Metastasis is initiated and sustained through therapy by cancer cells with stem-like and immune-evasive properties, termed metastasis-initiating cells (MIC). Recent progress suggests that MICs result from the adoption of a normal regenerative progenitor phenotype by malignant cells, a phenotype with intrinsic programs to survive the stresses of the metastatic process, undergo epithelial–mesenchymal transitions, enter slow-cycling states for dormancy, evade immune surveillance, establish supportive interactions with organ-specific niches, and co-opt systemic factors for growth and recurrence after therapy. Mechanistic understanding of the molecular mediators of MIC phenotypes and host tissue ecosystems could yield cancer therapeutics to improve patient outcomes.

Significance:

Understanding the origins, traits, and vulnerabilities of progenitor cancer cells with the capacity to initiate metastasis in distant organs, and the host microenvironments that support the ability of these cells to evade immune surveillance and regenerate the tumor, is critical for developing strategies to improve the prevention and treatment of advanced cancer. Leveraging recent progress in our understanding of the metastatic process, here we review the nature of MICs and their ecosystems and offer a perspective on how this knowledge is informing innovative treatments of metastatic cancers.

Clinically evident metastasis remains largely incurable, but progress is being made in understanding its basic biological principles and laying the foundation for improving long-term outcomes in patients with advanced cancer (1, 2). In spite of the aggressiveness of metastatic cancer, only a small proportion of cells released from a tumor end up forming a distant lesion. This is partly due to the extensive attrition that cancer cells suffer as they confront the stresses of cancer cell dissemination, dormancy, outgrowth, and resistance to therapy (2). Cancer cells must accumulate numerous phenotypic traits to survive these stresses and become proficient at initiating metastasis.

It is increasingly clear that metastasis is initiated by a subset of cancer cells with a stem cell–like phenotype that makes them more competent than the bulk cancer cell population at reinitiating tumor growth in distant sites (3–5). Cell “stemness” in mature tissues represents a phenotypically fluid condition rather than a developmentally imprinted and rigidly defined state. Stem cells in different tissues have distinct properties and developmental trajectories, as do stem cells that support tissue homeostasis versus those that are mobilized in response to injury to regenerate the disrupted tissue. Adult stem cells that maintain homeostasis may suffer oncogenic mutations and become tumor-initiating cancer stem cells (CSC). However, the cells that initiate metastasis are not necessarily CSCs that acquired a few extra metastatic traits. Recent progress indicates that metastatic colonization of distant organs is initiated by stem-like cancer cells that are distinct not only in terms of expressing certain specialized prometastatic traits but also in terms of their overall phenotype, which is more akin to that of regenerative stem cells. Hence, we refer to these cells as “metastasis-initiating cells” (MIC), a term that denotes what these cells can do and why they are of interest. Cells with MIC features are enriched in residual disease that survives cancer therapy, underscoring their importance not only in the original formation of distant metastases but also during relapse of increasingly lethal metastases after multiple treatment cycles in the clinic.

Understanding the origins, traits, and vulnerabilities of MICs and their host ecosystems is critical for developing strategies to improve the prevention and treatment of metastasis. Here we leverage the conceptual framework and open questions from previous reviews on the metastatic process (1, 2) and the cells and microenvironments that make it possible (3, 5) to offer an update on recent progress toward the resolution of these questions. We present a summary of the metastatic process and the traits that cancer cells must possess to become MICs. We then address the question of how MICs acquire these traits. We consider the hypothesis that MICs emerge by the progressive accumulation of an array of prometastatic traits through evolution of highly heterogeneous cancer cell populations under the selective pressures of the multiple steps of the metastatic process. We also consider the alternative hypothesis that cancer cells acquire many of their critical traits at once by adopting a regenerative progenitor phenotype with intrinsic tumor regrowth capacity. Finally, we offer a perspective on the implications of the growing understanding for improving the treatment of patients with metastatic cancer.

Metastasis starts with a phase of cancer cell dissemination from the primary tumor to regional and distal sites (Fig. 1A). In certain cancers, such as breast cancer, dissemination may start during early stages of tumor progression while the tumor is still small and appears to be confined by basement membrane within a ductal structure—i.e., ductal carcinoma in situ (6, 7). Dissemination can continue until the source tumor is removed, as demonstrated by the presence of circulating tumor cells (CTC) in patients undergoing surgical resection of the tumor (8). CTCs culminate their journey by exiting from blood capillaries (a process called extravasation) and infiltrating the new host parenchyma.

Figure 1.

Phases of metastasis. A, Metastasis proceeds through three distinct phases of dissemination, dormancy and colonization. MICs disseminate from the primary tumor and seed multiple organs, where they enter a subclinical state of dormancy. During dormancy, MICs may shuttle between quiescent and proliferative states, with proliferative cells being continually cleared by niche-specific or systemic immune defenses. MICs that acquire immune-evasive and organ-specific growth adaptations are able to exit dormancy and generate clinically evident macrometastatic colonies. During dissemination and dormancy, MICs are in a dynamic equilibrium with host immunity, whereas failure of immune surveillance results in metastatic outbreaks and organ colonization. B, The three phases of metastasis can overlap with the growth of primary tumors and may coexist in the same individual until removal of the primary tumor (stages I–III). The latter two phases continue to coexist during adjuvant therapy, with the eventual dominance of the colonization phase resulting in macrometastatic relapse (in cases that were originally diagnosed as stage I–III), or in patients diagnosed with de novo stage IV cancers.

Figure 1.

Phases of metastasis. A, Metastasis proceeds through three distinct phases of dissemination, dormancy and colonization. MICs disseminate from the primary tumor and seed multiple organs, where they enter a subclinical state of dormancy. During dormancy, MICs may shuttle between quiescent and proliferative states, with proliferative cells being continually cleared by niche-specific or systemic immune defenses. MICs that acquire immune-evasive and organ-specific growth adaptations are able to exit dormancy and generate clinically evident macrometastatic colonies. During dissemination and dormancy, MICs are in a dynamic equilibrium with host immunity, whereas failure of immune surveillance results in metastatic outbreaks and organ colonization. B, The three phases of metastasis can overlap with the growth of primary tumors and may coexist in the same individual until removal of the primary tumor (stages I–III). The latter two phases continue to coexist during adjuvant therapy, with the eventual dominance of the colonization phase resulting in macrometastatic relapse (in cases that were originally diagnosed as stage I–III), or in patients diagnosed with de novo stage IV cancers.

Close modal

The dissemination phase is followed by a phase of metastatic dormancy during which disseminated cancer cells do not yet grow to form a clinically manifest lesion because the host stroma presents physical, metabolic, and immune barriers that prevent this growth (9). Disseminated cancer cells survive under these conditions by entering a state of proliferative quiescence. During dormancy, metastasis remains a cryptic process that may eventually progress toward overt tumor growth or disappear by exhaustion of the disseminated cancer cell population under these barriers or by the effect of therapy.

The final phase of the metastatic process involves overt outgrowth by colonization of the host organ (Fig. 1A). Clinically detectable metastases may not become evident until months or decades after initial diagnosis and removal of a primary tumor. In some cancers overt metastasis occurs in multiple sites without marked preference for one or another organ. In other cancers, metastasis shows strong preference or “tropism” for certain organs. Notably, 2% to 5% of metastatic cancers are diagnosed as carcinomas of unknown primary, in which the primary tumor is not detected either because of its small size or because it may have spontaneously regressed after seeding aggressive metastases (10).

The three phases of the metastatic process—dissemination, dormancy, and outbreak—may coexist in time (Fig. 1B). Cancer cell dissemination may persist as long as the source tumor remains in place, long after the first disseminated cells accumulated as metastatic seeds in distant organs. Precocious metastatic lesions may break out from dormancy while the primary tumor is still in place, as occurs in patients whose cancer is first diagnosed as widely metastatic stage IV disease (Fig. 1B). When distant relapse occurs in a particular site long after the surgical elimination of the primary tumor, other organs may still harbor dormant metastatic cells that can independently evolve and eventually initiate metastatic outgrowth with their own set of organ-specific metastatic traits. An example is provided by late brain recurrence in patients with HER2-positive breast cancer who were treated with anti-HER2 agents that suppressed visceral metastasis (11).

A combination of invasive cell migration, extracellular matrix (ECM) remodeling, and the presence of leaky vasculature at the tumor invasion front enables the dispersion of cancer cells through the circulation, lymphatics, perineural, and perivascular routes, or via direct infiltration of adjacent body cavities (ref. 12; Fig. 2). The hematogenous circulation is the principal route in most types of cancer, but lymphatic dissemination and forms of extravascular spread are prominent routes in various types of cancer as well.

Figure 2.

Metastatic dissemination. The most common and best-characterized route of tumor dissemination is via the blood circulation (A–C). A, During hematogenous dissemination, cancer cells at the invasion front of primary tumors that undergo EMTs lead intravasation into neoangiogenic capillaries in the tumor, and thus access the venous circulation for dissemination to multiple organs. Cancer cells may intravasate individually or in clusters during collective migration. Invasion and intravasation involve remodeling of the tumor extracellular matrix and may be facilitated by tumor-resident fibroblasts and macrophages. B, In the circulation, cancer cells must rapidly adapt to overcome biomechanical, redox, and immunologic threats. Clustering of CTCs may enable paracrine growth factor signaling that promotes the niche-independent survival of disseminated epithelial cells. CTC clusters are enriched in stem-like cancer cells and can include other cell types including platelets and neutrophils, which in turn can protect CTCs from immune attack by secreting immunosuppressive factors. NK cell, natural killer cell. C, CTCs become trapped in the capillary beds of multiple organs and migrate into the organ parenchyma to seed nascent metastasis. Fenestrated capillary beds in the liver and bone marrow can facilitate extravasation. Nontumor cells with CTC clusters, or in the parenchyma, including platelets and monocytes, can facilitate endothelial permeability and transmigration by secreting endothelial disjunction factors. Alternatively, CTCs can secrete factors that induce endothelial necroptosis. D, Beyond hematogenous dissemination, cancer cells originating in certain primary tumors can also reach distant organs via alternative routes.

Figure 2.

Metastatic dissemination. The most common and best-characterized route of tumor dissemination is via the blood circulation (A–C). A, During hematogenous dissemination, cancer cells at the invasion front of primary tumors that undergo EMTs lead intravasation into neoangiogenic capillaries in the tumor, and thus access the venous circulation for dissemination to multiple organs. Cancer cells may intravasate individually or in clusters during collective migration. Invasion and intravasation involve remodeling of the tumor extracellular matrix and may be facilitated by tumor-resident fibroblasts and macrophages. B, In the circulation, cancer cells must rapidly adapt to overcome biomechanical, redox, and immunologic threats. Clustering of CTCs may enable paracrine growth factor signaling that promotes the niche-independent survival of disseminated epithelial cells. CTC clusters are enriched in stem-like cancer cells and can include other cell types including platelets and neutrophils, which in turn can protect CTCs from immune attack by secreting immunosuppressive factors. NK cell, natural killer cell. C, CTCs become trapped in the capillary beds of multiple organs and migrate into the organ parenchyma to seed nascent metastasis. Fenestrated capillary beds in the liver and bone marrow can facilitate extravasation. Nontumor cells with CTC clusters, or in the parenchyma, including platelets and monocytes, can facilitate endothelial permeability and transmigration by secreting endothelial disjunction factors. Alternatively, CTCs can secrete factors that induce endothelial necroptosis. D, Beyond hematogenous dissemination, cancer cells originating in certain primary tumors can also reach distant organs via alternative routes.

Close modal

Invasion and Intravasation

Malignant cells remodel the basement membrane to invade into tissue parenchyma, induce neoangiogenesis, move directionally toward blood vessels, and undergo transendothelial migration to enter the portal or systemic venous circulations (refs. 13, 14; Fig. 2A). These steps are enabled by dynamic changes in the repertoire and function of cell–cell adhesion molecules (cadherins and CAM family cell adhesion molecules), ECM remodeling enzymes (matrix metalloproteinases and ADAMTS endopeptidases), and cell–matrix adhesion molecules (integrins and syndecans; refs. 15, 16). Carcinoma cells undergo epithelial-to-mesenchymal transition (EMT), a phenotypic conversion process that enables cell migration and entry into the circulation (see below; refs. 1, 17). Stromal cells, including macrophages, fibroblasts, and neutrophils, can further stimulate invasion and intravasation by direct interactions with carcinoma cells (18, 19). Cancer cell–derived fibrogenic signals stimulate the deposition of desmoplastic matrix by mesenchymal cells in the tumor stroma (20, 21), which in turn facilitates the invasion of cancer cells and their proliferation through mechanosignaling cues (15, 16). The physical stress of migrating through tight interstitial spaces leads to nuclear envelope rupture and chromatin shearing, thus causing further genomic and phenotypic diversification of cancer cells that survive such stress (22, 23).

CTCs

Unlike tumor tissues, which can be accessed only via invasive biopsies, CTCs derived from primary or metastatic tumors can be readily and repeatedly sampled via simple blood draws from patients with cancer. This provides a unique window to study metastasis biology and for use as longitudinal clinical biomarkers of metastasis and therapy response (24, 25). For these reasons, CTCs have become a topic of intense study. Although the sensitivity and specificity of current CTC detection assays remain limiting for some applications, the prospect of delineating CTC characteristics that discriminate between high and low metastatic capacity is of biological and clinical significance.

Cancer cells circulate singly and in clusters (ref. 26; Fig. 2B). CTC clusters are observed in the blood of patients and tumor-bearing mice and have a superior ability to seed metastasis in experimental models (27). CTC clusters may also include tumor-derived stromal cells (28). CTC clustering provides intercellular spaces that concentrate paracrine growth-promoting signals (29) and enable cooperation between different cancer cell states within a cluster (30). CTC clusters are enriched in genome methylation patterns that denote a stem-like cancer cell state (31), and therefore have features of MICs. MICs upregulate expression of certain cell–cell and cell–matrix adhesion genes (32), raising the question: Are CTC clusters more metastatic because clustering augments the aggressiveness of cancer cells or because MICs are intrinsically prone to clustering?

Blood platelets coat CTCs, providing protection from shear stress and immune attack, particularly attack by natural killer (NK) cells. Platelets can also facilitate cancer cell extravasation by mediating adhesion to selectins on the endothelium and by enforcing EMT in the cancer cells. Platelets release factors including TGFβ and platelet-derived growth factor (PDGF) that suppress NK cells and promote EMT of CTCs (14, 33, 34). A small proportion of CTCs in patients with breast cancer are bound to neutrophils, an abundant white blood cell type; these CTCs are more proliferative and effective at forming metastases in mice than are neutrophil-free counterparts (35).

Extravasation

Within seconds to minutes of leaving a primary tumor, CTCs may already become trapped in the capillaries of distant organs. In experimental systems, CTCs can remain lodged in capillaries for days until they either extravasate or disappear (36). The half-life of cancer cells in the circulation is estimated to be just a few hours, based on the rate of decline in the CTC numbers following surgical removal of primary tumors in patients (8).

Differences in the tightness of capillary endothelial walls determine the difficulty of seeding metastasis in different organs, contributing to organ-specific patterns of metastatic spread (Fig. 2C). Liver and bone, the most common sites of metastasis from many cancers, contain sinusoid capillary beds with large intracellular fenestration gaps as well as gaps in the underlying basement membrane, which facilitate CTC extravasation. At the other end of the range, the blood–brain and blood–choroid plexus barriers restrict the movement of molecules and cells between the systemic circulation and the brain parenchyma and cerebrospinal fluid, respectively. Accordingly, metastasis to the brain and leptomeningeal spaces selects for MICs expressing factors that increase barrier permeability or disrupt the barrier altogether (37–39).

Extravasation can be facilitated by platelets coating CTCs (34) and cytokines secreted by perivascular macrophages in target organs or by the cancer cells themselves (ref. 13; Fig. 2C). As an alternative to squeezing through tight interstitial spaces between endothelial cells, cancer cells may induce endothelial cell death. Human and mouse cancer cells have been shown to induce necroptosis of endothelial cells in coculture and orthotopic lung metastasis models, whereas inhibition of RIPK1/3-dependent endothelial necroptosis reduced extravasation and metastasis in these models (40).

Following extravasation, metastatic cells remain situated near blood capillaries and migrate along the abluminal surface on the capillaries (36, 41–44). Integrins mediate interactions between cancer cells and capillaries (45). This is most apparent in experimental models of metastasis to the brain, where the elongated nature of capillaries facilitates imaging studies (41, 43). However, perivascular residence of extravasated cancer cells is also observed in other sites, including the liver, lungs, and bone marrow (43).

Lymphatic Dissemination

Cancer cells disseminate via the lymphatic circulation, but whether lymph nodes are an obligatory staging post for metastasis remains controversial and likely depends on tumor type (46, 47). Comparative genomics of regional lymph node and distant metastases from colorectal and breast cancers suggest that in the majority of cases, distinct clones within primary tumors give rise to nodal and distant metastases, with the latter undergoing more stringent clonal selection (48–50). In patients, highly aggressive triple-negative breast cancers often metastasize without apparent regional lymph node involvement (47). Consistent with a correlative, not causal, relationship between lymph node and distant metastasis, recent clinical trials have shown no survival benefit for regional lymph node dissection in patients with breast and ovarian cancers and melanoma (51–53). Even when injected directly into the lymph nodes of mice, cancer cells preferentially metastasize via the blood rather than through the lymphatics (54, 55). However, transient passage through the lymphatics may confer long-term survival benefits to MICs as they join the blood circulation (56).

Melanomas implanted in mice lacking dermal lymphatic vessels exhibited lower cytokine expression and leukocyte infiltration than those implanted in wild-type controls, suggesting that cancer cell interactions with immune cells in lymphatic structures are important for antitumor immunity (57). The distinctive metabolic milieu of lymph may also enable cancer cell adaptation to the stresses of metastasis. For example, exposure to oleic acid in lymph protectedmelanoma cells from ferroptosis and increased their ability to initiate metastasis during subsequent hematogenous dissemination (58).

Extravascular Spread

The metastatic spread of some tumors occurs largely or at least partly through routes that obviate the need to enter and exit the circulation (Fig. 2D). In ovarian cancer, cell clusters carried by the peritoneal fluid attach on the abdominal peritoneum where the cells grow (59). Peritoneal metastasis is also frequent in colorectal and pancreatic cancers. Melanomas and other cancers can spread without entering and exiting the circulation by engaging in extravascular migratory metastasis, a process of migration over the abluminal surface of vessels that is common during embryogenesis (60). Perineural spread is a long recognized, if underappreciated, form of metastatic spread in head and neck cancers and other squamous cell carcinomas (61). Regional spread of lung adenocarcinoma can occur by spread through air spaces where cancer cells detach from a primary tumor, migrate as clusters through air spaces, and reattach to the alveolar walls through vessel co-option to resume tumor growth, which is associated with tumor recurrence and poor survival of stage I lung adenocarcinomas (62, 63).

Dormant metastasis and the ability to predict the likelihood of an eventual outbreak in patients diagnosed with early-stage cancer are major considerations in the clinic (64). For example, patients with estrogen receptor–positive early-stage breast cancer who undergo complete surgical removal of the primary tumor continue to experience metastatic relapse as late as 20 years from their original diagnosis (64). Dormant metastasis is an understudied problem, owing to a dearth of appropriate experimental models. Models using aggressive metastatic cells sidestep the dormancy phase altogether. Some models of dormant metastasis rely on comparisons between nonisogenic cancer cell lines of varying propensity to develop metastatic colonies after inoculation in mice. Spontaneously arising models of dormancy have been characterized (9). New experimental models have been developed by in vivo selection of early-stage tumor-derived MICs that are competent to establish dormant metastasis and spontaneously generate late outbreaks, thus mimicking the course of the disease in patients (65). Studies on these various models have begun to shed light on the biology of MIC entry and exit from dormancy (Fig. 3).

Figure 3.

Metastatic dormancy and outbreak. A, MICs seeding distant organs enter into a variable period of dormancy, when then cannot be detected by clinical imaging technologies. Clinical dormancy reflects an equilibrium incorporating cell-intrinsic growth arrest, and stochastic MIC proliferation events, which are countered by elimination of proliferating cells. Secreted factors in the perivascular microenvironment may either inhibit MIC proliferation (e.g., TGFβ) or promote it (e.g., WNT), which in turn can be countered by MIC secretion of WNT inhibitors (e.g., DKK1). MICs spread on the abluminal surface of capillaries by adhering to the perivascular basement membrane, which provides proliferative inputs. However, stromal factors can induce proliferating MICs to differentiate and lose stemness properties (e.g., BMP). In contrast to quiescent MICs, proliferating MICs upregulate cell-surface expression of NK ligands and MHC class I molecules, facilitating their detection and destruction by NK cells and adaptive immune cells. B, Exit from dormancy requires close interactions between MICs and ECM. MICs induce innate immune responses that include the recruitment of neutrophils and the formation of extracellular web-like chromatin decondensates harboring ECM remodeling enzymes, termed NETs. NET-dependent cleavage of laminin, a key component of the ECM, activates β1-integrin signaling in MICs. MICs can also activate signaling through integrin-like kinase (ILK) by upregulating L1CAM, a cell-adhesion molecule that binds laminin on extracellular basement membranes, including that of blood capillaries. Integrin/ILK signaling enables cell stretching via the formation of actin-dependent protrusions, which in turn enables mechanosignaling to activate YAP-dependent transcriptional output and proliferation. Homophilic L1CAM interactions as cancer cells multiply can extend this signaling. Additional cues from the ECM, including the stem cell niche ECM components tenascin C and periostin, can potentiate growth factor signaling and enable MIC stemness and proliferation. C, Outgrowth of dormant metastasis might be prevented by bolstering barriers to MIC survival and proliferation or targeting the phenotypic adaptations required for escape from dormancy.

Figure 3.

Metastatic dormancy and outbreak. A, MICs seeding distant organs enter into a variable period of dormancy, when then cannot be detected by clinical imaging technologies. Clinical dormancy reflects an equilibrium incorporating cell-intrinsic growth arrest, and stochastic MIC proliferation events, which are countered by elimination of proliferating cells. Secreted factors in the perivascular microenvironment may either inhibit MIC proliferation (e.g., TGFβ) or promote it (e.g., WNT), which in turn can be countered by MIC secretion of WNT inhibitors (e.g., DKK1). MICs spread on the abluminal surface of capillaries by adhering to the perivascular basement membrane, which provides proliferative inputs. However, stromal factors can induce proliferating MICs to differentiate and lose stemness properties (e.g., BMP). In contrast to quiescent MICs, proliferating MICs upregulate cell-surface expression of NK ligands and MHC class I molecules, facilitating their detection and destruction by NK cells and adaptive immune cells. B, Exit from dormancy requires close interactions between MICs and ECM. MICs induce innate immune responses that include the recruitment of neutrophils and the formation of extracellular web-like chromatin decondensates harboring ECM remodeling enzymes, termed NETs. NET-dependent cleavage of laminin, a key component of the ECM, activates β1-integrin signaling in MICs. MICs can also activate signaling through integrin-like kinase (ILK) by upregulating L1CAM, a cell-adhesion molecule that binds laminin on extracellular basement membranes, including that of blood capillaries. Integrin/ILK signaling enables cell stretching via the formation of actin-dependent protrusions, which in turn enables mechanosignaling to activate YAP-dependent transcriptional output and proliferation. Homophilic L1CAM interactions as cancer cells multiply can extend this signaling. Additional cues from the ECM, including the stem cell niche ECM components tenascin C and periostin, can potentiate growth factor signaling and enable MIC stemness and proliferation. C, Outgrowth of dormant metastasis might be prevented by bolstering barriers to MIC survival and proliferation or targeting the phenotypic adaptations required for escape from dormancy.

Close modal

Causes of Cancer Cell Elimination

Clinical dormancy implies one of two complementary scenarios: either a cell-autonomous “temporary mitotic exit” as originally advanced by Geoffrey Hadfield in 1954 (66) or, alternatively, the existence of persistent attempts at colonization that are aborted by cell-extrinsic limitations on metastatic outgrowth. Although several studies have explored the niches, signals, and mechanisms that maintain both modes of dormancy (reviewed in refs. 9, 67, and 68), multiple lines of experimental and clinical data are converging on the view that dormancy reflects a dynamic equilibrium of MICs with antagonistic immune surveillance (Fig. 3A).

Cancer cells suffer extensive attrition after infiltrating distant organs, as shown in mouse models of metastasis and as inferred from the low counts of disseminated cancer cells relative to the number of CTCs that a primary tumor likely shed while it was present in a patient. Elimination of cancer cells after extravasation is driven by passive factors such as high oxidative stress in the lungs (69, 70), a dearth of growth factors and nutrients in the cerebrospinal fluid (38, 71), or, more generally, a lack of the appropriate microenvironment cues that had nursed the growth of MICs in the primary tumor. These passive mechanisms of disseminated cancer cell elimination are accompanied by the proactive role of tissue-resident immunity and other defenses, such as astrocytes in the brain (36), against intruding cancer cells. Tissue-resident macrophages, such as microglia in the brain and Kupffer cells in the liver, and NK cells attack infiltrated cancer cells (18, 72, 73). In experimental models, depletion of T cells and NK cells increases the survival and outgrowth of disseminated cancer cells (66, 74, 75). Tumor lymphocyte infiltration, indexed in the form of an “immunoscore,” is inversely correlated with metastatic burden in patients (76), and metastatic clones can show branched evolution as a result of different immunoediting pressures (77). Dramatically, immunosuppressed recipients of transplant organs from donors deemed cured of localized melanoma or glioblastoma developed donor-derived metastasis (78, 79). Underscoring the importance of immune surveillance of MICs, in patients with completely resected localized melanomas, postsurgical adjuvant therapy with immune-checkpoint inhibitors to boost antitumor immunity reduced later metastatic relapse and prolonged survival (80–83).

Entering Dormancy

Disseminated MICs that enter proliferative quiescence may be particularly equipped to survive these threats. Quiescence allows cancer cells to evade the innate and adaptive arms of immunity mediated by NK and T cells, respectively (65, 84–87). As an added benefit, dormancy renders disseminated cancer cells resistant to antimitotic therapy. The ability to enter a slow-cycling state is a property of some adult stem cells. Thus, to the extent that MICs are primed to enter protective dormancy, dormancy will enrich for MICs in disseminated cancer cell populations.

Entry of MICs into quiescence is stimulated by growth-inhibitory signals in the host microenvironment (Fig. 3A). TGFβ has cytostatic effects on normal epithelial cells and carcinoma cells by inducing the expression of cyclin-dependent kinase inhibitors, whereas it triggers tumor-suppressive apoptosis in premalignant epithelial cells. Carcinoma cells bypass the proapoptotic effect of TGFβ through various mechanisms while retaining the ability to respond to TGFβ with prometastatic effects, including entry into immunoprotective quiescence (88, 89). TGFβ has been shown to stimulate dormancy in experimental models of prostate, breast, and head and neck carcinomas (9). The three isoforms, TGFβ1, TGFβ2, and TGFβ3, are known to be produced in tumor microenvironments by various stromal components and cancer cells. Latent TGFβ1 in particular is present in perivascular niches of the bone marrow and brain parenchyma of mice and is activated by thrombospondin (TSP) to drive extravasated breast cancer cells into proliferative quiescence (ref. 44; Fig. 3A). In this context, TGFβ likely functions through canonical binding to the paired receptor kinases TGFBR1 and TGFBR2 with betaglycan, also known as type III TGFβ receptor, serving as a coreceptor that is particularly important for TGFβ2 binding (88).

Entry of disseminated MICs into dormancy is not merely a passive process resulting from the effect of local growth-inhibitory signals from the tumor microenvironment. Entry into dormancy can also be initiated by MICs themselves such that MICs that actively avoid mitogenic stimulation are selected for because quiescence enables immune evasion (65). MICs derived from early-stage lung and breast tumors actively secrete DKK1, an inhibitor of WNT factors that are potent mitogens for adult stem cells (Fig. 3A). This autocrine DKK1 enforces MIC dormancy in these models, and depleting DKK1 releases MICs from dormancy and delivers them to NK-mediated clearance (65).

These insights suggest that dormant metastasis results from a dynamic equilibrium in which MICs enter proliferative quiescence and thus avoid immune-mediated killing or enter the cell cycle for tumor growth at the risk of being eliminated (Fig. 3A). Another form of dormancy is tumor mass dormancy in which MICs start proliferating but are unable to satisfy the nutrient or oxygen requirements for growth beyond a small tumor mass due to lack of angiogenesis or other causes (9, 90). However, large clinical trials have shown little success with antiangiogenic drugs in the adjuvant or neoadjuvant setting when targeting latent micrometatases, suggesting that (with the limitations of the drugs available) the angiogenic switch may not be a major limiting factor to metastatic outgrowth in patients, or that current therapies are inadequate to suppress angiogenesis (91).

Exiting from Dormancy

Little is known about the signals that lead disseminated cancer cells to exit from dormancy and develop clinically detectable metastases. It is possible that disseminated cancer cells are “awakened” from years of quiescence by signals that jolt them into entering the cell cycle. However, the cited studies on depleting mice of immunity suggest that there is always a fraction of the disseminated cancer cell population attempting to proliferate, perhaps due to a growth-permitting balance between local mitogenic and antimitogenic signals (Fig. 3A). In line with this idea, depletion of DKK1 in models of dormant lung adenocarcinoma and breast cancer metastasis enables exit from dormancy, implying that WNT ligands in the perivascular niche reactivate the proliferation of dormant cells (65). Bone morphogenetic proteins can trigger differentiation of disseminated breast cancer cells in mouse models, which is prevented by BMP sequestering factors (92). Cell metabolism may also play a role. Cell autophagy in response to metabolic stress promotes the survival of dormant breast cancer cells in mouse models (93), and depleting autophagy related 3 (ATG3) or p62/sequestosome-1 promotes cancer cell exit from dormancy with expression of 6-phosphofructo-2-kinase, which supports glycolysis (94).

MICs have certain requirements for continued growth during exit from dormancy. MICs derived from breast, lung, colorectal, and renal cell carcinomas express L1CAM and use this cell adhesion molecule to bind to laminin in perivascular basement membranes, spreading on the abluminal surface of capillaries (Fig. 3B). Once engaged, L1CAM cooperates with β1 integrins, the integrin–actin bridging protein ILK, and p21-activating kinases (PAK) to activate transcription factors YAP and MRTF for the initiation of metastatic outgrowth (32, 43). ILK and β-parvin regulate actin filaments supporting the formation of filopodia-like protrusions, which are required for metastatic outgrowth (95). The L1CAM-expressing MICs grow forming sheaths around the vessels, engulfing the capillary network before the micrometastatic mass acquires a spheroidal shape (32, 43). The requirement for L1CAM has been shown in experimental models of dormant MIC outgrowth following depletion of NK cells as well as models of aggressive metastatic outgrowth without an intervening dormancy phase (43).

Recent studies have revealed a role for neutrophil extracellular traps (NET) in promoting YAP activation during cancer cell exit from dormancy (96, 97). NETs are protease-loaded chromatin webs that neutrophils release as an ultimate maneuver to entrap and clear pathogens (98). NETs are of interest for their roles in inflammation, autoimmune diseases, and cancer (99). Studies using intravital microscopy documented the presence of NETs around disseminated breast cancer cells in mouse models of metastasis and an inhibitory effect of DNAse I on metastatic growth, implying that cancer cell–triggered NETs promote metastasis (100). Indeed, NETs produced during inflammatory responses were shown to reactivate the growth of local dormant cancer cells through cleavage of basement membrane laminin by NET-associated elastase and matrix metalloproteinase 9 (MMP9; Fig. 3B). The cleaved laminin binds α3β1 integrin to activate YAP and proliferation of the cancer cells (97).

Colonization of distant organs is the most dramatic phase of the metastatic process clinically and the most complex biologically. The outgrowth of disseminated MICs into large metastatic colonies depends on the successful avoidance of organ-specific barriers and the productive interactions with specific components of the host tissue ecosystem. Many of these barriers and interactions vary with the site of metastasis. The lung, liver, or brain parenchyma, the lymph nodes, bone marrow, and leptomeninges present widely different tissue microenvironments. Not surprisingly, studies on metastatic colonization of these major target sites have uncovered different factors, metabolic changes, cell–cell interactions, and other determinants of metastatic colonization depending on the tumor type and the metastasis organ site. Detailed accounts of organ-specific metastatic colonization factors have recently been provided for metastasis to different organs (101, 102), and specifically to bone (103), central nervous system (39, 104), lung (105), and liver (106). Here we briefly summarize current concepts on general and organ-specific colonization processes.

Determinants of Organ Tropism

Metastatic tropism is manifest in the tendency of tumors to preferentially relapse in certain organs. Some cancers (e.g., small cell lung carcinomas and metastatic melanomas) form metastasis in multiple sites without marked organ preference, whereas other cancers preferentially relapse in certain organs (e.g., the bones in prostate and hormone receptor–positive breast carcinomas, the liver in colorectal carcinomas and ocular melanomas, or the lungs in soft-tissue sarcomas).

Metastatic tropism results from the combination of two distinct phenomena. One is the ability of cancer cells to reach particular organs through the circulation. Colon cancer is thought to predominantly metastasize to the liver because the mesenteric blood draining the intestines flows via the hepatic portal vein into the liver. The majority of CTCs derived from an intestinal tumor become trapped in the hepatic sinusoids and initiate metastases in the liver with far higher frequency than in the lungs, brain, or bones. It has been proposed that cells derived from certain tumors use organ-specific endothelium adhesion molecules to concentrate in the capillaries of those organs and preferentially extravasate there (107). However, experimental (65, 108, 109) and clinical (110) evidence suggests that, in most types of cancers, disseminated cells reach all organs and leave dormant cells everywhere. This suggests that metastasis develops from latent surviving MICs in the least antagonistic site first, and in other sites later. Thus, the metastatic organ tropism of a particular type of cancer is a function of the organs that MICs can travel to and, most critically, the probability that MICs will grow in a particular organ site depending on their intrinsic or acquired ability to sort the barriers and utilize the supportive niches in that site.

Surpassing Endothelial Barriers

As we noted earlier, differences in the permeability of capillary beds in different organs determine the ability of disseminated cancer cells to accumulate in these organs. Several mediators of cancer cell extravasation have been identified in experimental model systems, including cytokines (e.g., ANGPTL4 and HBEGF), secreted enzymes (e.g., cathepsin S), and monocytes that facilitate extravasation in the lung capillary walls (1, 2, 13) or through the blood–brain barrier (refs. 37–39, 111; Fig. 2C). Metastatic colon cancer cells that colonize the liver can gain lung metastatic ability by expressing the cytokine PTHrP, which mediates extravasation of these cells in the lungs after they reenter the circulation (112). In some instances, metastatic cells disrupt endothelial barriers to allow the influx of nutrients from the circulation. Breast and lung carcinoma cells that infiltrate the leptomeninges produce complement component 3 to activate the C3a receptor in the choroid plexus epithelium and disrupt the blood–CSF barrier, thereby allowing influx of growth factors and nutrients from blood into the nutritionally barren CSF (38).

Neutralizing Hostilities

The important role of immunity in suppressing the outgrowth of disseminated MICs can give rise to organ-specific differences depending on the ability of MICs from different types of cancers to evade tissue-resident immunity (18, 72, 73). The ability of disseminated cancer cells to regulate their own exposure to immune attack by modifying the expression of MHC class I molecules (113), NK-cell and effector T-cell ligands (114), cytosolic DNA-sensing pathways (115), and damage-associated RNAs as ligands for pattern-recognition receptors (116) is under study given their likely importance in metastatic colonization. Moreover, the defenses that invading cancer cells must neutralize to seed distant organs are not limited to classic immunity. Breast cancer and lung adenocarcinoma cells that infiltrate the brain must contend with the release of proapoptotic FAS ligand by reactive astrocytes. FAS ligand is released from its membrane-bound precursor in astrocytes by the protease plasmin, and cancer cells can prevent this by expressing serpin inhibitors of the plasminogen activator (36).

Metabolic Adaptations

Metabolic adaptation is an important aspect of metastasis. Unlike glycolytic primary tumors, MICs in multiple cancer types undergo a bioenergetic shift into an oxidative phosphorylation state (69, 117, 118). Organ-specific metabolic adaptations enable carcinoma MICs to reinitiate growth in tissues with variable bioavailability of oxygen, including in the lungs, where high oxygen levels cause oxidative stress that cancer cells can withstand by overexpressing NRF2 (refs. 118–120; Fig. 3C). Metastatic melanoma cells also undergo metabolic adaptations to withstand high levels of oxidative stress in the blood and lungs (69). Adaptations to the bioavailability of amino acids (121, 122), nucleotides (123), and other bioenergetic substrates (124, 125) have also been noted in metastasis models and human tissue samples and are likely to vary depending on the host tissue. In turn, metabolic stresses may shape the genomic and epigenomic reprogramming of cancer cells to enable metastasis (126). Other cancer cell adaptations serve to source scarce nutrients. For example, metastatic cells secrete the iron-chelating factor lipocalin A to scavenge scarce iron in the CSF (71).

Lipids are important for tumor growth. CD36, a scavenger receptor that can transport fatty acids and other molecules, is upregulated in MICs from oral carcinomas and other cancers and required for metastasis in various organs (127). Brain metastasis appears to place higher demands on MICs with regard to procuring their own lipids. The brain has a markedly lower level of triacylglycerol stores than other tissues. Breast cancer cell lines that are metastatic to the brain upregulate sterol response element binding transcription factor 1 (SREBF1) to promote lipid synthesis and fatty acid metabolism or alternatively upregulate CD36 for metastasis growth (128).

Metastatic Niches and Stromal Co-option

MICs that survive the organ-specific stresses described above additionally require support from the host stroma. Stromal support is thought to be important for MICs immediately after their infiltration of new tissues and establishment as long-term seeds for metastases. This may depend on a combination of cellular and extracellular cues that fulfills the survival and self-renewal requirements of MICs by mimicking the native niches that these cells or their predecessors occupied in their tissue of origin. Various cellular and extracellular components of natural stem cell niches support the viability of disseminated MICs (3, 5), including the ECM interacting components periostin and tenascin C, which cancer cells and fibroblasts produce to promote WNT and Notch signaling in MICs (refs. 129, 130; Fig. 3B). Evidence that disseminated cancer cells intrude into natural stem cell niches in the bone marrow has been provided in models of prostate (131) and breast cancer metastasis (132).

In addition to biochemical signals from the niche, biophysical and mechanical features of stromal niches can also promote metastasis (133). An expanding primary tumor mass exerts mechanical force on its tissue microenvironment, in turn triggering differentiation of myofibroblasts to produce extracellular proteins such as collagens and tenascin C (134), which further increase stiffness of the ECM. In turn, increased ECM stiffness can promote integrin-mediated focal adhesion assembly, RHOA–ROCK-mediated cell contractility, and mechanosignaling via the TGFβ, WNT, and YAP pathways (reviewed in ref. 133). Stiffness-mediated mechanosignaling may thus support MIC activities, including motility, anoikis evasion, and EMT. Consistently, in patients with breast and pancreatic cancers, tumors with increased ECM stiffness were associated with increased metastasis, therapy resistance, and poor prognosis (135–137).

Support from the stroma is also essential during the expansion of macrometastatic colonies, maintenance or aggressive metastatic lesions, and resistance to therapy. Most parenchymal components in sites of metastasis have been implicated in the support of survival, outgrowth, and/or chemoresistance of MICs through unidirectional or reciprocal signaling interactions with the cancer cells. This includes osteoblasts and osteoclasts in bone metastasis (101, 103, 138–140); astrocytes (141, 142), microglia (143), and neurons (144) in brain metastasis (39, 104); monocytes (145, 146), neutrophils (97, 129, 147), fibroblasts (130, 148), and epithelial cells (149) in lung metastasis (105); and mesenchymal, epithelial, macrophage, and other cells in liver metastasis (106). Cytokines and enzymes released by a primary tumor in soluble or vesicular form can alter the parenchyma of distant organs and establish “premetastatic niches,” zones that favor the immediate growth of cancer cells reaching these sites (150). It is unclear if premetastatic niches established by a primary tumor could support MICs as dormant seeds for long periods after a primary tumor is surgically removed.

Most studies on stromal interactions with metastatic cells have utilized experimental models in which aggressive cancer cells proceed to form metastatic colonies without an intervening dormancy phase. Therefore, with few exceptions, these studies do not directly distinguish whether a supportive interaction benefits MICs prior to dormancy, during the incipient outgrowth of micrometastasis after dormancy, or during the continued expansion of macrometastases. In spite of this shortcoming, several organ-specific metastatic mediators identified to date are accompanied with evidence of clinical relevance, providing potential targets for therapy in the treatment of metastasis.

EMT, referred to in passing above, is a key process during tumor progression and metastasis (1). Epithelial cells undergoing an EMT lose polarity and downregulate key cell-adhesion molecules, resulting in the loss of contacts with neighboring cells and an increased ability to migrate and invade adjacent tissue. The ability to undergo an EMT is a property of epithelial stem and progenitor cells, both normal and neoplastic, and reflects an intrinsic phenotypic plasticity of these cell states. EMTs play important roles in development and epithelial regeneration. During gastrulation, epiblast cells undergo an EMT to migrate to appropriate locations as they differentiate into mesoderm and endoderm progenitors. EMTs are also important in later morphogenic events and during wound healing in the adult (17, 21, 151). Cells that undergo an EMT return to an epithelial state through a mesenchymal-to-epithelial transition (MET; refs. 17, 21, 151). The regained epithelial state after an EMT–MET sequence may be different from the starting one, as it occurs during gastrulation.

In normal and neoplastic cells, EMTs are driven by SNAIL, SLUG, ZEB, TWIST, and other transcription factors (referred to as EMT-TF) that repress epithelial genes and stimulate the expression of mesenchymal components, together with micro-RNAs that balance this regulatory network (21). Stromal signals that trigger EMTs include TGFβ, WNT, and NFκB activators (17, 21, 151). TGFβ induces the expression of EMT-TFs in cooperation with RAS–MAPK signaling; TGFβ-activated SMAD transcription factors and MAPK-activated RREB1 jointly drive expression of SNAIL and SLUG coupled to cell type–dependent differentiation or fibrogenesis gene sets, in both normal and neoplastic cells (152). WNT-activated TCF transcription factors and NFκB transcription factor also converge on EMT-TF gene enhancers to regulate EMTs (21).

Carcinoma cells at the tumor invasion front undergoing an EMT can drive collective migrating by pulling cohesive ensembles of cancer cells that remain connected by cell–cell contacts (153). After efficiently disseminating and infiltrating distant organs using the mesenchymal properties acquired with the EMT, disseminated MICs undergo an MET to reinitiate tumor growth (154, 155). In human and mouse carcinomas, loss of cell-surface E-cadherin during EMT activates prosurvival signaling that is necessary in the detached state, but E-cadherin reexpression is required for the outgrowth of metastatic colonies (34). What signals trigger MET in disseminated MICs, and whether MET occurs before or after MICs pass through dormancy, remain open questions.

Epithelial and mesenchymal states are now viewed not as discrete endpoints but as a continuum of highly fluid, interconverting phenotypic states. Each particular epithelial progenitor cell type undergoes its own form of EMT, involving a distinct combination of EMT-TFs and different changes in cell morphology, adhesion, and motility (17). The extent of mesenchymal conversion achieved by an epithelial progenitor during an EMT also depends on the developmental state of this cell and reflects the phenotypic space that this state is programmed to sample. Many epithelial progenitors undergo what is referred to as a “partial” or “hybrid” EMT. However, the current trend is to consider every departure of epithelial cells from the polarized phenotype an EMT regardless of how far toward a stereotypical (“full”) mesenchymal phenotype this departure may reach (17). Although EMT is a common mechanism for metastatic dissemination, the cells that are most competent at forming metastasis are not necessarily those that adopt the most mesenchymal-like phenotype in their EMT (152, 156, 157). Some forms of dissemination, such as collective migration, may enable cancer cells to retain epithelial identity and intercellular contacts during migration and may not require an EMT.

Cancer cells require a large number of traits to perform each step of the metastatic process and additionally survive therapy if present. Some of the traits required to become a MIC are imparted by the basic tumor-initiating phenotype resulting from genetic alterations that activate driver oncogenes and that disrupt tumor suppressor genes. In addition to uncontrolled survival and proliferation functions, these traits include the ability to migrate, invade, and remodel ECM, all of which are conducive to metastasis. The basic traits or “hallmarks” of cancer included with the neoplastic phenotype remain essential throughout the course of tumor progression (158). Moreover, growing evidence indicates that cancer is initiated by oncogenic mutations in stem cells whose normal function is to maintain tissue homeostasis. Turned into CSCs, these cells retain self-renewal capacity, or “stemness,” which is essential for long-term tumor growth (4).

But even with these powerful traits in place, a vast majority of cancer cells leaving a tumor fail to form distant metastasis, instead succumbing to the stresses of the metastatic process. The many obstacles that MICs must sort on their way to establishing metastasis imply a need for additional traits beyond those provided by the CSC phenotype alone. Indeed, work over the past two decades has identified a large number of “metastasis genes” whose expression (or repression) enhances the ability of cancer cells to enter the circulation, extravasate, avert stromal attack, resist metabolic stress, enter dormancy, evade immune surveillance, reinitiate tumor growth, secure a supportive niche, and co-opt organ-specific stromal components (2, 5, 101).

Adaptability is another key trait of the MIC phenotype. Disseminated cancer cells must adapt to a succession of rapidly changing physical, metabolic, and immune challenges. To survive these stresses, MICs adjust their phenotype accordingly. Phenotypic plasticity, the ability of cells to dynamically change their gene-expression programs, is an overarching hallmark of tumor progression and metastasis, and a source of adaptability for MICs (159, 160). Phenotypic plasticity enables cancer cells to undergo EMT, adapt to specific microenvironments, and resist therapy (161, 162). Moreover, plasticity accentuates the phenotypic heterogeneity of genomically diverse cancer cell populations (163).

In sum, MICs acquire different classes of traits from different sources (Fig. 4A): basic prometastatic traits acquired with the neoplastic phenotype, additional specialized traits acquired under selection for tumor regeneration in specific organs, and a plastic phenotype adept at deploying these traits under rapidly changing conditions as MICs attempt to regenerate the tumor in new locations.

Figure 4.

Acquisition of MIC phenotypes. A, MICs arise from primary tumors but must acquire distinct phenotypic traits to successfully disseminate, seed, and colonize distant organs. MICs adopt key phenotypic traits of regenerative progenitors that respond to tissue injury, including phenotypic plasticity, the ability to restore heterogeneous and morphologically complex epithelial structures upon disruption of tissue structure, and the ability to evade killing by immune cells. In addition, MICs must acquire organ-specific adaptations that enable colonization of distinct microenvironments. B, Models of metastasis evolution. In the “parallel evolution model,” cancer cells disseminating from the primary tumor early during cancer progression seed distant organs and evolve genetically, independently from the primary tumor. Thus, mutations in the primary and metastatic tumors would be expected to be different. In contrast, in “linear evolution models,” MICs disseminate late during tumor evolution from the primary tumor, and thus are closely clonally related to the primary tumor. Linear evolution can be “gradual,” in which case tumors may consist of multiple distinct subclones with similar fitness and individual metastases may be derived from distinct subclones, or “punctuated,” in which case a dominant subclone arising in the primary tumor rapidly outcompetes and overtakes the entire population and seeds all metastases. Dashed arrows represent the length of metastasis latency time periods.

Figure 4.

Acquisition of MIC phenotypes. A, MICs arise from primary tumors but must acquire distinct phenotypic traits to successfully disseminate, seed, and colonize distant organs. MICs adopt key phenotypic traits of regenerative progenitors that respond to tissue injury, including phenotypic plasticity, the ability to restore heterogeneous and morphologically complex epithelial structures upon disruption of tissue structure, and the ability to evade killing by immune cells. In addition, MICs must acquire organ-specific adaptations that enable colonization of distinct microenvironments. B, Models of metastasis evolution. In the “parallel evolution model,” cancer cells disseminating from the primary tumor early during cancer progression seed distant organs and evolve genetically, independently from the primary tumor. Thus, mutations in the primary and metastatic tumors would be expected to be different. In contrast, in “linear evolution models,” MICs disseminate late during tumor evolution from the primary tumor, and thus are closely clonally related to the primary tumor. Linear evolution can be “gradual,” in which case tumors may consist of multiple distinct subclones with similar fitness and individual metastases may be derived from distinct subclones, or “punctuated,” in which case a dominant subclone arising in the primary tumor rapidly outcompetes and overtakes the entire population and seeds all metastases. Dashed arrows represent the length of metastasis latency time periods.

Close modal

How do MICs acquire their unique adaptability and other specialized traits? Heterogeneous cancer cell populations evolve under continual selective pressures during tumor progression and metastasis. The identification of large numbers of tumor-associated mutations by large-scale genomic sequencing of matched normal and primary tumor tissues provides a basis for the notion that metastasis-specific traits may emerge by selection from the genetic heterogeneity present in cancer cell populations. However, the search for mutations that specifically drive metastasis has met with few examples to date.

Genetic Drivers of Metastasis

Studies seeking to identify metastasis-specific mutations pose several challenges (164, 165). Metastasis typically evolves in patients over several years, and this temporal dimension cannot be adequately reproduced in mouse models. On the other hand, patient-based studies are logistically cumbersome, complicated by interpatient heterogeneity, and subject to sampling bias if only a small region of a large tumor is biopsied for genomic analysis (166). Therapy is another important confounder, because patients who experience metastatic relapse after initial removal of primary tumors will have typically received systemic anticancer therapies, and such metastases reflect therapy resistance and may not represent the natural history of metastasis per se. Despite these hurdles, several recent studies involving multiregion sampling of matched primary and metastatic cancers biopsied from large numbers of patients have illuminated the genetics of metastatic evolution (162, 165, 167–172). Notably, across tumors, there appear to be few recurrent metastasis-specific somatic mutations, including single-nucleotide variants (SNV), insertions, and deletions. Where such mutations have been identified, their functional significance is frequently linked to expanding the activity of the oncogenic driving pathway or conferring resistance to therapy (166, 168, 173, 174). However, activation of certain general growth-promoting pathways may translate into organ-specific metastatic advantage. For example, mutations in PIK3CA have been proposed to favor brain metastasis of breast cancer by driving lipid synthesis (128).

The association of genetic mutations with metastatic progression reflects a selection of prometastatic traits or an association of bystander mutational events with clones that gain a prometastatic phenotype by other means. A recent study of 1,421 samples from 394 tumors across 22 tumor types identified increased clonality of somatic copy-number alterations (SCNA) in metastases versus primary tumors, potentially indicating an as yet inadequately understood role for large-scale genomic rearrangements in driving metastasis (175). In non–small cell lung cancer, elevated copy-number heterogeneity across multiple sequenced regions of early-stage primary tumors was associated with increased risk of relapse (176), suggesting that ongoing chromosomal instability is an important mechanism to generate evolutionary space for the emergence of prometastatic traits within tumors.

Modes of Evolution

Computational analyses of SCNAs and SNVs enable reconstruction of phylogenetic trees to determine the sequence and selection of mutations during metastasis (177). Two models for metastatic evolution have long been considered (178). The “linear evolution” model envisions that primary tumors gradually accumulate mutations, with a late subclone acquiring metastatic capacity (Fig. 4B). The “parallel evolution” model, on the other hand, argues that cancer cells that disseminate early are more effective at forming metastasis because they evolve in the host microenvironment, unlike clones that remain in the primary tumor (179). In HER2+ mouse models of metastatic breast cancer, early disseminating cells were found to be more metastatically competent than late disseminators (180, 181). Using sensitive cell-free DNA technology, circulating DNA with tumor-associated mutations can be detected in patients with subclinical primary tumors in a variety of cancers, suggesting that early dissemination is possible. However, the rarity of metastasis-specific SNVs in patients argues against the parallel evolution model (165, 167). Other studies have identified both metastasis-specific recurrent alternations and alterations that are shared between primary and metastatic tumors, suggesting that both early and late metastatic dissemination can occur (162). Moreover, recent work suggests the presence of “punctuated evolution” in some tumors, with acquisition of mutations that rapidly clonally dominate the primary tumor and then seed metastasis in an evolutionary “Big Bang” (169, 182–185). Other tumors may evolve more gradually, with multiple clones of low metastatic fitness competing within the primary tumor.

Taken together, the evidence from clinical and murine studies suggests that metastasis imposes an evolutionary bottleneck that only select cells within primary cancer cell populations are able to overcome. Such metastases may be monoclonal or polyclonal (169, 182–187), suggesting potentially distinct modes of seeding or selection. It is worth noting that the prevalent linear interpretation of the clonality and timing of metastases may be confounded by the possibility of ongoing “self-seeding” of metastases, wherein cells from metastatic tumors not only may recirculate and seed new metastatic lesions, but may also infiltrate preexisting primary and metastatic tumors (188, 189).

Evolutionary Timing and Cancer Therapy

Intriguingly, in clear cell renal cell carcinoma, dominant clones resulting from punctuated evolution seed aggressive multiorgan metastasis with little intermetastatic heterogeneity, whereas oligometastases with indolent behavior arise from tumors displaying gradual evolution (ref. 184; Fig. 4B). Unlike multiorgan metastasis, oligometastasis in a single organ may be amenable to curative surgery or other local treatments, and thus provides an opportunity to use tumor clonality as a predictive biomarker for such therapies (190). In patients, there is a strong correlation between tumor size (a proxy for tumor age) and the probability of future metastatic relapse, which is inconsistent with the possibility of exclusive early metastatic seeding. However, in a large cohort of patients with breast cancer treated with mastectomy and no systemic therapy, tumor size was associated with early local recurrence and distant metastasis, but not with late metastasis (191). This observation suggests the possibility that in some tumors both early and late dissemination may coexist, with late, more proliferative disseminating cells driving mostly rapid relapse. In contrast, early disseminating cells may be particularly prone to latency for extended periods of time, either because of a capacity to enter a deep quiescent state or because those that grow are rapidly cleared by the immune system.

Although genomic alterations generate selectable survival traits in cancer cells, mutations alone cannot account for the fundamentally distinct biology of MICs. Unlike primary tumors, which are thought to largely arise through oncogenic transformation of homeostatic tissue stem and progenitor cells in their native niche (192–196), MICs must rapidly change their phenotypes to detach from the primary tumor, survive mechanical, metabolic, and immunologic stresses during dissemination to distant organs, adapt to the foreign microenvironments, access proliferative cues to reinitiate tumor growth in distant organs, and resist therapy. Dynamic adaptation to changing circumstances during the metastatic process requires faster change than a purely genetic mechanism, which allows for selection of variants only once per cell cycle, might support.

Although the dynamic expression of specific genes has long been associated with metastatic phenotypes, the master transcription factors and epigenetic remodeling mechanisms required for the different steps of the metastatic process in various tumors are only beginning to emerge (197). Genes required for chromatin modification (e.g., ARID1A/B and KMT2C/D) and DNA methylation (DNMT) are frequently mutated in advanced tumors, underscoring the importance of epigenetic modulation in metastasis (171). A fundamental question is whether metastatic adaptability involves greater overall chromatin accessibility, and hence the potential to activate the expression of multiple genes, as implied by studies in genetically engineered mouse models of lung cancer (198–200), or whether the ability to transition to one or more prometastatic phenotypic states by activation of specific transcription factors is sufficient, as shown in models of pancreatic ductal adenocarcinoma (201) and renal cell carcinoma (202). Recent work illuminated the role of histone H3 variants (203) and specific histone marks, such as H3K36me2 (204), as critical requirements for epigenomic reprogramming during metastasis. The question remains whether epigenetic alterations favor metastasis by randomly activating selectable genes or by enabling cancer cells to enter progenitor states that have a superior fitness to initiate metastasis.

Several key traits of the MIC phenotype are normally present in regenerative progenitor cells. In wound-healing processes (Fig. 5A), epithelial regenerative progenitors show intense phenotypic plasticity, undergo EMT, display migratory activity, interact with and remodel the ECM, and secrete signals that co-opt mesenchymal, endothelial, and immune stromal components to support the restoration of epithelial barrier integrity (205, 206). Recent work raises the possibility that carcinoma MICs may capture these traits all at once by adopting a regenerative phenotype.

Figure 5.

Model of the regenerative progenitor origin of MICs. A, In the intestine, proliferative LGR5+ crypt base cells maintain intestinal homeostasis. Injury disrupts intercellular adherens junctions and induces the emergence of an L1CAM+ regenerative progenitor phenotype, which is required for epithelial repair, regeneration of heterogeneous cell types, and resolution of the wound. B, Oncogenic driver mutations in homeostatic stem and progenitor cells create tumor-initiating cells (or “cancer stem cells,” CSCs), capable of adenoma formation within an intact epithelial niche. Additional mutations give rise to an invasive carcinoma, which further breaches the integrity of the epithelium and triggers the emergence of an L1CAM+ regenerative progenitor state in cancer cells. As regenerative progenitors, these malignant cells have the capacity to survive the loss of the epithelial niche during tumor dissemination, are competent to enter quiescence, evade immune surveillance and therapy, and upon the receipt of favorable growth signals, regenerate heterogeneous tumors in metastatic sites and after therapy.

Figure 5.

Model of the regenerative progenitor origin of MICs. A, In the intestine, proliferative LGR5+ crypt base cells maintain intestinal homeostasis. Injury disrupts intercellular adherens junctions and induces the emergence of an L1CAM+ regenerative progenitor phenotype, which is required for epithelial repair, regeneration of heterogeneous cell types, and resolution of the wound. B, Oncogenic driver mutations in homeostatic stem and progenitor cells create tumor-initiating cells (or “cancer stem cells,” CSCs), capable of adenoma formation within an intact epithelial niche. Additional mutations give rise to an invasive carcinoma, which further breaches the integrity of the epithelium and triggers the emergence of an L1CAM+ regenerative progenitor state in cancer cells. As regenerative progenitors, these malignant cells have the capacity to survive the loss of the epithelial niche during tumor dissemination, are competent to enter quiescence, evade immune surveillance and therapy, and upon the receipt of favorable growth signals, regenerate heterogeneous tumors in metastatic sites and after therapy.

Close modal

Phenotypic Plasticity in Regenerative Progenitors

Phenotypic plasticity, a key feature of tumor progression and metastasis (159, 160), is also a feature of normal adult stem cells, including those of the gut, lung, and skin. These cells are not locked in a fixed preexisting cell fate but rather enter and exit “stem-like” states in response to demands to preserve the structural and functional integrity of the tissue (4, 160, 207–209). In the intestinal mucosa, cells expressing the WNT signaling pathway component LGR5 function as stem cells in the maintenance of epithelial homeostasis (210). However, when LGR5+ cells are lost by genetic ablation or injury, LGR5 progenitor cells can become LGR5+ cells (206, 209). Moreover, unique stem cell states are mobilized by damage to revive the epithelium through transcription factor YAP (211).

Cells from the mouse small intestine can grow as organoids, three-dimensional structures that form in laminin-rich basement membrane matrix and mimic the cellular composition and architecture of the intestinal epithelium (212). Both LGR5+ and LGR5 cells sorted from normal mouse intestinal epithelium can form organoids (209, 213). Moreover, during the initial stages of organoid formation, LGR5+ cells convert to an LGR5 state that includes YAP activation, and later reexpress LGR5 and downregulate YAP to establish a differentiated epithelium (214). Thus, intestinal organoid formation appears to start as a process of epithelium recovery driven by LGR5 progenitors followed by restoration of differentiated structures driven by reemerging LGR5+ progenitors.

Dynamic phenotypic changes are also observed in cancer metastasis. In colorectal cancer, intestinal LGR5+ stem cells harboring constitutively activating mutations in the WNT signaling pathway act as tumor-initiating CSCs (195). However, intravital microscopy of experimental colorectal cancer metastasis in mice revealed that MICs disseminating from the primary tumor and seeding metastasis in the liver are predominantly LGR5 (214). To initiate metastatic tumor growth in the liver, some progeny of LGR5 cells reacquire LGR5 expression (214), whereas other cancers may acquire LGR5-independent regenerative capacity (215, 216).

Adopting a Regenerative Phenotype

Recent work has shown that MICs in colorectal cancer and other epithelial cancers dynamically express L1CAM (32). Originally identified as a neuronal cell-adhesion molecule, L1CAM is expressed in carcinomas at the primary tumor-invasion front and is associated with poor prognosis in multiple tumor types, including those of the breast, lung, colon, ovary, and kidney (217). L1CAM is not expressed in intact normal epithelia or required for epithelial homeostasis, but its expression is induced by the breach of epithelial intercellular adherens junctions during colitis and tissue injury, and is required for the survival of detached epithelial progenitors and for tissue repair (32). L1CAM expression in dissociated colorectal cancer cells is required for the initiation of organoid formation, where most L1CAM+ cells are LGR5. L1CAM+ cells are highly competent to initiate growth of transplanted colorectal cancer tumors and liver metastases. As MICs initiate tumor growth and regain epithelial structures, L1CAM expression is downregulated, but can be reactivated again when tumor architecture is disrupted by therapy. Repeated cycles of therapy select for cells with the capacity to upregulate L1CAM expression, which can more efficiently reinitiate tumor growth (32). Thus, L1CAM seems to mark certain regenerative progenitor states, and its expression in these cells mediates the regrowth of intestinal epithelium after injury and tumor regrowth during metastasis.

Several studies highlight a biphasic process of epithelial repair that is co-opted during metastasis. Upon wounding, an immediate emergency response is activated in which epithelial progenitors enter distinct migratory and proliferate states to seal the breach in the mucosa (refs. 218, 219; Fig. 5A). This “band-aid” phase is followed by a slower regenerative phase in which normal tissue stem/progenitor hierarchies are recreated. Borrowing from repair, dissociation of epithelial structures in invasive colorectal cancer induces malignant progenitors to adopt a highly plastic regenerative phenotype endowed with the capacity to undergo EMT, migrate, avert anoikis, and drive L1CAM-dependent regrowth (refs. 32, 43; Fig. 5B).

Further evidence for the regenerative nature of metastatic progression comes from single-cell RNA-sequencing studies on tumor tissue from patients with lung adenocarcinoma as well as lesions from a mouse model of metastatic latency and outbreak (220). Whereas primary tumors showed developmental lineage promiscuity and cell states that were intermediate between the various cell states of the regenerative lung epithelium, metastatic cells reverted to earlier states and recapitulated lung organogenesis and regeneration phenotypes with striking fidelity.

The adoption of a regenerative phenotype by MICs has several implications. Instead of having to select a predetermined rigid MIC fate or randomly acquiring a multitude of independent metastatic traits, colorectal cancer cells seem to sample among a range of phenotypically transient states, some of which, such as the L1CAM+ state, are adept at supporting survival of disseminated cells and the initiation of metastatic growth and are hence selected for during the metastatic process (32, 43).

Acquiring Organ-Specific Colonization Traits

The highly specialized nature of most organ-specific metastatic traits suggests that these are acquired through selection from heterogeneous MIC populations that complete their evolution after seeding distant organs, rather than being intrinsic properties of the stem cell states that give rise to MICs. However, not all organ-specific traits result from clonal evolution after dissemination. The microenvironment of a primary tumor can preselect clones with properties that are compatible with the environment of a particular distant organ, thus influencing the metastatic tropism of the disseminating cancer cells. For example, primary breast tumors with high TGFβ activity are associated with lung relapse, and this is linked to the ability of TGFβ to induce the expression of the cytokine ANGPTL4 in the cancer cells. ANGPTL4 causes endothelium disjunction to promote extravasation of circulating breast cancer cells that lodge in lung capillaries (221). Primary tumors may also preselect for MIC clones that thrive in distant organs whose stroma the primary tumor mimics. For example, breast carcinomas rich in mesenchymal cells that produce CXCL12 select for MIC clones that thrive in a CXCL12-rich microenvironment, and thus are predestined to colonize the CXCL12-rich stem cell niches of the bone marrow (222).

Metastasis is subject to important organ-specific barriers and adaptation, as mentioned in previous sections and recapitulated in Fig. 6A. However, metastasis is by definition a systemic disease (223). Despite the growing understanding of MIC-intrinsic and metastatic niche–dependent mechanisms, relatively little is known about the complex ways in which systemic physiologic factors influence MIC plasticity and selection, and vice versa. However, recent studies have provided evidence for the potential role of various systemic signals in the development of metastasis (Fig. 6B). Although the molecular basis for a majority of these effects remains to be determined, and their causal relationship with metastatic progression remains to be firmly established, this is an area of rising interest.

Figure 6.

Metastasis as a systemic disease with organ-specific features. A, A recap of organ-specific determinants of metastatic colonization. B, Systemic determinants of metastasis. During metastasis, cancer cells from primary tumors disseminate throughout the body via the blood and lymphatic circulations, among other routes. Tumor cells establish a dynamic equilibrium with both systemic immunity and organ-specific immune infiltrates. Systemic factors can modulate both primary and metastatic tumor growth as well as the antitumor immune responses. Although the catalog of systemic factors that can modulate metastasis is incomplete, current evidence implicates circulating chemokines and cytokines and signals transmitted via nerves and hormones in controlling tumor growth and metastasis. In addition, intratumoral and gut microbiomes can influence cancer progression either through direct interactions with cancer cells and immune cells, or indirectly, via metabolites released into the systemic circulation. The dynamic equilibrium between primary tumors, metastases, and their niches is influenced by systemic factors that affect MIC growth and therapy resistance.

Figure 6.

Metastasis as a systemic disease with organ-specific features. A, A recap of organ-specific determinants of metastatic colonization. B, Systemic determinants of metastasis. During metastasis, cancer cells from primary tumors disseminate throughout the body via the blood and lymphatic circulations, among other routes. Tumor cells establish a dynamic equilibrium with both systemic immunity and organ-specific immune infiltrates. Systemic factors can modulate both primary and metastatic tumor growth as well as the antitumor immune responses. Although the catalog of systemic factors that can modulate metastasis is incomplete, current evidence implicates circulating chemokines and cytokines and signals transmitted via nerves and hormones in controlling tumor growth and metastasis. In addition, intratumoral and gut microbiomes can influence cancer progression either through direct interactions with cancer cells and immune cells, or indirectly, via metabolites released into the systemic circulation. The dynamic equilibrium between primary tumors, metastases, and their niches is influenced by systemic factors that affect MIC growth and therapy resistance.

Close modal

Systemic Niche Components

Both primary and metastatic tumors can release cytokines, chemokines, and hormones either directly into the systemic circulation or packaged into exosomes, which in turn can modulate distant niches to favor or resist metastatic colonization, including organ-specific metastatic dissemination (146, 150, 224, 225). Although many critical mediators and mechanisms of premetastatic niche formation remain to be uncovered, studies to date suggest that organ-specific stromal cell reprogramming induces changes in ECM and matrix-associated proteins, which in turn enable recruitment of specific myeloid populations that support future MIC growth (226). Indeed, bone marrow–derived myeloid cells can play critical roles in either limiting or promoting MIC survival and growth, and the mechanisms enabling their context-specific recruitment, differentiation, and activity at various steps in the metastatic process have been a focus of growing interest (18, 97, 129).

Systemic Immunity, Inflammation, and Metabolism

The critical role of systemic immunity in controlling metastasis is now firmly established. Physiologic and pathologic phenomena that modulate systemic immunity indirectly control metastasis, in addition to having direct effects on MICs (Fig. 6). Stress and depression have been associated with early metastasis and poor prognosis in patients with breast, lung, and hepatobiliary carcinomas (227). In cell line xenograft models of metastatic breast cancer, increased corticosteroid stress hormone levels generated during tumor progression resulted in increased glucocorticoid receptor activation, downstream ROR1 upregulation, metastatic growth, and chemoresistance (228). In addition to direct effects on MICs, glucocorticoids have widespread immunosuppressive effects, which in turn can promote MIC outgrowth (229, 230). In addition to steroids, increased levels of the stress-induced catecholamine norepinephrine induced changes in neutrophils that enabled MIC escape from dormancy in syngeneic lung cancer models (231).

Systemic inflammation and immunosuppression resulting from a range of physiologic and pathologic conditions, including aging, postpartum breast involution, asthma, and osteoarthritis, have been associated with increased metastasis risk and reduced survival from a range of epithelial cancers (232–236). In a recent study, inflammation induced by surgical wounding was shown to promote MIC outgrowth from dormancy in breast cancer mouse models (237), suggesting a potential explanation for the early peak in metastatic recurrence within one year of curative breast surgery in patients (238). Based on these observations, several groups have proposed the prophylactic use of nonsteroidal anti-inflammatory agents and beta-blockers in the perioperative setting, although clear clinical evidence for a significant antimetastatic effect is lacking (237, 239).

Tumors in turn may alter host metabolism, immunity, and physiology locally as well as systemically (Fig. 6). Circulating cytokines and metabolites derived from metastatic tumors can have a dramatic impact on tissues such as skeletal muscle and adipose. Indeed, many patients with metastatic cancer develop cachexia, a debilitating muscle-wasting syndrome that is associated with poor prognosis and accelerated death (240). A better understanding of the molecular basis for how metastatic disease promotes cachexia is important for improving the care of patients with metastatic cancer.

Nerves and Metastasis

The nervous system is emerging as a potential systemic modulator of metastasis (241, 242). Innervation is essential for physiologic regeneration (243), but the role of nerves in metastasis had long been surmised to be only as passive conduits for tumor dissemination in cases of perineural invasion (61). Growing evidence, however, suggests that neurons can be active determinants of cancer progression and outcome (refs. 241, 242; Fig. 6). Direct innervation of tumors can contribute to tumor progression at both the primary and metastatic sites. In mouse cancer models, autonomic denervation of the prostate, stomach, and pancreas inhibited tumor progression and cell dissemination from these sites (244). In addition to effects mediated by direct tumor innervation, increasing understanding of neuronal regulation of immunity suggests a systemic means of neuronal metastatic regulation, although little mechanistic insight is currently available (245). For example, in a syngeneic metastatic mammary carcinoma model, denervation of sensory neurons suppressed NK cell activity, resulting in increased lung and cardiac metastases (246).

Diet and Exercise

Epidemiologic and experimental studies have identified correlations between lifestyle factors such as diet and exercise with cancer incidence, therapy resistance, and survival (247–249). Proposed mechanisms of tumor modulation include direct effects on cancer cells (for example, by altering insulin signaling or lipid metabolism), indirect effects on tumor inflammation and immune surveillance, or, especially for diet, tertiary effects mediated by the gut microbiome (127, 250–255). Obesity, which is associated with both poor diet and low exercise, is an independent risk factor for the future development of distant metastasis and death in patients with surgically resected primary breast cancer (254). With some notable exceptions (e.g., refs 127, 255), the majority of animal studies of diet, exercise, and microbiome modulation to date have been performed in models of tumor initiation or subcutaneous tumor cell transplantation, and the functional and mechanistic significance of these studies to the critical steps of MIC dissemination, dormancy, and outbreak, as well as therapy resistance in patients with advanced cancer, remain unresolved.

It is important to discern whether lifestyle factors that may influence tumor progression represent modifiable, and potentially therapeutically targetable, risk factors in patients who have already been diagnosed with primary tumors. A meta-analysis of 49,095 early-stage breast and colon cancer survivors suggested that survivors who increased their physical activity from before diagnosis and surgery showed a statistically significant decreased total mortality risk in comparison with those who did not change their activity level (256). Prospective clinical studies randomizing newly diagnosed patients with cancer to specific dietary or exercise interventions will help clarify the actionability of these factors in preventing and treating metastasis (257).

Microbiome

The role of the microbiome as a player in influencing cancer biology is becoming increasingly evident (refs. 258, 259; Fig. 6). The mucosal microbiome can interact directly with epithelial cells in niches such as the luminal gastrointestinal tract, respiratory tract, and skin to influence tumor progression through local inflammation or immunosuppression (258, 259). Gut pathobiotic strains of fusobacterium have been shown to colonize colorectal tumors and drive metastasis to the liver (260). Mouse xenografts derived from such human liver metastases continue to be colonized by fusobacterium, and antibiotics that target fusobacterium can inhibit metastasis in mouse models (260). Intriguingly, many cancers that are not directly exposed to environmental bacteria, including breast and bone cancers, have recently been shown to also harbor intratumoral microbiota, although the functional significance of such colonization remains unclear (261). In addition to local effects, metabolites secreted by microbiota may have diverse systemic effects on immunity, hormones, and organ function, most of which are currently poorly understood (262, 263). Other systemic factors, such as diet, can rapidly alter microbiome composition (264, 265), and patient-specific microbiota can in turn influence responses to therapy (266, 267). Given this complexity, there is an increasingly evident need to characterize and control for the microbiome composition as an independent variable in animal and human studies of metastasis.

Aging

In some cancers, such as melanoma, age at cancer diagnosis is associated with increased tumor aggression and metastasis (268). Three broad mechanisms have been proposed for age-related local and systemic acceleration of metastasis (269, 270). First, age-dependent accumulation of senescent cells, and their associated secretomes, in the tumor microenvironment may promote tumor invasion, premetastatic niche formation, and metastatic outgrowth (271–273). Coinjection of senescent fibroblasts, but not normal fibroblasts, promoted metastatic progression in mouse models (274). Second, age-related deterioration of extracellular collagen matrices may facilitate tumor cell invasion and promote transendothelial migration (275, 276). Third, age-associated systemic low-grade chronic inflammation, termed “inflammaging” (277), can disrupt tissue architecture and promote tumor dissemination (275, 278), whereas immunosenescence may prevent effective immune editing of disseminated MICs emerging from dormancy (270, 279). Further delineation of the mechanisms by which aging intersects with metastatic progression is needed to define therapeutic strategies to prevent and treat cancer in aging populations.

A cardinal feature of metastatic disease is its ability to resist therapy. We recently reviewed the clinical contexts and approaches for targeting metastasis (280). Here we focus on the properties of MICs that enable therapy resistance and potential approaches to overcome these.

Because tumors comprise numerous genetically distinct subclones, cells harboring therapy resistance mutations can be selected for during metastasis as the patient undergoes treatment for initially localized disease. Genetic analyses of matched primary tumors and metastases show that metastases from patients who had received adjuvant therapy for treatment of initially localized cancer had reduced clonal heterogeneity in comparison with metastases from previously untreated patients, suggesting that treatment imposes an evolutionary bottleneck for MIC emergence (171). This is particularly evident in the case of treatments that target specific oncogenic signaling pathways, where therapy with inhibitors of these pathways selects for the emergence of cancer cells with specific recurrent resistance mutations. Examples include EGFRT790M mutations in patients treated with the first-generation tyrosine kinase inhibitors gefitinib and erlotinib (281), and ESR1 mutations in patients with estrogen receptor–positive breast cancer treated with antiestrogen therapy (173). Clinical and experimental studies characterizing such targeted therapy–specific resistance mutations have enabled the rapid development of improved drugs that anticipate and target resistance mutations, with improved therapeutic results (282).

In addition to these genetically acquired mechanisms of drug resistance, MIC phenotypic properties may confer intrinsic therapy resistance (153, 280, 283). In most cancers, the mainstay of the treatment of metastasis remains cytotoxic chemotherapy, which preferentially targets rapidly proliferating cells. Thus, MICs that are capable of entering quiescence may more readily evade chemotherapy than do proliferative cancer cells. When therapies result in partial destruction of glandular metastatic tumors, they may in turn disrupt epithelial adherens junctions and induce MICs to enter a regenerative progenitor state, as observed in colorectal cancer (32). Successive rounds of therapy may further select for residual cells that are epigenetically primed to reenter regenerative states (284), suggesting an explanation for the successively restricted duration of clinical response typically observed with sequential lines of therapy in patients with advanced cancer. In addition, the stem-like properties of MICs may include the expression of ATP-binding cassette drug efflux transporters that could confer multidrug resistance (285). Dying or senescent cancer cells induced by therapy that partially debulks metastatic tumors can release cytokines as secretomes, to which MICs can respond by activating growth factor signaling programs to regenerate tumors (286–288).

The immune-evasive properties of MICs are likely also of importance in resisting both tumor cell–directed therapy and systemic immunotherapy. In addition to MIC-intrinsic mechanisms of immune escape, desmoplasia in tumors is an important mechanism of therapy resistance. In pancreatic ductal adenocarcinoma, Hedgehog signaling induces fibrosis, which in turn poses a physical barrier for drug delivery to tumor cells that are surrounded by fibroblasts (20, 289, 290). In many cancer types, immune cells are recruited to tumors from the systemic circulation but are spatially segregated from tumor cells by fibroblast barriers, resulting in immunologically “cold” tumors that do not respond to PD-1 checkpoint immunotherapy (291, 292). Targeting these fibroblast barriers using Hedgehog inhibitors disrupts desmoplasia in mouse models of pancreatic ductal adenocarcinoma (290). TGFβ is strongly immunosuppressive and fibrogenic (88, 89); the use of TGFβ inhibitors in colorectal and breast cancers increases drug and immune cell infiltration into the tumor, reversing immunotherapy resistance in preclinical models and correlating with superior immunotherapy responses in patients (293, 294). However, the translation of these approaches to patients has been challenging to date due to complex effects of drugs that target developmental signaling pathways on the tumor microenvironment, as well as broader systemic toxicities (295–297). Improved understanding of the mechanisms of tumor fibrogenesis and immune cell communication may yield more specific targets for therapeutic intervention.

The plasticity of MICs arguably poses the greatest challenge to therapeutic success, because MICs that are capable of sampling among numerous transcriptional and epigenetic states may be surmised to rapidly select for gene-expression programs that enable therapeutic escape. Therefore, understanding the molecular mechanisms of MIC plasticity may yield improved therapeutics (160, 298). For example, epigenetic modulators could be used to inhibit or reverse MIC plasticity or to force MIC differentiation into terminal, nonproliferative states. Targeting genotype-agnostic conserved regenerative states may block the relapse of metastatic disease that is being debulked by antineoplastic chemotherapy. Characterizing the endpoints of plasticity, such as neuroendocrine lineage plasticity in prostate cancer, may enable the development of new drugs that target the emergent neuroendocrine state instead of or in addition to the preexisting androgen-driven epithelial state (161).

In summary, recent work is illuminating the fundamental principles of MICs and their ecosystems, and importantly shedding light on the mechanisms by which metastatic tumors emerge and develop as phenotypically distinct, therapy-resistant entities in comparison with primary tumors. In brief:

  • Metastasis develops through three distinct phases, namely dissemination, dormancy, and outbreak, which can coexist until the removal of the primary tumor.

  • Each phase of metastasis reflects a dynamic equilibrium of MICs with host immunity, with the eventual outbreak of MICs as macrometastatic colonies reflecting a failure of immune surveillance.

  • Although preexisting genetically distinct tumor subclones with high metastatic propensity are selected for in primary tumors, phenotypic plasticity, enabled by epigenetic and metabolic adaptations, is an overarching hallmark of MICs.

  • Phenotypic plasticity endows MICs with the ability to sample multiple transcriptional states and dynamically adapt to diverse stresses during each phase of metastasis.

  • MICs can adopt epithelial regenerative progenitor states that possess stem-like, immune-evasive, tissue-regenerative, and therapy-resistance capacities.

  • The dynamic equilibrium between MICs and their organ-specific microenvironmental niches can be modulated by systemic factors that affect either or both components.

  • Identifying the vulnerabilities of conserved regenerative MIC states, and the molecular determinants of communication between MICs and their ecosystems, could yield improved therapeutics to prevent and treat metastasis, and thus improve outcomes for patients with advanced cancer.

With the advent of immunotherapy and continued advances in targeted therapy, the last two decades have witnessed considerable improvements in outcomes of patients with some types of metastatic cancers (268). These advances are driven by increasingly closer collaborative bidirectional interactions between preclinical and clinical scientists, and improved, more representative patient-derived and animal models of metastatic cancer. Significant therapeutic advances are resulting from the rapid recognition, molecular characterization, and clinical translation of biologically discrete phenotypes of metastatic disease. Examples include curative local therapy for oligometastatic disease, immune activation phenotypes in extreme outlier patients with exceptional response to therapy, and mechanism-based therapies for metastases in specific organs, such as the brain, where even small improvements in tumor shrinkage can yield significant improvements in patient survival (280).

Further improvements in patient outcomes will emerge from a deeper understanding of the mechanisms that underlie the plasticity and dormancy of MICs as well as the complex interactions between the MICs, their niches, and systemic factors. To do so, preclinical models used to study metastasis and test therapies for metastatic cancer must recapitulate the genomic complexity, plasticity, and niche biology of advanced human cancers, rather than relying exclusively on simpler systems that may fail to capture these critical features.

In 1889, Steven Paget first hypothesized that metastasis might result from a fertile “seed” landing on congenial niche “soil” (299). Although investigations in the subsequent 132 years have vindicated Paget's view, a third dimension of the metastasis ecosystem—the “forest” of systemic factors—is emerging. By understanding and targeting the interactions between the seeds, soil, and forest, we may be able to convert metastasis, which is today still typically a death sentence, into a manageable illness.

J. Massagué reports grants from NIH during the conduct of the study; and personal fees from Scholar Rock Inc and Biogen Inc. outside the submitted work; in addition, J. Massagué has a patent for US7829066 issued, a patent for EU2831593 issued, a patent for EU3047039 issued, a patent for US10413522 issued, a patent for PCT/US2016/062880 pending, and a patent for PCT/US2017/045145 pending; and owns Scholar Rock Inc. stock. K. Ganesh reports grants from NIH, the Damon Runyon Cancer Research Foundation, and Burroughs Wellcome Funds during the conduct of the study; in addition, K. Ganesh has a patent for PCT/US2017/045145 pending.

The authors' work in the subject area of this review is supported by NIH grants R35CA252978 (J. Massagué), K08CA23021 (K. Ganesh), and P30-CA008748 (MSKCC), and the Alan and Sandra Gerry Metastasis and Tumor Ecosystems Center at MSKCC (J. Massagué).

1.
Lambert
AW
,
Pattabiraman
DR
,
Weinberg
RA
. 
Emerging biological principles of metastasis
.
Cell
2017
;
168
:
670
91
.
2.
Massagué
J
,
Obenauf
AC
. 
Metastatic colonization by circulating tumour cells
.
Nature
2016
;
529
:
298
306
.
3.
Oskarsson
T
,
Batlle
E
,
Massagué
J
. 
Metastatic stem cells: sources, niches, and vital pathways
.
Cell Stem Cell
2014
;
14
:
306
21
.
4.
Batlle
E
,
Clevers
H
. 
Cancer stem cells revisited
.
Nat Med
2017
;
23
:
1124
34
.
5.
Celià-Terrassa
T
,
Kang
Y
. 
Metastatic niche functions and therapeutic opportunities
.
Nat Cell Biol
2018
;
20
:
868
77
.
6.
Hüsemann
Y
,
Geigl
JB
,
Schubert
F
,
Musiani
P
,
Meyer
M
,
Burghart
E
, et al
Systemic spread is an early step in breast cancer
.
Cancer Cell
2008
;
13
:
58
68
.
7.
Sänger
N
,
Effenberger
KE
,
Riethdorf
S
,
Van Haasteren
V
,
Gauwerky
J
,
Wiegratz
I
, et al
Disseminated tumor cells in the bone marrow of patients with ductal carcinoma in situ
.
Int J Cancer
2011
;
129
:
2522
6
.
8.
Stott
SL
,
Lee
RJ
,
Nagrath
S
,
Yu
M
,
Miyamoto
DT
,
Ulkus
L
, et al
Isolation and characterization of circulating tumor cells from patients with localized and metastatic prostate cancer
.
Sci Transl Med
2010
;
2
:
25ra3
.
9.
Risson
E
,
Nobre
AR
,
Maguer-Satta
V
,
Aguirre-Ghiso
JA
. 
The current paradigm and challenges ahead for the dormancy of disseminated tumor cells
.
Nature Cancer
2020
;
1
:
672
80
.
10.
Varadhachary
GR
. 
Carcinoma of unknown primary origin
.
Gastrointest Cancer Res
2007
;
1
:
229
35
.
11.
Lin
NU
,
Winer
EP
. 
Brain metastases: the HER2 paradigm
.
Clin Cancer Res
2007
;
13
:
1648
55
.
12.
Sleeman
JP
,
Nazarenko
I
,
Thiele
W
. 
Do all roads lead to Rome? Routes to metastasis development
.
Int J Cancer
2011
;
128
:
2511
26
.
13.
Reymond
N
,
d'Agua
BB
,
Ridley
AJ
. 
Crossing the endothelial barrier during metastasis
.
Nat Rev Cancer
2013
;
13
:
858
70
.
14.
Labelle
M
,
Hynes
RO
. 
The initial hours of metastasis: the importance of cooperative host-tumor cell interactions during hematogenous dissemination
.
Cancer Discov
2012
;
2
:
1091
9
.
15.
Kai
F
,
Drain
AP
,
Weaver
VM
. 
The extracellular matrix modulates the metastatic journey
.
Dev Cell
2019
;
49
:
332
46
.
16.
Hamidi
H
,
Ivaska
J
. 
Every step of the way: integrins in cancer progression and metastasis
.
Nat Rev Cancer
2018
;
18
:
533
48
.
17.
Yang
J
,
Antin
P
,
Berx
G
,
Blanpain
C
,
Brabletz
T
,
Bronner
M
, et al
Guidelines and definitions for research on epithelial-mesenchymal transition
.
Nat Rev Mol Cell Biol
2020
;
21
:
341
52
.
18.
Quail
DF
,
Joyce
JA
. 
Microenvironmental regulation of tumor progression and metastasis
.
Nat Med
2013
;
19
:
1423
37
.
19.
Pignatelli
J
,
Bravo-Cordero
JJ
,
Roh-Johnson
M
,
Gandhi
SJ
,
Wang
Y
,
Chen
X
, et al
Macrophage-dependent tumor cell transendothelial migration is mediated by Notch1/Mena(INV)-initiated invadopodium formation
.
Sci Rep
2016
;
6
:
37874
.
20.
Yauch
RL
,
Gould
SE
,
Scales
SJ
,
Tang
T
,
Tian
H
,
Ahn
CP
, et al
A paracrine requirement for hedgehog signalling in cancer
.
Nature
2008
;
455
:
406
10
.
21.
Nieto
MA
,
Huang
RY
,
Jackson
RA
,
Thiery
JP
. 
EMT: 2016
.
Cell
2016
;
166
:
21
45
.
22.
Denais
CM
,
Gilbert
RM
,
Isermann
P
,
McGregor
AL
,
te Lindert
M
,
Weigelin
B
, et al
Nuclear envelope rupture and repair during cancer cell migration
.
Science
2016
;
352
:
353
8
.
23.
van Helvert
S
,
Storm
C
,
Friedl
P
. 
Mechanoreciprocity in cell migration
.
Nat Cell Biol
2018
;
20
:
8
20
.
24.
Alix-Panabieres
C
,
Schwarzenbach
H
,
Pantel
K
. 
Circulating tumor cells and circulating tumor DNA
.
Annu Rev Med
2012
;
63
:
199
215
.
25.
Pantel
K
,
Alix-Panabières
C
. 
Liquid biopsy and minimal residual disease–latest advances and implications for cure
.
Nat Rev Clin Oncol
2019
;
16
:
409
24
.
26.
Fidler
IJ
. 
The relationship of embolic homogeneity, number, size and viability to the incidence of experimental metastasis
.
Eur J Cancer
1973
;
9
:
223
7
.
27.
Aceto
N
,
Toner
M
,
Maheswaran
S
,
Haber
DA
. 
En route to metastasis: circulating tumor cell clusters and epithelial-to-mesenchymal transition
.
Trends Cancer
2015
;
1
:
44
52
.
28.
Duda
DG
,
Duyverman
AM
,
Kohno
M
,
Snuderl
M
,
Steller
EJ
,
Fukumura
D
, et al
Malignant cells facilitate lung metastasis by bringing their own soil
.
Proc Natl Acad Sci U S A
2010
;
107
:
21677
82
.
29.
Wrenn
ED
,
Yamamoto
A
,
Moore
BM
,
Huang
Y
,
McBirney
M
,
Thomas
AJ
, et al
Regulation of collective metastasis by nanolumenal signaling
.
Cell
2020
;
183
:
395
410
.
30.
Campbell
NR
,
Rao
A
,
Zhang
M
,
Baron
M
,
Heilmann
S
,
Deforet
M
, et al
Cell state diversity promotes metastasis through heterotypic cluster formation in melanoma
.
bioRxiv
2020
:
2020.08.24.265140
.
31.
Gkountela
S
,
Castro-Giner
F
,
Szczerba
BM
,
Vetter
M
,
Landin
J
,
Scherrer
R
, et al
Circulating tumor cell clustering shapes DNA methylation to enable metastasis seeding
.
Cell
2019
;
176
:
98
112
.
32.
Ganesh
K
,
Basnet
H
,
Kaygusuz
Y
,
Laughney
AM
,
He
L
,
Sharma
R
, et al
L1CAM defines the regenerative origin of metastasis-initiating cells in colorectal cancer
.
Nature Cancer
2020
;
1
:
28
45
.
33.
Haemmerle
M
,
Stone
RL
,
Menter
DG
,
Afshar-Kharghan
V
,
Sood
AK
. 
The platelet lifeline to cancer: challenges and opportunities
.
Cancer Cell
2018
;
33
:
965
83
.
34.
Padmanaban
V
,
Krol
I
,
Suhail
Y
,
Szczerba
BM
,
Aceto
N
,
Bader
JS
, et al
Neutrophils escort circulating tumour cells to enable cell cycle progression
.
Nature
2019
;
573
:
439
44
.
35.
Szczerba
BM
,
Castro-Giner
F
,
Vetter
M
,
Krol
I
,
Gkountela
S
,
Landin
J
, et al
Neutrophils escort circulating tumour cells to enable cell cycle progression
.
Nature
2019
;
566
:
553
7
.
36.
Valiente
M
,
Obenauf
AC
,
Jin
X
,
Chen
Q
,
Zhang
XH
,
Lee
DJ
, et al
Serpins promote cancer cell survival and vascular co-option in brain metastasis
.
Cell
2014
;
156
:
1002
16
.
37.
Bos
PD
,
Zhang
XH
,
Nadal
C
,
Shu
W
,
Gomis
RR
,
Nguyen
DX
, et al
Genes that mediate breast cancer metastasis to the brain
.
Nature
2009
;
459
:
1005
9
.
38.
Boire
A
,
Zou
Y
,
Shieh
J
,
Macalinao
DG
,
Pentsova
E
,
Massagué
J
. 
Complement component 3 adapts the cerebrospinal fluid for leptomeningeal metastasis
.
Cell
2017
;
168
:
1101
13
.
39.
Achrol
AS
,
Rennert
RC
,
Anders
C
,
Soffietti
R
,
Ahluwalia
MS
,
Nayak
L
, et al
Brain metastases
.
Nat Rev Dis Primers
2019
;
5
:
5
.
40.
Strilic
B
,
Yang
L
,
Albarran-Juarez
J
,
Wachsmuth
L
,
Han
K
,
Muller
UC
, et al
Tumour-cell-induced endothelial cell necroptosis via death receptor 6 promotes metastasis
.
Nature
2016
;
536
:
215
8
.
41.
Kienast
Y
,
von Baumgarten
L
,
Fuhrmann
M
,
Klinkert
WE
,
Goldbrunner
R
,
Herms
J
, et al
Real-time imaging reveals the single steps of brain metastasis formation
.
Nat Med
2010
;
16
:
116
22
.
42.
Im
JH
,
Fu
W
,
Wang
H
,
Bhatia
SK
,
Hammer
DA
,
Kowalska
MA
, et al
Coagulation facilitates tumor cell spreading in the pulmonary vasculature during early metastatic colony formation
.
Cancer Res
2004
;
64
:
8613
9
.
43.
Er
EE
,
Valiente
M
,
Ganesh
K
,
Zou
Y
,
Agrawal
S
,
Hu
J
, et al
Pericyte-like spreading by disseminated cancer cells activates YAP and MRTF for metastatic colonization
.
Nat Cell Biol
2018
;
20
:
966
78
.
44.
Ghajar
CM
,
Peinado
H
,
Mori
H
,
Matei
IR
,
Evason
KJ
,
Brazier
H
, et al
The perivascular niche regulates breast tumour dormancy
.
Nat Cell Biol
2013
;
15
:
807
17
.
45.
Wang
H
,
Fu
W
,
Im
JH
,
Zhou
Z
,
Santoro
SA
,
Iyer
V
, et al
Tumor cell alpha3beta1 integrin and vascular laminin-5 mediate pulmonary arrest and metastasis
.
J Cell Biol
2004
;
164
:
935
41
.
46.
Naxerova
K
. 
Defining the role of lymph node metastasis in systemic breast cancer evolution
.
EBioMedicine
2020
;
57
:
102852
.
47.
Tjan-Heijnen
V
,
Viale
G
. 
The lymph node and the metastasis
.
N Engl J Med
2018
;
378
:
2045
6
.
48.
Reiter
JG
,
Hung
WT
,
Lee
IH
,
Nagpal
S
,
Giunta
P
,
Degner
S
, et al
Lymph node metastases develop through a wider evolutionary bottleneck than distant metastases
.
Nat Genet
2020
;
52
:
692
700
.
49.
Naxerova
K
,
Reiter
JG
,
Brachtel
E
,
Lennerz
JK
,
van de Wetering
M
,
Rowan
A
, et al
Origins of lymphatic and distant metastases in human colorectal cancer
.
Science
2017
;
357
:
55
60
.
50.
Venet
D
,
Fimereli
D
,
Rothe
F
,
Boeckx
B
,
Maetens
M
,
Majjaj
S
, et al
Phylogenetic reconstruction of breast cancer reveals two routes of metastatic dissemination associated with distinct clinical outcome
.
EBioMedicine
2020
;
56
:
102793
.
51.
Giuliano
AE
,
Hunt
KK
,
Ballman
KV
,
Beitsch
PD
,
Whitworth
PW
,
Blumencranz
PW
, et al
Axillary dissection vs no axillary dissection in women with invasive breast cancer and sentinel node metastasis: a randomized clinical trial
.
JAMA
2011
;
305
:
569
75
.
52.
Faries
MB
,
Thompson
JF
,
Cochran
AJ
,
Andtbacka
RH
,
Mozzillo
N
,
Zager
JS
, et al
Completion dissection or observation for sentinel-node metastasis in melanoma
.
N Engl J Med
2017
;
376
:
2211
22
.
53.
Harter
P
,
Sehouli
J
,
Lorusso
D
,
Reuss
A
,
Vergote
I
,
Marth
C
, et al
A randomized trial of lymphadenectomy in patients with advanced ovarian neoplasms
.
N Engl J Med
2019
;
380
:
822
32
.
54.
Pereira
ER
,
Kedrin
D
,
Seano
G
,
Gautier
O
,
Meijer
EFJ
,
Jones
D
, et al
Lymph node metastases can invade local blood vessels, exit the node, and colonize distant organs in mice
.
Science
2018
;
359
:
1403
7
.
55.
Brown
M
,
Assen
FP
,
Leithner
A
,
Abe
J
,
Schachner
H
,
Asfour
G
, et al
Lymph node blood vessels provide exit routes for metastatic tumor cell dissemination in mice
.
Science
2018
;
359
:
1408
11
.
56.
Jones
D
,
Pereira
ER
,
Padera
TP
. 
Growth and immune evasion of lymph node metastasis
.
Front Oncol
2018
;
8
:
36
.
57.
Lund
AW
,
Wagner
M
,
Fankhauser
M
,
Steinskog
ES
,
Broggi
MA
,
Spranger
S
, et al
Lymphatic vessels regulate immune microenvironments in human and murine melanoma
.
J Clin Invest
2016
;
126
:
3389
402
.
58.
Ubellacker
JM
,
Tasdogan
A
,
Ramesh
V
,
Shen
B
,
Mitchell
EC
,
Martin-Sandoval
MS
, et al
Lymph protects metastasizing melanoma cells from ferroptosis
.
Nature
2020
;
585
:
113
8
.
59.
Lengyel
E
. 
Ovarian cancer development and metastasis
.
Am J Pathol
2010
;
177
:
1053
64
.
60.
Lugassy
C
,
Kleinman
HK
,
Vermeulen
PB
,
Barnhill
RL
. 
Angiotropism, pericytic mimicry and extravascular migratory metastasis: an embryogenesis-derived program of tumor spread
.
Angiogenesis
2020
;
23
:
27
41
.
61.
Liebig
C
,
Ayala
G
,
Wilks
JA
,
Berger
DH
,
Albo
D
. 
Perineural invasion in cancer: a review of the literature
.
Cancer
2009
;
115
:
3379
91
.
62.
Kadota
K
,
Nitadori
JI
,
Sima
CS
,
Ujiie
H
,
Rizk
NP
,
Jones
DR
, et al
Tumor spread through air spaces is an important pattern of invasion and impacts the frequency and location of recurrences after limited resection for small stage I lung adenocarcinomas
.
J Thorac Oncol
2015
;
10
:
806
14
.
63.
Yagi
Y
,
Aly
RG
,
Tabata
K
,
Barlas
A
,
Rekhtman
N
,
Eguchi
T
, et al
Three-dimensional histologic, immunohistochemical, and multiplex immunofluorescence analyses of dynamic vessel co-option of spread through air spaces in lung adenocarcinoma
.
J Thorac Oncol
2020
;
15
:
589
600
.
64.
Pan
H
,
Gray
R
,
Braybrooke
J
,
Davies
C
,
Taylor
C
,
McGale
P
, et al
20-year risks of breast-cancer recurrence after stopping endocrine therapy at 5 years
.
N Engl J Med
2017
;
377
:
1836
46
.
65.
Malladi
S
,
Macalinao
DG
,
Jin
X
,
He
L
,
Basnet
H
,
Zou
Y
, et al
Metastatic latency and immune evasion through autocrine inhibition of WNT
.
Cell
2016
;
165
:
45
60
.
66.
Hadfield
G
. 
The dormant cancer cell
.
Br Med J
1954
;
2
:
607
10
.
67.
Phan
TG
,
Croucher
PI
. 
The dormant cancer cell life cycle
.
Nat Rev Cancer
2020
;
20
:
398
411
.
68.
Shen
S
,
Vagner
S
,
Robert
C
. 
Persistent cancer cells: the deadly survivors
.
Cell
2020
;
183
:
860
74
.
69.
Piskounova
E
,
Agathocleous
M
,
Murphy
MM
,
Hu
Z
,
Huddlestun
SE
,
Zhao
Z
, et al
Oxidative stress inhibits distant metastasis by human melanoma cells
.
Nature
2015
;
527
:
186
91
.
70.
Le Gal
K
,
Ibrahim
MX
,
Wiel
C
,
Sayin
VI
,
Akula
MK
,
Karlsson
C
, et al
Antioxidants can increase melanoma metastasis in mice
.
Sci Transl Med
2015
;
7
:
308re8
re8
.
71.
Chi
Y
,
Remsik
J
,
Kiseliovas
V
,
Derderian
C
,
Sener
U
,
Alghader
M
, et al
Cancer cells deploy lipocalin-2 to collect limiting iron in leptomeningeal metastasis
.
Science
2020
;
369
:
276
82
.
72.
Mohme
M
,
Riethdorf
S
,
Pantel
K
. 
Circulating and disseminated tumour cells–mechanisms of immune surveillance and escape
.
Nat Rev Clin Oncol
2017
;
14
:
155
67
.
73.
Garner
H
,
de Visser
KE
. 
Immune crosstalk in cancer progression and metastatic spread: a complex conversation
.
Nat Rev Immunol
2020
;
20
:
483
97
.
74.
Eyles
J
,
Puaux
AL
,
Wang
X
,
Toh
B
,
Prakash
C
,
Hong
M
, et al
Tumor cells disseminate early, but immunosurveillance limits metastatic outgrowth, in a mouse model of melanoma
.
J Clin Invest
2010
;
120
:
2030
9
.
75.
Bidwell
BN
,
Slaney
CY
,
Withana
NP
,
Forster
S
,
Cao
Y
,
Loi
S
, et al
Silencing of Irf7 pathways in breast cancer cells promotes bone metastasis through immune escape
.
Nat Med
2012
;
18
:
1224
31
.
76.
Van den Eynde
M
,
Mlecnik
B
,
Bindea
G
,
Fredriksen
T
,
Church
SE
,
Lafontaine
L
, et al
The link between the multiverse of immune microenvironments in metastases and the survival of colorectal cancer patients
.
Cancer Cell
2018
;
34
:
1012
26
.
77.
Angelova
M
,
Mlecnik
B
,
Vasaturo
A
,
Bindea
G
,
Fredriksen
T
,
Lafontaine
L
, et al
Evolution of metastases in space and time under immune selection
.
Cell
2018
;
175
:
751
65
.
78.
MacKie
RM
,
Reid
R
,
Junor
B
. 
Fatal melanoma transferred in a donated kidney 16 years after melanoma surgery
.
N Engl J Med
2003
;
348
:
567
8
.
79.
Xiao
D
,
Craig
JC
,
Chapman
JR
,
Dominguez-Gil
B
,
Tong
A
,
Wong
G
. 
Donor cancer transmission in kidney transplantation: a systematic review
.
Am J Transplant
2013
;
13
:
2645
52
.
80.
Eggermont
AMM
,
Blank
CU
,
Mandala
M
,
Long
GV
,
Atkinson
V
,
Dalle
S
, et al
Adjuvant pembrolizumab versus placebo in resected stage III melanoma
.
N Engl J Med
2018
;
378
:
1789
801
.
81.
Weber
J
,
Mandala
M
,
Del Vecchio
M
,
Gogas
HJ
,
Arance
AM
,
Cowey
CL
, et al
Adjuvant nivolumab versus ipilimumab in resected stage III or IV melanoma
.
N Engl J Med
2017
;
377
:
1824
35
.
82.
Eggermont
AM
,
Chiarion-Sileni
V
,
Grob
JJ
,
Dummer
R
,
Wolchok
JD
,
Schmidt
H
, et al
Prolonged survival in stage III melanoma with ipilimumab adjuvant therapy
.
N Engl J Med
2016
;
375
:
1845
55
.
83.
Zimmer
L
,
Livingstone
E
,
Hassel
JC
,
Fluck
M
,
Eigentler
T
,
Loquai
C
, et al
Adjuvant nivolumab plus ipilimumab or nivolumab monotherapy versus placebo in patients with resected stage IV melanoma with no evidence of disease (IMMUNED): a randomised, double-blind, placebo-controlled, phase 2 trial
.
Lancet
2020
;
395
:
1558
68
.
84.
Pommier
A
,
Anaparthy
N
,
Memos
N
,
Kelley
ZL
,
Gouronnec
A
,
Yan
R
, et al
Unresolved endoplasmic reticulum stress engenders immune-resistant, latent pancreatic cancer metastases
.
Science
2018
;
360
:
eaao4908
.
85.
Pantel
K
,
Schlimok
G
,
Kutter
D
,
Schaller
G
,
Genz
T
,
Wiebecke
B
, et al
Frequent down-regulation of major histocompatibility class I antigen expression on individual micrometastatic carcinoma cells
.
Cancer Res
1991
;
51
:
4712
5
.
86.
Koebel
CM
,
Vermi
W
,
Swann
JB
,
Zerafa
N
,
Rodig
SJ
,
Old
LJ
, et al
Adaptive immunity maintains occult cancer in an equilibrium state
.
Nature
2007
;
450
:
903
7
.
87.
Eyob
H
,
Ekiz
HA
,
Derose
YS
,
Waltz
SE
,
Williams
MA
,
Welm
AL
. 
Inhibition of ron kinase blocks conversion of micrometastases to overt metastases by boosting antitumor immunity
.
Cancer Discov
2013
;
3
:
751
60
.
88.
David
CJ
,
Massagué
J
. 
Contextual determinants of TGFbeta action in development, immunity and cancer
.
Nat Rev Mol Cell Biol
2018
;
19
:
419
35
.
89.
Batlle
E
,
Massagué
J
. 
Transforming growth factor-beta signaling in immunity and cancer
.
Immunity
2019
;
50
:
924
40
.
90.
Naumov
GN
,
Akslen
LA
,
Folkman
J
. 
Role of angiogenesis in human tumor dormancy: animal models of the angiogenic switch
.
Cell Cycle
2006
;
5
:
1779
87
.
91.
Ye
W
. 
The complexity of translating anti-angiogenesis therapy from basic science to the clinic
.
Dev Cell
2016
;
37
:
114
25
.
92.
Gao
H
,
Chakraborty
G
,
Lee-Lim
AP
,
Mo
Q
,
Decker
M
,
Vonica
A
, et al
The BMP inhibitor Coco reactivates breast cancer cells at lung metastatic sites
.
Cell
2012
;
150
:
764
79
.
93.
Vera-Ramirez
L
,
Vodnala
SK
,
Nini
R
,
Hunter
KW
,
Green
JE
. 
Autophagy promotes the survival of dormant breast cancer cells and metastatic tumour recurrence
.
Nat Commun
2018
;
9
:
1944
.
94.
La Belle Flynn
A
,
Calhoun
BC
,
Sharma
A
,
Chang
JC
,
Almasan
A
,
Schiemann
WP
. 
Autophagy inhibition elicits emergence from metastatic dormancy by inducing and stabilizing Pfkfb3 expression
.
Nat Commun
2019
;
10
:
3668
.
95.
Shibue
T
,
Brooks
MW
,
Weinberg
RA
. 
An integrin-linked machinery of cytoskeletal regulation that enables experimental tumor initiation and metastatic colonization
.
Cancer Cell
2013
;
24
:
481
98
.
96.
De Cock
JM
,
Shibue
T
,
Dongre
A
,
Keckesova
Z
,
Reinhardt
F
,
Weinberg
RA
. 
Inflammation triggers Zeb1-dependent escape from tumor latency
.
Cancer Res
2016
;
76
:
6778
84
.
97.
Albrengues
J
,
Shields
MA
,
Ng
D
,
Park
CG
,
Ambrico
A
,
Poindexter
ME
, et al
Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice
.
Science
2018
;
361
:
eaao4227
.
98.
Brinkmann
V
,
Reichard
U
,
Goosmann
C
,
Fauler
B
,
Uhlemann
Y
,
Weiss
DS
, et al
Neutrophil extracellular traps kill bacteria
.
Science
2004
;
303
:
1532
5
.
99.
Branzk
N
,
Papayannopoulos
V
. 
Molecular mechanisms regulating NETosis in infection and disease
.
Semin Immunopathol
2013
;
35
:
513
30
.
100.
Park
J
,
Wysocki
RW
,
Amoozgar
Z
,
Maiorino
L
,
Fein
MR
,
Jorns
J
, et al
Cancer cells induce metastasis-supporting neutrophil extracellular DNA traps
.
Sci Transl Med
2016
;
8
:
361ra138
.
101.
Obenauf
AC
,
Massagué
J
. 
Surviving at a distance: organ-specific metastasis
.
Trends Cancer
2015
;
1
:
76
91
.
102.
Gao
Y
,
Bado
I
,
Wang
H
,
Zhang
W
,
Rosen
JM
,
Zhang
XH
. 
Metastasis organotropism: redefining the congenial soil
.
Dev Cell
2019
;
49
:
375
91
.
103.
Esposito
M
,
Guise
T
,
Kang
Y
. 
The biology of bone metastasis
.
Cold Spring Harb Perspect Med
2018
;
8
:
a031252
.
104.
Quail
DF
,
Joyce
JA
. 
The microenvironmental landscape of brain tumors
.
Cancer Cell
2017
;
31
:
326
41
.
105.
Altorki
NK
,
Markowitz
GJ
,
Gao
D
,
Port
JL
,
Saxena
A
,
Stiles
B
, et al
The lung microenvironment: an important regulator of tumour growth and metastasis
.
Nat Rev Cancer
2019
;
19
:
9
31
.
106.
Brodt
P
. 
Role of the microenvironment in liver metastasis: from pre- to prometastatic niches
.
Clin Cancer Res
2016
;
22
:
5971
82
.
107.
Ruoslahti
E
. 
Vascular zip codes in angiogenesis and metastasis
.
Biochem Soc Trans
2004
;
32
:
397
402
.
108.
Minn
AJ
,
Kang
Y
,
Serganova
I
,
Gupta
GP
,
Giri
DD
,
Doubrovin
M
, et al
Distinct organ-specific metastatic potential of individual breast cancer cells and primary tumors
.
J Clin Invest
2005
;
115
:
44
55
.
109.
Chambers
AF
,
Groom
AC
,
MacDonald
IC
. 
Dissemination and growth of cancer cells in metastatic sites
.
Nat Rev Cancer
2002
;
2
:
563
72
.
110.
Iacobuzio-Donahue
CA
,
Michael
C
,
Baez
P
,
Kappagantula
R
,
Hooper
JE
,
Hollman
TJ
. 
Cancer biology as revealed by the research autopsy
.
Nat Rev Cancer
2019
;
19
:
686
97
.
111.
Sevenich
L
,
Bowman
RL
,
Mason
SD
,
Quail
DF
,
Rapaport
F
,
Elie
BT
, et al
Analysis of tumour- and stroma-supplied proteolytic networks reveals a brain-metastasis-promoting role for cathepsin S
.
Nat Cell Biol
2014
;
16
:
876
88
.
112.
Urosevic
J
,
Garcia-Albeniz
X
,
Planet
E
,
Real
S
,
Cespedes
MV
,
Guiu
M
, et al
Colon cancer cells colonize the lung from established liver metastases through p38 MAPK signalling and PTHLH
.
Nat Cell Biol
2014
;
16
:
685
94
.
113.
Romero
I
,
Garrido
F
,
Garcia-Lora
AM
. 
Metastases in immune-mediated dormancy: a new opportunity for targeting cancer
.
Cancer Res
2014
;
74
:
6750
7
.
114.
Raulet
DH
,
Marcus
A
,
Coscoy
L
. 
Dysregulated cellular functions and cell stress pathways provide critical cues for activating and targeting natural killer cells to transformed and infected cells
.
Immunol Rev
2017
;
280
:
93
101
.
115.
Kwon
J
,
Bakhoum
SF
. 
The cytosolic DNA-sensing cGAS-STING pathway in cancer
.
Cancer Discov
2020
;
10
:
26
39
.
116.
Nabet
BY
,
Qiu
Y
,
Shabason
JE
,
Wu
TJ
,
Yoon
T
,
Kim
BC
, et al
Exosome RNA unshielding couples stromal activation to pattern recognition receptor signaling in cancer
.
Cell
2017
;
170
:
352
66
.
117.
Davis
RT
,
Blake
K
,
Ma
D
,
Gabra
MBI
,
Hernandez
GA
,
Phung
AT
, et al
Transcriptional diversity and bioenergetic shift in human breast cancer metastasis revealed by single-cell RNA sequencing
.
Nat Cell Biol
2020
;
22
:
310
20
.
118.
Basnet
H
,
Tian
L
,
Ganesh
K
,
Huang
YH
,
Macalinao
DG
,
Brogi
E
, et al
Flura-seq identifies organ-specific metabolic adaptations during early metastatic colonization
.
Elife
2019
;
8
:
e43627
.
119.
Dupuy
F
,
Tabaries
S
,
Andrzejewski
S
,
Dong
Z
,
Blagih
J
,
Annis
MG
, et al
PDK1-dependent metabolic reprogramming dictates metastatic potential in breast cancer
.
Cell Metab
2015
;
22
:
577
89
.
120.
Nguyen
A
,
Loo
JM
,
Mital
R
,
Weinberg
EM
,
Man
FY
,
Zeng
Z
, et al
PKLR promotes colorectal cancer liver colonization through induction of glutathione synthesis
.
J Clin Invest
2016
;
126
:
681
94
.
121.
Knott
SRV
,
Wagenblast
E
,
Khan
S
,
Kim
SY
,
Soto
M
,
Wagner
M
, et al
Asparagine bioavailability governs metastasis in a model of breast cancer
.
Nature
2018
;
554
:
378
81
.
122.
Chen
J
,
Lee
HJ
,
Wu
X
,
Huo
L
,
Kim
SJ
,
Xu
L
, et al
Gain of glucose-independent growth upon metastasis of breast cancer cells to the brain
.
Cancer Res
2015
;
75
:
554
65
.
123.
Yamaguchi
N
,
Weinberg
EM
,
Nguyen
A
,
Liberti
MV
,
Goodarzi
H
,
Janjigian
YY
, et al
PCK1 and DHODH drive colorectal cancer liver metastatic colonization and hypoxic growth by promoting nucleotide synthesis
.
Elife
2019
;
8
:
e52135
124.
Loo
JM
,
Scherl
A
,
Nguyen
A
,
Man
FY
,
Weinberg
E
,
Zeng
Z
, et al
Extracellular metabolic energetics can promote cancer progression
.
Cell
2015
;
160
:
393
406
.
125.
Mashimo
T
,
Pichumani
K
,
Vemireddy
V
,
Hatanpaa
KJ
,
Singh
DK
,
Sirasanagandla
S
, et al
Acetate is a bioenergetic substrate for human glioblastoma and brain metastases
.
Cell
2014
;
159
:
1603
14
.
126.
Lehuede
C
,
Dupuy
F
,
Rabinovitch
R
,
Jones
RG
,
Siegel
PM
. 
Metabolic plasticity as a determinant of tumor growth and metastasis
.
Cancer Res
2016
;
76
:
5201
8
.
127.
Pascual
G
,
Avgustinova
A
,
Mejetta
S
,
Martin
M
,
Castellanos
A
,
Attolini
CS
, et al
Targeting metastasis-initiating cells through the fatty acid receptor CD36
.
Nature
2017
;
541
:
41
5
.
128.
Jin
X
,
Demere
Z
,
Nair
K
,
Ali
A
,
Ferraro
GB
,
Natoli
T
, et al
A metastasis map of human cancer cell lines
.
Nature
2020
;
588
:
331
6
.
129.
Wculek
SK
,
Malanchi
I
. 
Neutrophils support lung colonization of metastasis-initiating breast cancer cells
.
Nature
2015
;
528
:
413
7
.
130.
Oskarsson
T
,
Acharyya
S
,
Zhang
XH
,
Vanharanta
S
,
Tavazoie
SF
,
Morris
PG
, et al
Breast cancer cells produce tenascin C as a metastatic niche component to colonize the lungs
.
Nat Med
2011
;
17
:
867
74
.
131.
Shiozawa
Y
,
Pedersen
EA
,
Havens
AM
,
Jung
Y
,
Mishra
A
,
Joseph
J
, et al
Human prostate cancer metastases target the hematopoietic stem cell niche to establish footholds in mouse bone marrow
.
J Clin Invest
2011
;
121
:
1298
312
.
132.
Wang
H
,
Tian
L
,
Liu
J
,
Goldstein
A
,
Bado
I
,
Zhang
W
, et al
The osteogenic niche is a calcium reservoir of bone micrometastases and confers unexpected therapeutic vulnerability
.
Cancer Cell
2018
;
34
:
823
39
.
133.
Northey
JJ
,
Przybyla
L
,
Weaver
VM
. 
Tissue force programs cell fate and tumor aggression
.
Cancer Discov
2017
;
7
:
1224
37
.
134.
Tomasek
JJ
,
Gabbiani
G
,
Hinz
B
,
Chaponnier
C
,
Brown
RA
. 
Myofibroblasts and mechano-regulation of connective tissue remodelling
.
Nat Rev Mol Cell Biol
2002
;
3
:
349
63
.
135.
Laklai
H
,
Miroshnikova
YA
,
Pickup
MW
,
Collisson
EA
,
Kim
GE
,
Barrett
AS
, et al
Genotype tunes pancreatic ductal adenocarcinoma tissue tension to induce matricellular fibrosis and tumor progression
.
Nat Med
2016
;
22
:
497
505
.
136.
Acerbi
I
,
Cassereau
L
,
Dean
I
,
Shi
Q
,
Au
A
,
Park
C
, et al
Human breast cancer invasion and aggression correlates with ECM stiffening and immune cell infiltration
.
Integr Biol
2015
;
7
:
1120
34
.
137.
Maller
O
,
Drain
AP
,
Barrett
AS
,
Borgquist
S
,
Ruffell
B
,
Zakharevich
I
, et al
Tumour-associated macrophages drive stromal cell-dependent collagen crosslinking and stiffening to promote breast cancer aggression
.
Nat Mater
2020
.
138.
Yin
JJ
,
Selander
K
,
Chirgwin
JM
,
Dallas
M
,
Grubbs
BG
,
Wieser
R
, et al
TGF-beta signaling blockade inhibits PTHrP secretion by breast cancer cells and bone metastases development
.
J Clin Invest
1999
;
103
:
197
206
.
139.
Zheng
H
,
Bae
Y
,
Kasimir-Bauer
S
,
Tang
R
,
Chen
J
,
Ren
G
, et al
Therapeutic antibody targeting tumor- and osteoblastic niche-derived jagged1 sensitizes bone metastasis to chemotherapy
.
Cancer Cell
2017
;
32
:
731
47
.
140.
Zhang
J
,
Dai
J
,
Qi
Y
,
Lin
DL
,
Smith
P
,
Strayhorn
C
, et al
Osteoprotegerin inhibits prostate cancer-induced osteoclastogenesis and prevents prostate tumor growth in the bone
.
J Clin Invest
2001
;
107
:
1235
44
.
141.
Chen
Q
,
Boire
A
,
Jin
X
,
Valiente
M
,
Er
EE
,
Lopez-Soto
A
, et al
Carcinoma-astrocyte gap junctions promote brain metastasis by cGAMP transfer
.
Nature
2016
;
533
:
493
8
.
142.
Priego
N
,
Zhu
L
,
Monteiro
C
,
Mulders
M
,
Wasilewski
D
,
Bindeman
W
, et al
STAT3 labels a subpopulation of reactive astrocytes required for brain metastasis
.
Nat Med
2018
;
24
:
1024
35
.
143.
Guldner
IH
,
Wang
Q
,
Yang
L
,
Golomb
SM
,
Zhao
Z
,
Lopez
JA
, et al
CNS-native myeloid cells drive immune suppression in the brain metastatic niche through Cxcl10
.
Cell
2020
;
183
:
1234
48
.
144.
Zeng
Q
,
Michael
IP
,
Zhang
P
,
Saghafinia
S
,
Knott
G
,
Jiao
W
, et al
Synaptic proximity enables NMDAR signalling to promote brain metastasis
.
Nature
2019
;
573
:
526
31
.
145.
Chen
Q
,
Zhang
XH
,
Massagué
J
. 
Macrophage binding to receptor VCAM-1 transmits survival signals in breast cancer cells that invade the lungs
.
Cancer Cell
2011
;
20
:
538
49
.
146.
Acharyya
S
,
Oskarsson
T
,
Vanharanta
S
,
Malladi
S
,
Kim
J
,
Morris
PG
, et al
A CXCL1 paracrine network links cancer chemoresistance and metastasis
.
Cell
2012
;
150
:
165
78
.
147.
Teijeira
A
,
Garasa
S
,
Gato
M
,
Alfaro
C
,
Migueliz
I
,
Cirella
A
, et al
CXCR1 and CXCR2 chemokine receptor agonists produced by tumors induce neutrophil extracellular traps that interfere with immune cytotoxicity
.
Immunity
2020
;
52
:
856
71
.
148.
Malanchi
I
,
Santamaria-Martinez
A
,
Susanto
E
,
Peng
H
,
Lehr
HA
,
Delaloye
JF
, et al
Interactions between cancer stem cells and their niche govern metastatic colonization
.
Nature
2011
;
481
:
85
9
.
149.
Ombrato
L
,
Nolan
E
,
Kurelac
I
,
Mavousian
A
,
Bridgeman
VL
,
Heinze
I
, et al
Metastatic-niche labelling reveals parenchymal cells with stem features
.
Nature
2019
;
572
:
603
8
.
150.
Peinado
H
,
Zhang
H
,
Matei
IR
,
Costa-Silva
B
,
Hoshino
A
,
Rodrigues
G
, et al
Pre-metastatic niches: organ-specific homes for metastases
.
Nat Rev Cancer
2017
;
17
:
302
17
.
151.
Pastushenko
I
,
Brisebarre
A
,
Sifrim
A
,
Fioramonti
M
,
Revenco
T
,
Boumahdi
S
, et al
Identification of the tumour transition states occurring during EMT
.
Nature
2018
;
556
:
463
8
.
152.
Su
J
,
Morgani
SM
,
David
CJ
,
Wang
Q
,
Er
EE
,
Huang
YH
, et al
TGF-beta orchestrates fibrogenic and developmental EMTs via the RAS effector RREB1
.
Nature
2020
;
577
:
566
71
.
153.
Shibue
T
,
Weinberg
RA
. 
EMT, CSCs, and drug resistance: the mechanistic link and clinical implications
.
Nat Rev Clin Oncol
2017
;
14
:
611
29
.
154.
Tsai
JH
,
Donaher
JL
,
Murphy
DA
,
Chau
S
,
Yang
J
. 
Spatiotemporal regulation of epithelial-mesenchymal transition is essential for squamous cell carcinoma metastasis
.
Cancer Cell
2012
;
22
:
725
36
.
155.
Ocana
OH
,
Corcoles
R
,
Fabra
A
,
Moreno-Bueno
G
,
Acloque
H
,
Vega
S
, et al
Metastatic colonization requires the repression of the epithelial-mesenchymal transition inducer Prrx1
.
Cancer Cell
2012
;
22
:
709
24
.
156.
Pastushenko
I
,
Blanpain
C
. 
EMT transition states during tumor progression and metastasis
.
Trends Cell Biol
2019
;
29
:
212
26
.
157.
Pastushenko
I
,
Mauri
F
,
Song
Y
,
de Cock
F
,
Meeusen
B
,
Swedlund
B
, et al
Fat1 deletion promotes hybrid EMT state, tumour stemness and metastasis
.
Nature
2021
;
589
:
448
55
.
158.
Hanahan
D
,
Weinberg
RA
. 
Hallmarks of cancer: the next generation
.
Cell
2011
;
144
:
646
74
.
159.
Gupta
PB
,
Pastushenko
I
,
Skibinski
A
,
Blanpain
C
,
Kuperwasser
C
. 
Phenotypic plasticity: driver of cancer initiation, progression, and therapy resistance
.
Cell Stem Cell
2019
;
24
:
65
78
.
160.
Yuan
S
,
Norgard
RJ
,
Stanger
BZ
. 
Cellular plasticity in cancer
.
Cancer Discov
2019
;
9
:
837
51
.
161.
Quintanal-Villalonga
A
,
Chan
JM
,
Yu
HA
,
Pe'er
D
,
Sawyers
CL
,
Sen
T
, et al
Lineage plasticity in cancer: a shared pathway of therapeutic resistance
.
Nat Rev Clin Oncol
2020
;
17
:
360
71
.
162.
Priestley
P
,
Baber
J
,
Lolkema
MP
,
Steeghs
N
,
de Bruijn
E
,
Shale
C
, et al
Pan-cancer whole-genome analyses of metastatic solid tumours
.
Nature
2019
;
575
:
210
6
.
163.
Biswas
D
,
Birkbak
NJ
,
Rosenthal
R
,
Hiley
CT
,
Lim
EL
,
Papp
K
, et al
A clonal expression biomarker associates with lung cancer mortality
.
Nat Med
2019
;
25
:
1540
8
.
164.
Hunter
KW
,
Amin
R
,
Deasy
S
,
Ha
NH
,
Wakefield
L
. 
Genetic insights into the morass of metastatic heterogeneity
.
Nat Rev Cancer
2018
;
18
:
211
23
.
165.
Reiter
JG
,
Baretti
M
,
Gerold
JM
,
Makohon-Moore
AP
,
Daud
A
,
Iacobuzio-Donahue
CA
, et al
An analysis of genetic heterogeneity in untreated cancers
.
Nat Rev Cancer
2019
;
19
:
639
50
.
166.
Turajlic
S
,
Swanton
C
. 
Metastasis as an evolutionary process
.
Science
2016
;
352
:
169
75
.
167.
Makohon-Moore
AP
,
Zhang
M
,
Reiter
JG
,
Bozic
I
,
Allen
B
,
Kundu
D
, et al
Limited heterogeneity of known driver gene mutations among the metastases of individual patients with pancreatic cancer
.
Nat Genet
2017
;
49
:
358
66
.
169.
Reiter
JG
,
Makohon-Moore
AP
,
Gerold
JM
,
Heyde
A
,
Attiyeh
MA
,
Kohutek
ZA
, et al
Minimal functional driver gene heterogeneity among untreated metastases
.
Science
2018
;
361
:
1033
7
.
169.
Notta
F
,
Chan-Seng-Yue
M
,
Lemire
M
,
Li
Y
,
Wilson
GW
,
Connor
AA
, et al
A renewed model of pancreatic cancer evolution based on genomic rearrangement patterns
.
Nature
2016
;
538
:
378
82
.
170.
Yates
LR
,
Knappskog
S
,
Wedge
D
,
Farmery
JHR
,
Gonzalez
S
,
Martincorena
I
, et al
Genomic evolution of breast cancer metastasis and relapse
.
Cancer Cell
2017
;
32
:
169
84
.
171.
Hu
Z
,
Li
Z
,
Ma
Z
,
Curtis
C
. 
Multi-cancer analysis of clonality and the timing of systemic spread in paired primary tumors and metastases
.
Nat Genet
2020
;
52
:
701
8
.
172.
Yachida
S
,
Jones
S
,
Bozic
I
,
Antal
T
,
Leary
R
,
Fu
B
, et al
Distant metastasis occurs late during the genetic evolution of pancreatic cancer
.
Nature
2010
;
467
:
1114
7
.
173.
Razavi
P
,
Chang
MT
,
Xu
G
,
Bandlamudi
C
,
Ross
DS
,
Vasan
N
, et al
The genomic landscape of endocrine-resistant advanced breast cancers
.
Cancer Cell
2018
;
34
:
427
38
.
174.
Sanchez-Vega
F
,
Mina
M
,
Armenia
J
,
Chatila
WK
,
Luna
A
,
La
KC
, et al
Oncogenic signaling pathways in the cancer genome atlas
.
Cell
2018
;
173
:
321
37
.
175.
Watkins
TBK
,
Lim
EL
,
Petkovic
M
,
Elizalde
S
,
Birkbak
NJ
,
Wilson
GA
, et al
Pervasive chromosomal instability and karyotype order in tumour evolution
.
Nature
2020
;
587
:
126
32
.
176.
Jamal-Hanjani
M
,
Wilson
GA
,
McGranahan
N
,
Birkbak
NJ
,
Watkins
TBK
,
Veeriah
S
, et al
Tracking the evolution of non-small-cell lung cancer
.
N Engl J Med
2017
;
376
:
2109
21
.
177.
Turajlic
S
,
Sottoriva
A
,
Graham
T
,
Swanton
C
. 
Resolving genetic heterogeneity in cancer
.
Nat Rev Genet
2019
;
20
:
404
16
.
178.
Hu
Z
,
Curtis
C
. 
Looking backward in time to define the chronology of metastasis
.
Nat Commun
2020
;
11
:
3213
.
179.
Klein
CA
. 
Parallel progression of primary tumours and metastases
.
Nat Rev Cancer
2009
;
9
:
302
12
.
180.
Hosseini
H
,
Obradovic
MMS
,
Hoffmann
M
,
Harper
KL
,
Sosa
MS
,
Werner-Klein
M
, et al
Early dissemination seeds metastasis in breast cancer
.
Nature
2016
;
540
:
552
8
.
181.
Harper
KL
,
Sosa
MS
,
Entenberg
D
,
Hosseini
H
,
Cheung
JF
,
Nobre
R
, et al
Mechanism of early dissemination and metastasis in Her2(+) mammary cancer
.
Nature
2016
;
540
:
588
92
.
182.
Turajlic
S
,
Xu
H
,
Litchfield
K
,
Rowan
A
,
Horswell
S
,
Chambers
T
, et al
Deterministic evolutionary trajectories influence primary tumor growth: TRACERx renal
.
Cell
2018
;
173
:
595
610
.
183.
Sottoriva
A
,
Kang
H
,
Ma
Z
,
Graham
TA
,
Salomon
MP
,
Zhao
J
, et al
A Big Bang model of human colorectal tumor growth
.
Nat Genet
2015
;
47
:
209
16
.
184.
Turajlic
S
,
Xu
H
,
Litchfield
K
,
Rowan
A
,
Chambers
T
,
Lopez
JI
, et al
Tracking cancer evolution reveals constrained routes to metastases: TRACERx renal
.
Cell
2018
;
173
:
581
94
.
185.
Noorani
A
,
Li
X
,
Goddard
M
,
Crawte
J
,
Alexandrov
LB
,
Secrier
M
, et al
Genomic evidence supports a clonal diaspora model for metastases of esophageal adenocarcinoma
.
Nat Genet
2020
;
52
:
74
83
.
186.
Sanborn
JZ
,
Chung
J
,
Purdom
E
,
Wang
NJ
,
Kakavand
H
,
Wilmott
JS
, et al
Phylogenetic analyses of melanoma reveal complex patterns of metastatic dissemination
.
Proc Natl Acad Sci U S A
2015
;
112
:
10995
1000
.
187.
Maddipati
R
,
Stanger
BZ
. 
Pancreatic cancer metastases harbor evidence of polyclonality
.
Cancer Discov
2015
;
5
:
1086
97
.
188.
Kim
MY
,
Oskarsson
T
,
Acharyya
S
,
Nguyen
DX
,
Zhang
XH
,
Norton
L
, et al
Tumor self-seeding by circulating cancer cells
.
Cell
2009
;
139
:
1315
26
.
189.
Comen
E
,
Norton
L
,
Massagué
J
. 
Clinical implications of cancer self-seeding
.
Nat Rev Clin Oncol
2011
;
8
:
369
77
.
190.
Weichselbaum
RR
,
Hellman
S
. 
Oligometastases revisited
.
Nat Rev Clin Oncol
2011
;
8
:
378
82
.
191.
Demicheli
R
,
Abbattista
A
,
Miceli
R
,
Valagussa
P
,
Bonadonna
G
. 
Time distribution of the recurrence risk for breast cancer patients undergoing mastectomy: further support about the concept of tumor dormancy
.
Breast Cancer Res Treat
1996
;
41
:
177
85
.
192.
Lapidot
T
,
Sirard
C
,
Vormoor
J
,
Murdoch
B
,
Hoang
T
,
Caceres-Cortes
J
, et al
A cell initiating human acute myeloid leukaemia after transplantation into SCID mice
.
Nature
1994
;
367
:
645
8
.
193.
Bonnet
D
,
Dick
JE
. 
Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell
.
Nat Med
1997
;
3
:
730
7
.
194.
Shimokawa
M
,
Ohta
Y
,
Nishikori
S
,
Matano
M
,
Takano
A
,
Fujii
M
, et al
Visualization and targeting of LGR5(+) human colon cancer stem cells
.
Nature
2017
;
545
:
187
92
.
195.
Barker
N
,
Ridgway
RA
,
van Es
JH
,
van de Wetering
M
,
Begthel
H
,
van den Born
M
, et al
Crypt stem cells as the cells-of-origin of intestinal cancer
.
Nature
2009
;
457
:
608
11
.
196.
Visvader
JE
. 
Cells of origin in cancer
.
Nature
2011
;
469
:
314
22
.
197.
Flavahan
WA
,
Gaskell
E
,
Bernstein
BE
. 
Epigenetic plasticity and the hallmarks of cancer
.
Science
2017
;
357
:
eaal2380
.
198.
Denny
SK
,
Yang
D
,
Chuang
CH
,
Brady
JJ
,
Lim
JS
,
Gruner
BM
, et al
Nfib promotes metastasis through a widespread increase in chromatin accessibility
.
Cell
2016
;
166
:
328
42
.
199.
Marjanovic
ND
,
Hofree
M
,
Chan
JE
,
Canner
D
,
Wu
K
,
Trakala
M
, et al
Emergence of a high-plasticity cell state during lung cancer evolution
.
Cancer Cell
2020
;
38
:
229
46
.
200.
LaFave
LM
,
Kartha
VK
,
Ma
S
,
Meli
K
,
Del Priore
I
,
Lareau
C
, et al
Epigenomic state transitions characterize tumor progression in mouse lung adenocarcinoma
.
Cancer Cell
2020
;
38
:
212
28
.
201.
Roe
JS
,
Hwang
CI
,
Somerville
TDD
,
Milazzo
JP
,
Lee
EJ
,
Da Silva
B
, et al
Enhancer reprogramming promotes pancreatic cancer metastasis
.
Cell
2017
;
170
:
875
88
.
202.
Vanharanta
S
,
Shu
W
,
Brenet
F
,
Hakimi
AA
,
Heguy
A
,
Viale
A
, et al
Epigenetic expansion of VHL-HIF signal output drives multiorgan metastasis in renal cancer
.
Nat Med
2013
;
19
:
50
6
.
203.
Gomes
AP
,
Ilter
D
,
Low
V
,
Rosenzweig
A
,
Shen
ZJ
,
Schild
T
, et al
Dynamic incorporation of histone H3 variants into chromatin is essential for acquisition of aggressive traits and metastatic colonization
.
Cancer Cell
2019
;
36
:
402
17
.
204.
Yuan
S
,
Natesan
R
,
Sanchez-Rivera
FJ
,
Li
J
,
Bhanu
NV
,
Yamazoe
T
, et al
Global regulation of the histone mark H3K36me2 underlies epithelial plasticity and metastatic progression
.
Cancer Discov
2020
;
10
:
854
71
.
205.
Dekoninck
S
,
Blanpain
C
. 
Stem cell dynamics, migration and plasticity during wound healing
.
Nat Cell Biol
2019
;
21
:
18
24
.
206.
de Sousa
EMF
,
de Sauvage
FJ
. 
Cellular plasticity in intestinal homeostasis and disease
.
Cell Stem Cell
2019
;
24
:
54
64
.
207.
Rompolas
P
,
Deschene
ER
,
Zito
G
,
Gonzalez
DG
,
Saotome
I
,
Haberman
AM
, et al
Live imaging of stem cell and progeny behaviour in physiological hair-follicle regeneration
.
Nature
2012
;
487
:
496
9
.
208.
Tata
PR
,
Mou
H
,
Pardo-Saganta
A
,
Zhao
R
,
Prabhu
M
,
Law
BM
, et al
Dedifferentiation of committed epithelial cells into stem cells in vivo
.
Nature
2013
;
503
:
218
23
.
209.
Tian
H
,
Biehs
B
,
Warming
S
,
Leong
KG
,
Rangell
L
,
Klein
OD
, et al
A reserve stem cell population in small intestine renders Lgr5-positive cells dispensable
.
Nature
2011
;
478
:
255
9
.
210.
Barker
N
,
van Es
JH
,
Kuipers
J
,
Kujala
P
,
van den Born
M
,
Cozijnsen
M
, et al
Identification of stem cells in small intestine and colon by marker gene Lgr5
.
Nature
2007
;
449
:
1003
7
.
211.
Ayyaz
A
,
Kumar
S
,
Sangiorgi
B
,
Ghoshal
B
,
Gosio
J
,
Ouladan
S
, et al
Single-cell transcriptomes of the regenerating intestine reveal a revival stem cell
.
Nature
2019
;
569
:
121
5
.
212.
Sato
T
,
Vries
RG
,
Snippert
HJ
,
van de Wetering
M
,
Barker
N
,
Stange
DE
, et al
Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche
.
Nature
2009
;
459
:
262
5
.
213.
Serra
D
,
Mayr
U
,
Boni
A
,
Lukonin
I
,
Rempfler
M
,
Challet Meylan
L
, et al
Self-organization and symmetry breaking in intestinal organoid development
.
Nature
2019
;
569
:
66
72
.
214.
Fumagalli
A
,
Oost
KC
,
Kester
L
,
Morgner
J
,
Bornes
L
,
Bruens
L
, et al
Plasticity of Lgr5-negative cancer cells drives metastasis in colorectal cancer
.
Cell Stem Cell
2020
;
26
:
569
78
.
215.
Han
T
,
Goswami
S
,
Hu
Y
,
Tang
F
,
Zafra
MP
,
Murphy
C
, et al
Lineage reversion drives WNT independence in intestinal cancer
.
Cancer Discov
2020
;
10
:
1590
609
.
216.
Morral
C
,
Stanisavljevic
J
,
Hernando-Momblona
X
,
Mereu
E
,
Alvarez-Varela
A
,
Cortina
C
, et al
Zonation of ribosomal DNA transcription defines a stem cell hierarchy in colorectal cancer
.
Cell Stem Cell
2020
;
26
:
845
61
.
217.
Altevogt
P
,
Doberstein
K
,
Fogel
M
. 
L1CAM in human cancer
.
Int J Cancer
2016
;
138
:
1565
76
.
218.
Miyoshi
H
,
Ajima
R
,
Luo
CT
,
Yamaguchi
TP
,
Stappenbeck
TS
. 
Wnt5a potentiates TGF-beta signaling to promote colonic crypt regeneration after tissue injury
.
Science
2012
;
338
:
108
13
.
219.
Wang
Y
,
Chiang
IL
,
Ohara
TE
,
Fujii
S
,
Cheng
J
,
Muegge
BD
, et al
Long-term culture captures injury-repair cycles of colonic stem cells
.
Cell
2019
;
179
:
1144
59
.
220.
Laughney
AM
,
Hu
J
,
Campbell
NR
,
Bakhoum
SF
,
Setty
M
,
Lavallee
VP
, et al
Regenerative lineages and immune-mediated pruning in lung cancer metastasis
.
Nat Med
2020
;
26
:
259
69
.
221.
Padua
D
,
Zhang
XH
,
Wang
Q
,
Nadal
C
,
Gerald
WL
,
Gomis
RR
, et al
TGFbeta primes breast tumors for lung metastasis seeding through angiopoietin-like 4
.
Cell
2008
;
133
:
66
77
.
222.
Zhang
XH
,
Jin
X
,
Malladi
S
,
Zou
Y
,
Wen
YH
,
Brogi
E
, et al
Selection of bone metastasis seeds by mesenchymal signals in the primary tumor stroma
.
Cell
2013
;
154
:
1060
73
.
223.
Alečković
M
,
McAllister
SS
,
Polyak
K
. 
Metastasis as a systemic disease: molecular insights and clinical implications
.
Biochim Biophys Acta Rev Cancer
2019
;
1872
:
89
102
.
224.
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
.
225.
Hoshino
A
,
Kim
HS
,
Bojmar
L
,
Gyan
KE
,
Cioffi
M
,
Hernandez
J
, et al
Extracellular vesicle and particle biomarkers define multiple human cancers
.
Cell
2020
;
182
:
1044
61
.
226.
Kaczanowska
S
,
Kaplan
RN
. 
Mapping the switch that drives the pre-metastatic niche
.
Nature Cancer
2020
;
1
:
577
9
.
227.
Moreno-Smith
M
,
Lutgendorf
SK
,
Sood
AK
. 
Impact of stress on cancer metastasis
.
Future Oncol
2010
;
6
:
1863
81
.
228.
Obradovic
MMS
,
Hamelin
B
,
Manevski
N
,
Couto
JP
,
Sethi
A
,
Coissieux
MM
, et al
Glucocorticoids promote breast cancer metastasis
.
Nature
2019
;
567
:
540
4
.
229.
Flint
TR
,
Janowitz
T
,
Connell
CM
,
Roberts
EW
,
Denton
AE
,
Coll
AP
, et al
Tumor-induced IL-6 reprograms host metabolism to suppress anti-tumor immunity
.
Cell Metab
2016
;
24
:
672
84
.
230.
Webster
JI
,
Tonelli
L
,
Sternberg
EM
. 
Neuroendocrine regulation of immunity
.
Annu Rev Immunol
2002
;
20
:
125
63
.
231.
Perego
M
,
Tyurin
VA
,
Tyurina
YY
,
Yellets
J
,
Nacarelli
T
,
Lin
C
, et al
Reactivation of dormant tumor cells by modified lipids derived from stress-activated neutrophils
.
Sci Transl Med
2020
;
12
:
eabb5817
.
232.
Harper
EI
,
Sheedy
EF
,
Stack
MS
. 
With great age comes great metastatic ability: ovarian cancer and the appeal of the aging peritoneal microenvironment
.
Cancers (Basel)
2018
;
10
:
E230
.
233.
Amant
F
,
von Minckwitz
G
,
Han
SN
,
Bontenbal
M
,
Ring
AE
,
Giermek
J
, et al
Prognosis of women with primary breast cancer diagnosed during pregnancy: results from an international collaborative study
.
J Clin Oncol
2013
;
31
:
2532
9
.
234.
Pennock
ND
,
Martinson
HA
,
Guo
Q
,
Betts
CB
,
Jindal
S
,
Tsujikawa
T
, et al
Ibuprofen supports macrophage differentiation, T cell recruitment, and tumor suppression in a model of postpartum breast cancer
.
J Immunother Cancer
2018
;
6
:
98
.
235.
Taranova
AG
,
Maldonado
D
 3rd
,
Vachon
CM
,
Jacobsen
EA
,
Abdala-Valencia
H
,
McGarry
MP
, et al
Allergic pulmonary inflammation promotes the recruitment of circulating tumor cells to the lung
.
Cancer Res
2008
;
68
:
8582
9
.
236.
Roy
LD
,
Ghosh
S
,
Pathangey
LB
,
Tinder
TL
,
Gruber
HE
,
Mukherjee
P
. 
Collagen induced arthritis increases secondary metastasis in MMTV-PyV MT mouse model of mammary cancer
.
BMC Cancer
2011
;
11
:
365
.
237.
Krall
JA
,
Reinhardt
F
,
Mercury
OA
,
Pattabiraman
DR
,
Brooks
MW
,
Dougan
M
, et al
The systemic response to surgery triggers the outgrowth of distant immune-controlled tumors in mouse models of dormancy
.
Sci Transl Med
2018
;
10
:
eaan3464
.
238.
Demicheli
R
,
Retsky
MW
,
Hrushesky
WJ
,
Baum
M
. 
Tumor dormancy and surgery-driven interruption of dormancy in breast cancer: learning from failures
.
Nat Clin Pract Oncol
2007
;
4
:
699
710
.
239.
Benish
M
,
Bartal
I
,
Goldfarb
Y
,
Levi
B
,
Avraham
R
,
Raz
A
, et al
Perioperative use of beta-blockers and COX-2 inhibitors may improve immune competence and reduce the risk of tumor metastasis
.
Ann Surg Oncol
2008
;
15
:
2042
52
.
240.
Biswas
AK
,
Acharyya
S
. 
Understanding cachexia in the context of metastatic progression
.
Nat Rev Cancer
2020
;
20
:
274
84
.
241.
Faulkner
S
,
Jobling
P
,
March
B
,
Jiang
CC
,
Hondermarck
H
. 
Tumor neurobiology and the war of nerves in cancer
.
Cancer Discov
2019
;
9
:
702
10
.
242.
Kuol
N
,
Stojanovska
L
,
Apostolopoulos
V
,
Nurgali
K
. 
Role of the nervous system in cancer metastasis
.
J Exp Clin Cancer Res
2018
;
37
:
5
.
243.
Kumar
A
,
Brockes
JP
. 
Nerve dependence in tissue, organ, and appendage regeneration
.
Trends Neurosci
2012
;
35
:
691
9
.
244.
Magnon
C
,
Hall
SJ
,
Lin
J
,
Xue
X
,
Gerber
L
,
Freedland
SJ
, et al
Autonomic nerve development contributes to prostate cancer progression
.
Science
2013
;
341
:
1236361
.
245.
Schiller
M
,
Ben-Shaanan
TL
,
Rolls
A
. 
Neuronal regulation of immunity: why, how and where?
Nat Rev Immunol
2021
;
21
:
20
36
.
246.
Erin
N
,
Boyer
PJ
,
Bonneau
RH
,
Clawson
GA
,
Welch
DR
. 
Capsaicin-mediated denervation of sensory neurons promotes mammary tumor metastasis to lung and heart
.
Anticancer Res
2004
;
24
:
1003
9
.
247.
Cannioto
RA
,
Hutson
A
,
Dighe
S
,
McCann
W
,
McCann
SE
,
Zirpoli
GR
, et al
Physical activity before, during, and after chemotherapy for high-risk breast cancer: relationships with survival
.
J Natl Cancer Inst
2021
;
113
:
54
63
.
248.
Koelwyn
GJ
,
Quail
DF
,
Zhang
X
,
White
RM
,
Jones
LW
. 
Exercise-dependent regulation of the tumour microenvironment
.
Nat Rev Cancer
2017
;
17
:
620
32
.
249.
Chambers
AF
. 
Influence of diet on metastasis and tumor dormancy
.
Clin Exp Metastasis
2009
;
26
:
61
6
.
250.
Hopkins
BD
,
Pauli
C
,
Du
X
,
Wang
DG
,
Li
X
,
Wu
D
, et al
Suppression of insulin feedback enhances the efficacy of PI3K inhibitors
.
Nature
2018
;
560
:
499
503
.
251.
Klil-Drori
AJ
,
Azoulay
L
,
Pollak
MN
. 
Cancer, obesity, diabetes, and antidiabetic drugs: is the fog clearing?
Nat Rev Clin Oncol
2017
;
14
:
85
99
.
252.
Lien
EC
,
Vander Heiden
MG
. 
A framework for examining how diet impacts tumour metabolism
.
Nat Rev Cancer
2019
;
19
:
651
61
.
253.
Christ
A
,
Lauterbach
M
,
Latz
E
. 
Western diet and the immune system: an inflammatory connection
.
Immunity
2019
;
51
:
794
811
.
254.
Ewertz
M
,
Jensen
MB
,
Gunnarsdóttir
K
,
Højris
I
,
Jakobsen
EH
,
Nielsen
D
, et al
Effect of obesity on prognosis after early-stage breast cancer
.
J Clin Oncol
2011
;
29
:
25
31
.
255.
Rubio-Patiño
C
,
Bossowski
JP
,
De Donatis
GM
,
Mondragón
L
,
Villa
E
,
Aira
LE
, et al
Low-protein diet induces IRE1α-dependent anticancer immunosurveillance
.
Cell Metab
2018
;
27
:
828
42
.
256.
Schmid
D
,
Leitzmann
MF
. 
Association between physical activity and mortality among breast cancer and colorectal cancer survivors: a systematic review and meta-analysis
.
Ann Oncol
2014
;
25
:
1293
311
.
257.
Kanarek
N
,
Petrova
B
,
Sabatini
DM
. 
Dietary modifications for enhanced cancer therapy
.
Nature
2020
;
579
:
507
17
.
258.
Helmink
BA
,
Khan
MAW
,
Hermann
A
,
Gopalakrishnan
V
,
Wargo
JA
. 
The microbiome, cancer, and cancer therapy
.
Nat Med
2019
;
25
:
377
88
.
259.
Geller
LT
,
Barzily-Rokni
M
,
Danino
T
,
Jonas
OH
,
Shental
N
,
Nejman
D
, et al
Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine
.
Science
2017
;
357
:
1156
60
.
260.
Bullman
S
,
Pedamallu
CS
,
Sicinska
E
,
Clancy
TE
,
Zhang
X
,
Cai
D
, et al
Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer
.
Science
2017
;
358
:
1443
8
.
261.
Nejman
D
,
Livyatan
I
,
Fuks
G
,
Gavert
N
,
Zwang
Y
,
Geller
LT
, et al
The human tumor microbiome is composed of tumor type-specific intracellular bacteria
.
Science
2020
;
368
:
973
80
.
262.
Chen
YE
,
Fischbach
MA
,
Belkaid
Y
. 
Skin microbiota-host interactions
.
Nature
2018
;
553
:
427
36
.
263.
Fischbach
MA
. 
Microbiome: focus on causation and mechanism
.
Cell
2018
;
174
:
785
90
.
264.
Cotillard
A
,
Kennedy
SP
,
Kong
LC
,
Prifti
E
,
Pons
N
,
Le Chatelier
E
, et al
Dietary intervention impact on gut microbial gene richness
.
Nature
2013
;
500
:
585
8
.
265.
David
LA
,
Maurice
CF
,
Carmody
RN
,
Gootenberg
DB
,
Button
JE
,
Wolfe
BE
, et al
Diet rapidly and reproducibly alters the human gut microbiome
.
Nature
2014
;
505
:
559
63
.
266.
Routy
B
,
Le Chatelier
E
,
Derosa
L
,
Duong
CPM
,
Alou
MT
,
Daillere
R
, et al
Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors
.
Science
2018
;
359
:
91
7
.
267.
Gopalakrishnan
V
,
Spencer
CN
,
Nezi
L
,
Reuben
A
,
Andrews
MC
,
Karpinets
TV
, et al
Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients
.
Science
2018
;
359
:
97
103
.
268.
Siegel
RL
,
Miller
KD
,
Jemal
A
. 
Cancer statistics, 2020
.
CA Cancer J Clin
2020
;
70
:
7
30
.
269.
Fane
M
,
Weeraratna
AT
. 
Normal aging and its role in cancer metastasis
.
Cold Spring Harb Perspect Med
2020
;
10
:
a037341
.
270.
Fane
M
,
Weeraratna
AT
. 
How the ageing microenvironment influences tumour progression
.
Nat Rev Cancer
2020
;
20
:
89
106
.
271.
Coppe
JP
,
Desprez
PY
,
Krtolica
A
,
Campisi
J
. 
The senescence-associated secretory phenotype: the dark side of tumor suppression
.
Annu Rev Pathol
2010
;
5
:
99
118
.
272.
Campisi
J
. 
Aging, cellular senescence, and cancer
.
Annu Rev Physiol
2013
;
75
:
685
705
.
273.
Kaur
A
,
Webster
MR
,
Marchbank
K
,
Behera
R
,
Ndoye
A
,
Kugel
CH
 3rd
, et al
sFRP2 in the aged microenvironment drives melanoma metastasis and therapy resistance
.
Nature
2016
;
532
:
250
4
.
274.
Krtolica
A
,
Parrinello
S
,
Lockett
S
,
Desprez
P-Y
,
Campisi
J
. 
Senescent fibroblasts promote epithelial cell growth and tumorigenesis: a link between cancer and aging
.
Proc Natl Acad Sci
2001
;
98
:
12072
7
.
275.
Kaur
A
,
Ecker
BL
,
Douglass
SM
,
Kugel
CH
 3rd
,
Webster
MR
,
Almeida
FV
, et al
Remodeling of the collagen matrix in aging skin promotes melanoma metastasis and affects immune cell motility
.
Cancer Discov
2019
;
9
:
64
81
.
276.
Ecker
BL
,
Kaur
A
,
Douglass
SM
,
Webster
MR
,
Almeida
FV
,
Marino
GE
, et al
Age-related changes in HAPLN1 increase lymphatic permeability and affect routes of melanoma metastasis
.
Cancer Discov
2019
;
9
:
82
95
.
277.
López-Otín
C
,
Blasco
MA
,
Partridge
L
,
Serrano
M
,
Kroemer
G
. 
The hallmarks of aging
.
Cell
2013
;
153
:
1194
217
.
278.
Leonardi
GC
,
Accardi
G
,
Monastero
R
,
Nicoletti
F
,
Libra
M
. 
Ageing: from inflammation to cancer
.
Immun Ageing
2018
;
15
:
1
.
279.
Kugel
CH
 III
,
Douglass
SM
,
Webster
MR
,
Kaur
A
,
Liu
Q
,
Yin
X
, et al
Age correlates with response to anti-PD1, reflecting age-related differences in intratumoral effector and regulatory T-cell populations
.
Clin Cancer Res
2018
;
24
:
5347
56
.
280.
Ganesh
K
,
Massagué
J
. 
Targeting metastatic cancer
.
Nat Med
2021
;
27
:
34
44
.
281.
Balak
MN
,
Gong
Y
,
Riely
GJ
,
Somwar
R
,
Li
AR
,
Zakowski
MF
, et al
Novel D761Y and common secondary T790M mutations in epidermal growth factor receptor–mutant lung adenocarcinomas with acquired resistance to kinase inhibitors
.
Clin Cancer Res
2006
;
12
:
6494
501
.
282.
Vasan
N
,
Baselga
J
,
Hyman
DM
. 
A view on drug resistance in cancer
.
Nature
2019
;
575
:
299
309
.
283.
Al-Hajj
M
,
Becker
MW
,
Wicha
M
,
Weissman
I
,
Clarke
MF
. 
Therapeutic implications of cancer stem cells
.
Curr Opin Genet Dev
2004
;
14
:
43
7
.
284.
Naik
S
,
Larsen
SB
,
Gomez
NC
,
Alaverdyan
K
,
Sendoel
A
,
Yuan
S
, et al
Inflammatory memory sensitizes skin epithelial stem cells to tissue damage
.
Nature
2017
;
550
:
475
80
.
285.
Zhou
S
,
Schuetz
JD
,
Bunting
KD
,
Colapietro
AM
,
Sampath
J
,
Morris
JJ
, et al
The ABC transporter Bcrp1/ABCG2 is expressed in a wide variety of stem cells and is a molecular determinant of the side-population phenotype
.
Nat Med
2001
;
7
:
1028
34
.
286.
Obenauf
AC
,
Zou
Y
,
Ji
AL
,
Vanharanta
S
,
Shu
W
,
Shi
H
, et al
Therapy-induced tumour secretomes promote resistance and tumour progression
.
Nature
2015
;
520
:
368
72
.
287.
Coppé
JP
,
Patil
CK
,
Rodier
F
,
Sun
Y
,
Muñoz
DP
,
Goldstein
J
, et al
Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor
.
PLoS Biol
2008
;
6
:
2853
68
.
288.
Faget
DV
,
Ren
Q
,
Stewart
SA
. 
Unmasking senescence: context-dependent effects of SASP in cancer
.
Nat Rev Cancer
2019
;
19
:
439
53
.
289.
Bailey
JM
,
Swanson
BJ
,
Hamada
T
,
Eggers
JP
,
Singh
PK
,
Caffery
T
, et al
Sonic hedgehog promotes desmoplasia in pancreatic cancer
.
Clin Cancer Res
2008
;
14
:
5995
6004
.
290.
Olive
KP
,
Jacobetz
MA
,
Davidson
CJ
,
Gopinathan
A
,
McIntyre
D
,
Honess
D
, et al
Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer
.
Science
2009
;
324
:
1457
61
.
291.
Gajewski
TF
,
Meng
Y
,
Blank
C
,
Brown
I
,
Kacha
A
,
Kline
J
, et al
Immune resistance orchestrated by the tumor microenvironment
.
Immunol Rev
2006
;
213
:
131
45
.
292.
Gajewski
TF
,
Woo
SR
,
Zha
Y
,
Spaapen
R
,
Zheng
Y
,
Corrales
L
, et al
Cancer immunotherapy strategies based on overcoming barriers within the tumor microenvironment
.
Curr Opin Immunol
2013
;
25
:
268
76
.
293.
Tauriello
DVF
,
Palomo-Ponce
S
,
Stork
D
,
Berenguer-Llergo
A
,
Badia-Ramentol
J
,
Iglesias
M
, et al
TGFbeta drives immune evasion in genetically reconstituted colon cancer metastasis
.
Nature
2018
;
554
:
538
43
.
294.
Mariathasan
S
,
Turley
SJ
,
Nickles
D
,
Castiglioni
A
,
Yuen
K
,
Wang
Y
, et al
TGFbeta attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells
.
Nature
2018
;
554
:
544
8
.
295.
Catenacci
DVT
,
Junttila
MR
,
Karrison
T
,
Bahary
N
,
Horiba
MN
,
Nattam
SR
, et al
Randomized phase Ib/II study of gemcitabine plus placebo or vismodegib, a hedgehog pathway inhibitor, in patients with metastatic pancreatic cancer
.
J Clin Oncol
2015
;
33
:
4284
92
.
296.
Ciardiello
D
,
Elez
E
,
Tabernero
J
,
Seoane
J
. 
Clinical development of therapies targeting TGFβ: current knowledge and future perspectives
.
Ann Oncol
2020
;
31
:
1336
49
.
297.
Lee
JJ
,
Perera
RM
,
Wang
H
,
Wu
D-C
,
Liu
XS
,
Han
S
, et al
Stromal response to Hedgehog signaling restrains pancreatic cancer progression
.
Proc Natl Acad Sci
2014
;
111
:
E3091
E100
.
298.
Boumahdi
S
,
de Sauvage
FJ
. 
The great escape: tumour cell plasticity in resistance to targeted therapy
.
Nat Rev Drug Discov
2020
;
19
:
39
56
.
299.
Paget
S
. 
The distribution of secondary growths in cancer of the breast. 1889
.
Cancer Metastasis Rev
1989
;
8
:
98
101
.