You are on page 1of 6

REVIEW ARTICLE

Paget’s “Seed and Soil” Theory of Cancer Metastasis:


An Idea Whose Time has Come
Mohammed Akhtar, MD, FCAP, FRCPath, FRCPA,*
Abdulrazzaq Haider, MD,* Sameera Rashid, MD,†
and Ajayeb Dakhilalla M.H. Al-Nabet, PhD*

cause the patient’s death. Thus, metastasis is the most life-


Abstract: The concept that the pattern of metastatic spread of threatening event in patients with cancer.1–3
cancer is not random and that cancer cells exhibit preferences when It has long been observed that most cancers show an
metastasizing to organs, dates back to 1889 when Steven Paget organ-specific pattern of metastases. For example, colon
published his “seed and soil” hypothesis. He proposed that the
spread of tumor cells is governed by interaction and cooperation
carcinomas metastasize usually to liver and lung but rarely
between the cancer cells (seed) and the host organ (soil). Extensive to bone, skin, brain, and kidneys. In contrast, breast carci-
studies during the last several decades have provided a better nomas, frequently form metastases in most of these organs.
understanding of the process of metastatic spread of cancer and Similarly, prostate cancer metastasizes to the bone and only
several stages such as intravasation, extravasation, tumor latency, rarely to lung and liver. Whether the distribution of meta-
and development of micrometastasis and macrometastasis have static disease is a random process determined by the patterns
been defined. Furthermore, recent studies have shown that the of blood supply and lymphatic flow or due to the active
target organs may be prepared for metastatic deposits by interaction between the tumor cells and the host organ has
the development of premetastatic niches. This specialized micro- long been a matter of debate.2–4
environment is involved in promoting tumor cell homing,
colonization, and subsequent growth at the target organ. The
The purpose of this review is to provide a brief per-
premetastatic niche consists of accumulation of aberrant immune spective on our understanding of the mechanisms of meta-
cells and extracellular matrix proteins in target organs. The primary stasis and to provide an update on recent findings with
tumor plays a key role in the development of premetastatic niches regard to the interactions between the cancer cells and host
by producing tumor-derived soluble factors which mobilize bone organ harboring the metastasis and to highlight changes in
marrow-derived hematopoietic cells to the premetastatic niche. the microenvironment of these organs before and after the
Exosomes-derived from the primary tumor also contribute to can- arrival of the metastatic cancer cells.
cer-favorable microenvironment in the premetastatic niches. These
changes prime the initially healthy organ microenvironment and
render it amenable for subsequent metastatic cell colonization. SEED AND THE SOIL THEORY
Steven Paget (son of the famous English surgeon and
Key Words: seed, soil, metastasis, exosomes, niche, premetastatic,
pathologist Sir James Paget) proposed the “seed and soil”
macrophage, intravasation, extravasation, bone marrow
theory of metastasis, which was based on analysis of 735
(Adv Anat Pathol 2019;26:69–74) fatal cases of breast cancer, complete with the autopsy, as
well as many other cancer cases from the literature. His
findings were published in Lancet in 1889. He argued that
the distribution of metastases cannot be due to chance: the
C ancer metastasis is the spread of cancer cells beyond
where the tumor originated to other parts of the body
with the formation of new tumors. It is the single event that
“seed” refers to certain tumor cells with metastatic potential,
and the “soil” is any organ or tissue providing a proper
results in the death of most patients with cancer. At the time environment for the growth of the seeds. Paget suggested
of cancer diagnosis, at least half of the patients already that the spread of metastatic cells was organ specific and not
present clinically detectable metastatic disease. Although the merely anatomic and involved interaction between the
primary tumor, in most cases may be eradicated by ther- cancer cells and the host organ. He concluded that meta-
apeutic modalities such as surgical resection, radiotherapy, stases developed only when the seed and soil were
and chemotherapy, its metastases, when distributed at dis- compatible.5 This concept was in opposition to the pre-
tant sites in the body, are most difficult to eradicate by any vailing mechanistic hypothesis of Rudolf Virchow who
of the currently available therapeutic means, and finally, considered metastasis as the arrest of tumor cell emboli in
the vasculature.2,6
In 1928, James Ewing7 challenged Paget’s “seed and
soil” theory and hypothesized that metastatic dissemination
From the *Department of Laboratory Medicine and Pathology, Divi- occurs by purely mechanical factors that are a result of the
sion of Anatomic Pathology; and †Department of Laboratory anatomic structure of the vascular system. Thus, it would
Medicine and Pathology, Hamad Medical Corporation, Doha,
Qatar.
be completely accounted for by the vascular connections of
The authors have no funding or conflicts of interest to disclose. the primary tumor so that tumor cell emboli are much more
Reprints: Mohammed Akhtar MD, FACP, FRCPath, FRCPA, likely to be mechanically trapped in the circulatory network
Department of Laboratory Medicine and Pathology, Hamad of the first connected organ, which will then sustain the
Medical Corporation, P.O. Box 3050, Doha, Qatar (e-mail:
dr.mohammed.akhtar@gmail.com).
highest burden of metastatic colonization.
All figures can be viewed online in color at www.anatomicpathology.com. Other organs receive less tumor cells and develop fewer
Copyright © 2018 Wolters Kluwer Health, Inc. All rights reserved. metastatic tumors. That became the dominant view point

Adv Anat Pathol  Volume 26, Number 1, January 2019 www.anatomicpathology.com | 69


Copyright r 2019 Wolters Kluwer Health, Inc. All rights reserved.
Akhtar et al Adv Anat Pathol  Volume 26, Number 1, January 2019

and for the next several decades the concept of “seed and
soil” languished in the shadows. This proposal, however,
does not explain the observation that some organs, such as
brain, bone, and adrenals, are served by a small fraction of
the circulatory volume, yet they are frequently involved by
metastatic deposits of certain cancers. In contrast, organs,
such as heart, muscle, skin, kidney, and spleen, each of
which receives a considerable supply of blood, only occa-
sionally develop metastatic disease.3
In 1979, Sugarbaker8 postulated that while the regional
metastases could be due to anatomic or mechanical issues
such as lymphatic drainage as stated by Ewing, metastases
to distant organs are site specific and require a different
explanation. Seminal work by Hart and Fidler in the 1980s FIGURE 2. Following intravasation the tumor cell may be sur-
supported Paget’s “seed and soil” theory by showing pref- rounded by platelets and fibrin which protect the cancer cell from
erential homing of tumor cells of B16 melanoma in specific the immune system. Please see this image in color online.
distant sites. Their work conclusively demonstrated that
while potentially metastatic tumor cells reached the vascu- as hemodynamic shear and from sensitized killer mono-
lature of all organs, the development of metastases occurred nuclear cells. At the same time, there is an activation of
selectively in certain organs but not others.9 thrombosis with fibrin deposition. The aggregation of pla-
telets, fibrin, and tumor cells creates a tumor-platelet com-
plex that essentially acts as an embolus (Fig. 2). From this
THE START OF THE METASTATIC JOURNEY point, these tumor cells move away from the primary site
The process of metastasis starts with the release of and circulate in the blood circulation where they would
malignant cells from its adhesive attachments to other cells encounter resistance by the immune system and the mech-
and the extracellular matrix including the basement mem- anical stresses of blood flow. Some tumor cells will even-
branes. Further, the cancer cells acquire mesenchymal-like tually survive and adopt a process to leave the blood
properties via a process called epithelial mesenchymal circulation, known as extravasation which is the reverse of
transition. This is mediated by cadherin molecule switching intravasation and in which cells adhere and penetrate the
involving downregulation of E-cadherin and upregulation of blood vessel wall again. The tumor cells that escape from the
mesenchymal cadherins such as N-cadherin, integrin-αvβ6, circulation, invade the host tissues.10–13 These cells undergo
vimentin, and matrix metalloproteinase.9 Such cells assume mesenchymal-epithelial transformation and are now des-
motile phenotype and acquire migratory capabilities and ignated as disseminated tumor cells (DTCs) (Fig. 3).
can interact with components of the extracellular matrix and
contribute to it by synthesizing and organizing new com-
ponents. They also remodel the extracellular matrix through HOMING OF CIRCULATING TUMOR CELLS
the production of matrix-degrading metalloproteinases. Various explanations have been proposed for the site
Thus, the loss of cell-cell adhesions allows malignant tumor selectivity and organ tropism of blood-borne distant meta-
cells to dissociate from the primary tumor and changes in stases, including tumor cell surface characteristics, adhesion
cell-matrix interaction enable the cells to invade the sur- between tumor cells and the target organ components and
rounding stroma. As these motile cells pass through the response to specific host tissue growth factors and chemo-
basement membrane and extracellular matrix, some tumor kines. An explanation for the different sites of tumor growth
cells will penetrate the blood vessels, thus entering the cir- involves interactions between the metastatic cells and the
culation (intravasation) (Fig. 1). Tumor cells that have organ environment, possibly in terms of specific binding to
successfully invaded the blood vessels adhere to blood pla- endothelial cells. Endothelial cells in the vasculature of
telets that protect them from destructive physical forces such different organs express different cell surface receptors
and growth factors that influence the phenotype of the

FIGURE 3. At the host organ the tumor cell penetrates the vas-
FIGURE 1. Diagram showing the carcinoma cells invading the cular wall and enters the host organ tissue. This is usually followed
stroma and undergoing EMT. The invading tumor cell may enter a by MET. DTC indicates disseminated tumor cells; MET, mesen-
blood vessel and enter circulation (intravasation). EMT indicates chymal to epithelial transformation. Please see this image in color
epithelial to mesenchymal transformation. online.

70 | www.anatomicpathology.com Copyright © 2018 Wolters Kluwer Health, Inc. All rights reserved.
Copyright r 2019 Wolters Kluwer Health, Inc. All rights reserved.
Adv Anat Pathol  Volume 26, Number 1, January 2019 Atypical Thymoma

corresponding metastases. Greene and Harvey first sug-


gested that the organ distribution patterns of metastatic foci
were dependent on the formation of sufficient adhesive
bonds between arrested tumor cells and endothelial cells,
and they hypothesized that these interactions were similar
to those of lymphocyte/endothelial cells at sites of
inflammation.14–16
Circulating immune and stem cells are known to use
chemokine-mediated signaling to home on specific organs.
Chemokines are growth factor–like molecules that bind to
G-coupled receptors. They induce leukocytes to adhere
tightly to endothelial cells and to migrate towards a gra-
dient. Tumor cells also use similar mechanisms to direct
metastatic organ preference. The best-known example is the
expression of chemokine receptor 4 (CXCR4) in breast
cancer. Tumors with CXCR4 expression migrate to organs
that express high levels of its ligand CXCL12. CXCL12 is
expressed in the common sites of metastasis for breast
FIGURE 5. Photomicrograph featuring a micrometastasis of
cancer: lung, liver, bone marrow, and brain. Similarly, breast cancer in a lymph node. Please see this image in color
melanoma cells may be attracted to certain organs using online.
CCL27/CCR10 chemokine receptor combination.16,17
despite active tumor cell proliferation, the tumor size
remains too small for clinical detection by conventional
TUMOR LATENCY methods.19–21
The fact that tumor cells arrive and establish residence The duration of metastatic latency varies between cancer
at a new and potentially hostile environment does not mean types, and for the most aggressive ones it is very short, resulting
that they will necessarily survive and proliferate in the new in high relapse and mortality rates following diagnosis. In lung
location. Survival and subsequent proliferation, however, is cancer, the metastatic latency interval usually lasts only a few
a prerequisite for development of a secondary tumor at the weeks. In this type of cancer, malignant cells acquire metastatic
new location. Only a small fraction of these cells ultimately traits for rapid and massive cell dissemination, followed by
survives in the new tissue location. It has been estimated that colonization of multiple secondary organs. The short latency in
<0.01% of circulating tumor cells eventually succeed in lung cancer implies that malignant cells in the primary tumor
forming secondary growths.18 acquire most of the metastatic traits, thus enabling them to
After extravasation, a common theme of the metastatic overtake organs immediately after arrival. In contrast, a well-
process is settlement of DTCs into latency, which can last known example of a tumor type with very long latency is
from several weeks to decades (Fig. 4). In cellular latency prostate cancer. Similarly, some of the breast cancer metastasis
isolated tumor cells enter a state of proliferative quiescence. may also remain dormant only to present with widespread
Indeed, in patient bone marrow samples most DTCs are metastasis years and even decades later.22
found as quiescent single cells. Another form of tumor
latency is the establishment of micrometastasis (Fig. 5). This
is metastatic lesion where tumor cells form small aggregates THE METASTATIC NICHE AND FORMATION
in which rate of proliferation and tumor cell attrition are OF MACROMETASTASIS
exactly balanced. The loss of tumor cells may be due to DTCs need time to alter or unleash the required
apoptosis, insufficient vascularization, or to constant culling functions for tumor initiation and expansion in the secon-
by immune defenses. Consequently, in a micrometastasis dary site. For the successful establishment of a metastasis
several crucial events must take place, the most important
among which are cell cycle activity and angiogenesis. It is
obvious then that there are intrinsic differences in genomic
makeup of tumor cells that must be evaluated to understand
the factors intrinsic to the cell (seed). However, it is equally
important to acknowledge the microenvironmental con-
dition of the host tissue, the soil that plays a key role in the
acquisition of proliferative phenotype by the tumor cells.
Studies using, radioactive labeling of the injected can-
cer cells showed that they were equally likely to be trapped
in a variety of tissue.18 So, just landing in a tissue is not
enough for cancer cells to develop a secondary tumor;
rather, some role of the tissue itself may be crucial in sus-
taining the new growth. The findings of Tarin et al23 that the
development of secondary cancer was rare even upon direct
FIGURE 4. The DTC enter a stage of tumor latency which may
last for a few weeks to decades. During latency tumor cells may be deposition of millions of tumor cells into the vena cava
disposed as isolated cells or in the form of micrometastasis. They (to reduce ascites from ovarian cancer) is a good indication
may also be destroyed by the immune system or undergo of the importance of other factors involved.
apoptosis. DTC indicates disseminated tumor cells. Please see this In stem cell biology the specialized microenvironment
image in color online. that supports stem cell maintenance and actively regulates

Copyright © 2018 Wolters Kluwer Health, Inc. All rights reserved. www.anatomicpathology.com | 71
Copyright r 2019 Wolters Kluwer Health, Inc. All rights reserved.
Akhtar et al Adv Anat Pathol  Volume 26, Number 1, January 2019

cell function and proliferation is termed as niche.24 A similar


model has been suggested to delineate the interactions of
malignant cells with their microenvironment at metastatic
sites. This microenvironment comprises supportive non-
neoplastic stromal cells, soluble factors, vascular networks,
nutrients and metabolic components, and the structural
extracellular matrix architecture. Although the precise
genetic makeup of a cell is undoubtedly pivotal in deter-
mining its malignant phenotype, the metastatic niche model
stipulates that microenvironmental factors are also impor-
tant in permitting malignant cells to realize their metastatic
potential. Thus, with the right genetic makeup of the tumor
cells accompanied by a permissive and supportive environ-
ment in the host organ, the tumor cells may proliferate and FIGURE 6. Cartoon depicting the formation of a premetastatic
ultimately develop into a clinically detectable macrometa- niche. The primary tumor produces TDSFs that start the formation of
stasis. The metastatic niche model suggests that a suitably the premetastatic niche by transfer of BDHCs to the target organ.
conducive microenvironment must evolve for tumor cells The primary tumor also causes accumulation of TAMs in the pre-
to be able to engraft and proliferate at secondary sites with metastatic niche. These cells protect disseminated tumor cells from
the transition from micrometastatic to macrometastatic the immune system. TDSFs also cause inflammation in the devel-
status.25 The idea that cancer cells require some nourish- oping premetastatic niche with accumulation of inflammatory cells
ment and support from their environment to develop is an and formation of the intercellular matrix. Increased vascularity also
important focus of research today, with the aim of unrav- contributes to the niche formation. Tumor-derived exosome play a
crucial role in maintaining a cancer friendly microenvironment in the
eling the molecular mechanisms that bring seed and soil niche. BDHC indicates bone marrow-derived hematopoietic cells;
together to promote metastasis. TAM, tumor-associated macrophages; TDSF, tumor-derived soluble
factors. Please see this image in color online.

THE CONCEPT OF PREMETASTATIC NICHE


The premetastatic niche can be defined as a supportive and
receptive tissue microenvironment undergoing a series of which would otherwise be more likely to be effective in halting
molecular and cellular changes to form the metastatic-des- metastasis. Myeloid progenitor cells, recruited at various stages
ignated sites, prior to the arrival of the metastatic tumor. in their cell cycle, are believed to constitute much of the
Dr Lyden and colleagues pioneered the research on the pre- premetastatic niche, as they can protect the tumor cells from the
metastatic niche and the role and significance of the premeta- standard immune response as the cancer cells attempt to colonize
static niche in metastasis has attracted more and more attention the premetastatic niche. Given their important role in protecting
in recent years.26 Their landmark study was the first demon- the growing metastasis from immune system attacks, myeloid
stration of a microenvironment designed to attract tumor cells cells are a key factor in the development of the premetastatic
to a target organ and set the stage for future work to discover niche, and thus eventually in promoting metastases.29,31
additional factors that contribute to premetastatic niche Chemokines, also play a significant role in the creation of
formation. premetastatic niches and metastases. The primary tumor,
The process of premetastatic niche formation in distant to evade detection by the immune system, uses chemokines
organs is initiated by the primary tumor that produces tumor- to increase recruitment of bone marrow-derived myeloid cells
derived secreted factors prior to tumor dissemination.27 These to secondary organs. In addition, cancer cells from the
factors include vascular endothelial growth factor (VEGF-A) primary tumor can be used to induce inflammation in the
and placental growth factor among others. These factors future site of the premetastatic niche in the secondary organ,
increase the proliferation of fibroblast-like stromal cells, which which is like the immune response created by an infection.
contribute to local deposition of fibronectin. Tumor-derived Thus, the large presence of immune cells allows the pre-
secreted factors promote premetastatic niche formation by metastatic niche to ward off attacks by the immune system
mobilizing and recruiting VEGFR1+ bonemarrow-derived and therefore allow the tumor to metastasize without
hematopoietic progenitor cells directly from the bone marrow inhibition.29,32
to the premetastatic niche (Fig. 6). These cells express VLA−4 The formation of a premetastatic niche not only involves
that binds to fibronectin and allows them to assemble at the the recruitment of foreign cells, such as immune cells, but also
site. Most notably, the VEGFR1+ niche cells act as harbingers the reprogramming of the resident stromal cells to facilitate
of organ-specific carcinoma spread. Others, such as tumor metastatic growth. Normal lung fibroblasts express miR-30
necrosis factor alpha (TNF-a) and transforming growth factor family members to restrain MMPs, such as MMP9, to stabilize
b (TGF-b), along with VEGF-A, induce the expression of the lung vasculature. Cancer cells reprogram fibroblasts to
S100A8 and S100A9 in the lung to develop premetastatic decrease their expression of miR-30 family members, resulting
niches.25–30 in enhanced MMP activity, vascular permeability, and meta-
Premetastatic niche formation is facilitated in large part by stasis. Factors secreted from the primary tumor induce the
the presence of a suppressed immune system. Primary tumors expression of α-smooth muscle actin—in premetastatic fibro-
recruit myeloid cells, which are the precursors to immune cells. blasts, activating them to induce remodeling extracellular matrix
These cells enable the tumor cells to avoid detection by the through secretion of fibronectin, Lysyl Oxidases LOX and
immune system as they metastasize, and thus allow the meta- LOXL2, thereby generating a more permissive microenviron-
stasis to flourish. Once the primary tumors have entered the ment for survival and outgrowth of DTC.28,29,33–35
bloodstream, myeloid cells that have been recruited by the tumor Thus, immune suppression, combined with hypoxia
protect the cancer cells from detection by the immune system, and changes in extracellular matrix, among other processes,

72 | www.anatomicpathology.com Copyright © 2018 Wolters Kluwer Health, Inc. All rights reserved.
Copyright r 2019 Wolters Kluwer Health, Inc. All rights reserved.
Adv Anat Pathol  Volume 26, Number 1, January 2019 Atypical Thymoma

are essential steps in creating premetastatic niches that allow in intercellular communication. The capability of exosomes
tumor cells to grow in a foreign and hostile environment to transfer proteins, DNA, mRNA, as well as non-coding
without being destroyed by the typical response of the RNAs has made them an attractive focus of research into
immune system. the pathogenesis of different diseases, including cancer.40,41
It has been noted that cancer cells secrete much higher
amounts of exosomes in comparison with nontransformed
ROLE OF EXOSOMES IN TUMOR METASTASIS cells. These exosomes not only influence proximal tumor cells
Exosomes are spherical to cup shaped, lipid bilayer and stromal cells in local microenvironment, but also can
membrane nanovesicles 40 to 100 nm in diameter. These exert systemic effects when participating in blood circulation.
vesicles are secreted by many cells and can be found in most Exosomes have been shown to be implicated in the induction
body fluids such as urine and blood as well as in super- of apoptosis of cytotoxic T cells, expansion and function of
natants of cultured cells. Exosomes must be differentiated regulatory T cells (Treg cells), induction of M2 polarization of
from other secreted cellular entities such as microvesicles macrophages, inhibition of cytotoxicity of natural killer cells,
(50 to 1000 nm in diameter) and ectosomes, which are inhibition of differentiation of dendritic cells, expansion and
microvesicles derived from neutrophils or monocytes and activation of myeloid-derived suppressor cells and mobi-
apoptotic bodies (500 to 2000 nm in diameter).36,37 lization of neutrophils. Exosomes released under hypoxic
Exosome formation is a fine-tuned process which conditions can stimulate angiogenesis through interactions
includes 4 stages: initiation, endocytosis, multivesicular with endothelial cells.
bodies formation, and exosome secretion. Multivesicular Exosomal transforming growth factor β (TGFβ) can
bodies are endocytic structures formed by the budding of an induce differentiation of fibroblasts into tumor-supporting
endosomal membrane into the lumen of the compartment myofibroblasts and exosomes from ovarian cancer are
(Fig. 7). After vesicular accumulation, the multivesicular able to convert adipose-derived mesenchymal stem cells
bodies are either sorted for cargo degradation in the lyso- into myofibroblast-like cells, supporting tumor growth and
some or released into the extracellular space as exosomes by angiogenesis.36,41
fusing with the plasma membrane.38 As exosomes can transfer specific proteins and nucleic
In addition to lipids, nucleic acids, and proteins have acids to recipient cells in the tumor microenvironment or at
also been detected in exosomes. The protein composition of specific distant sites, cancers have used exosomes as a tool
tumor cell-derived exosomes has been well characterized for by which cancer cells can transfer malignant phenotype to
several cancers by using different proteomic methods. To normal cells and establish a fertile local and distant micro-
date, 4563 proteins, 1639 mRNAs, and 764 miRNAs have environment to help cancer cell growth. Exosomes con-
been identified in exosomes from different species and tis- tribute to tumor metastasis by enhancing tumor cell
sues by independent examinations.39 migration and invasion, remodeling the extracellular matrix
The exosomes can transfer their constituentsand cargo and establishing premetastatic niche.36,41–43
to neighboring or distant cells with preservation of their
function. Several mechanisms for the uptake of exosomes by
recipient cells, such as exosome fusion with the membrane of CONCLUSIONS
the recipient cell, endocytosis by phagocytosis, and receptor- A better understanding of the mechanisms of meta-
ligand interaction.40 Initially discovered as the garbage bags static disease in recent years seems to support the seed and
for removal of unwanted material from cells, the role of soil theory proposed by Steven Pagets almost 130 years ago.
exosomes in immune response and cancer is gradually rec- Preventing metastasis in high-risk patients would be far
ognized. Exosomes are now considered important mediators better than having to treat it later. Recent recognition of the
concept of the premetastatic niche allows researchers to
consider several new possibilities for treating cancer.44
Research on the mechanisms that control and support
the viability of latent metastatic cells should yield clues for
targeting cancer with the goal of preventing metastasis.
Factors from the primary tumor that structurally alter the
secondary organ to facilitate its colonization by tumor cells
could also potentially be targeted to stop metastasis. Rec-
ognition of the role of exosomes in tumor progression is also
an important potential target for early detection of cancer
and prevention of premetastatic niche formation.

REFERENCES
1. Liotta LA, Stetler-Stevenson WG. Principles of Molecular Cell
FIGURE 7. Early endosome is formed from the plasma Biology of Cancer: Cancer Metastasis, 4th ed. Philadelphia, PA:
membrane via endocytic pathway. MVB can be formed by the JB Lippincott Co; 1993.
invagination of the endosomal membrane. During the biogenesis 2. Chambers AF, Groom AC, Mac Donald IC. Dissemination
of exosomes and before their secretion, proteins (cytosolic pro- and growth of cancer cells in metastatic sites. Nat Rev Cancer.
teins, receptors), nucleic acids (eg, mRNA, miRNA, DNA), and 2002;2:563–572.
lipids are uploaded to exosomes. Dependent on the function and 3. Ribatti D, Mangialardi G, Vacca A. Stephen Paget and the
content, MVB then can be directed to fuse with the plasma ‘seed and soil’ theory of metastatic dissemination. Clin Exp
membrane and release to the extracellular space as exosomes. Med. 2006;6:145–149.
Alternatively, contents of MVB may be degraded by fusion with 4. Nguyen DX, Bos PD, Massagué J. Metastasis: from dissem-
lysosomes. MVB indicates multivesicular body. Please see this ination to organ specific colonization. Nat Rev Cancer. 2009;
image in color online. 9:274–284.

Copyright © 2018 Wolters Kluwer Health, Inc. All rights reserved. www.anatomicpathology.com | 73
Copyright r 2019 Wolters Kluwer Health, Inc. All rights reserved.
Akhtar et al Adv Anat Pathol  Volume 26, Number 1, January 2019

5. Paget S. The distribution of secondary growths in cancer of the 25. Psaila B, Lyden D. The metastatic niche: adapting the foreign
breast. Lancet. 1889;133:571–573. soil. Nat Rev Cancer. 2009;9:285–293.
6. Pachmayr E, Treese C, Stein U. Underlying mechanisms for 26. Kaplan RN, Riba RD, Zacharoulis S, et al. VEGFR1-positive
distant metastasis —molecular biology. Visc Med. 2017;33: haematopoietic bone marrow progenitors initiate the pre-
11–20. metastatic niche. Nature. 2005;438:820–827.
7. Ewing J. ed. Neoplastic Diseases: a Treatise on Tumours 27. Peinado H, Lavotshkin S, Lyden D. The secreted factors
Metastasis, 3rd ed. Philadelphia: Saunders; 1928:76–88. responsible for pre-metastatic niche formation: old sayings and
8. Sugarbaker EV. Cancer metastasis: a product of tumor host new thoughts. Semin Cancer Biol. 2011;21:139–146.
interactions. Curr Probl Cancer. 1979;3:1–59. 28. Chin AR, Wang SE. Cancer tills the premetastatic field:
9. Hart IR, Fidler IJ. Role of organ selectivity in the determi- mechanistic basis and clinical implications. Clin Cancer Res.
nation of metastatic patterns of B16 melanoma. Cancer Res. 2016;22:3725–3733.
1980;40:2281–2287. 29. Peinado H, Zhang H, Matei IR, et al. Pre-metastatic niches: organ-
10. Nguyen DX, Bos PD, Massague J. Metastasis: from dissem- specific homes for metastases. Nat Rev Cancer. 2017;17:302–317.
ination to organ-specific colonization. Nat Rev Cancer. 30. Liu Y, Cao X. Characteristics and significance of the
2009;9:274–284. premetastatic niche. Cancer Cell. 2016;30:668–681.
11. Martin TA, Ye L, Sanders AJ, et al. Cancer invasion and 31. Kitamura T, Qian B-Z, Pollard JW. Immune cell promotion of
metastasis: molecular and cellular perspective. In: Jandial metastasis. Nat Rev Immunol. 2015;15:73–86.
Rahul, ed. Metastatic Cancer: Clinical and Biological Perspec- 32. Law AMK, Lim E, Ormandy CJ, et al. The innate and adaptive
tives. Austin, TX: Landes Bioscience; 2013. infiltrating immune systems as targets for breast cancer
12. Kent W, Hunter KW, Crawford NPS, et al. Mechanisms of immunotherapy. Endocr Relat Cancer. 2017;24:R123–R144.
metastasis. Breast Cancer Res. 2008;10(suppl 1):S2 1–S2 10. 33. Aguado BA, Bushnell GG, Rao SS, et al. Engineering the pre-
13. Munguti J, Sammy M. Mechanisms of tumor metastasis: metastatic niche. Nat Biomed Eng. 2017;1:1–28.
anatomical mimicry? OA Anatomy. 2013;1:23. 34. Cox TR, Bird D, Baker A-M, et al. LOX-mediated collagen
14. Bogenrieder T, Herlyn M. Axis of evil: molecular mechanisms crosslinking is responsible for fibrosis-enhanced metastasis.
of cancer metastasis. Oncogene. 2003;22:6524–6536. Cancer Res. 2013;73:1721–1732.
15. Greene HS, Harvey EK. The relationship between the 35. Lu P, Weaver VM, Werb Z. The extracellular matrix: a dynamic
dissemination of tumor cells and the distribution of metastases. niche in cancer progression. J Cell Boil. 2012;196:395–406.
Cancer Res. 1964;24:799–811. 36. Weidle HU, Birzele F, Kollmorgen G, et al. The multiple roles
16. Sarvaiya PJ, Guo D, Ulasov I, et al. Chemokines in tumor of exosomes in metastasis. Cancer Genomics Proteomics.
progression and metastasis. Oncotarget. 2013;4:2171–2185. 2017;14:1–16.
17. Mukherjee D, Zhao J. The role of chemokine receptor CXCR4 37. Thery C, Zitvogel L, Amigorena S. Exosomes: composition,
in breast cancer metastasis. Am J Cancer Res. 2013;3:46–57. biogenesis and function. Nat Rev Immunol. 2002;2:569–579.
18. Fidler IJ. Metastasis: quantitative analysis of distribution and 38. Li W, Li C, Zhou T, et al. Role of exosomal proteins in cancer
fate of tumor emboli labeled with 125 I-5-iodo-2′-deoxyuridine. diagnosis. Mol Cancer. 2017;16:145.
J Natl Cancer Inst. 1970;45:773–782. 39. Zhang X, Yuan X, Shi H, et al. Exosomes in cancer: small
19. Pollard JW. Defining metastatic cell latency. N Engl J Med. particle, big player. J Hematol Oncol. 2015;8:83.
2016;375:280–282. 40. Mulcahy LA, Pink RC, Carter DRF. Routes and mechanisms
20. Massagué J, Obenauf JA AC. Metastatic colonization. Nature. of extracellular vesicle uptake. J Extracell Vesicles. 2014;3:10.
2016;529:298–306. 41. Rashed MH, Bayraktar E, Helal GK, et al. Exosomes: from
21. Lambert WA, Pattabiraman DR, Weinberg RA. Emerging garbage bins to promising therapeutic targets. Int J Mol Sci.
biological principles of metastasis. Cell. 2017;168:4670–4691. 2017;18:538.
22. Gomis RR, Gawrzak S. Tumor cell dormancy. Mol Oncol. 42. Young Hwa Soung YH, Nguyen T, Cao H, et al. Emerging
2017;11:62–78. roles of exosomes in cancer invasion and metastasis. BMB Rep.
23. Tarin D, Price J, Kettlewell M, et al. Mechanisms of human 2016;49:18–25.
tumor metastasis studied in patients with peritoneovenous 43. Jia Y, Chen Y, Wang Q, et al. Exosome: emerging biomarker in
shunts. Cancer Res. 1984;44:3584–3592. breast cancer. Oncotarget. 2017;8:41717–41733.
24. Morrison SJ, Spradling AC. Stem cells and niches: mechanisms 44. Zoccoli A, Iuliani M, Pantano F, et al. Premetastatic niche:
that promote stem cell maintenance throughout life. Cell. ready for new therapeutic interventions? Exp Opin Ther
2008;132:598–611. Targets. 2012;16(suppl 2):S119–S129.

74 | www.anatomicpathology.com Copyright © 2018 Wolters Kluwer Health, Inc. All rights reserved.
Copyright r 2019 Wolters Kluwer Health, Inc. All rights reserved.

You might also like