperative survival in patients with colorectal cancer. Br. J. Cancer 109, 401–407 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.McMillan DC The systemic inflammation-based Glasgow Prognostic Score: a decade of experience in patients with cancer. Cancer Treat. Rev 39, 534–540 (2012). [DOI] [PubMed] [Google Scholar]

RESOURCES