Nothing Special   »   [go: up one dir, main page]

Cancer and Tumor Microenvironment

Download as pdf or txt
Download as pdf or txt
You are on page 1of 20

NIH Public Access

Author Manuscript
Cancer Chemother Pharmacol. Author manuscript; available in PMC 2010 April 22.
Published in final edited form as:
NIH-PA Author Manuscript

Cancer Chemother Pharmacol. 2009 March ; 63(4): 571–582. doi:10.1007/s00280-008-0881-9.

Cancer and the tumor microenvironment: a review of an


essential relationship

Flaubert Mbeunkui1 and Donald J. Johann Jr2


1Department of Molecular and Structural Biochemistry, North Carolina State University, 128 Polk

Hall, Campus Box 7622, Raleigh, NC 27695, USA fmbeunk@ncsu.edu


2Center for Cancer Research, Medical Oncology Branch, National Cancer Institute, National
Institutes of Health, Bethesda, MD 20892, USA johannd@mail.nih.gov

Abstract
Purpose—The role of the microenvironment during the initiation and progression of
carcinogenesis is now realized to be of critical importance, both for enhanced understanding of
fundamental cancer biology, as well as exploiting this source of relatively new knowledge for
NIH-PA Author Manuscript

improved molecular diagnostics and therapeutics.


Methods—This review focuses on: (1) the approaches of preparing and analyzing secreted
proteins, (2) the contribution of tumor microenvironment elements in cancer, and (3) the potential
molecular targets for cancer therapy.
Results—The microenvironment of a tumor is an integral part of its physiology, structure, and
function. It is an essential aspect of the tumor proper, since it supplies a nurturing environment for
the malignant process. A fundamental deranged relationship between tumor and stromal cells is
essential for tumor cell growth, progression, and development of life threatening metastasis.
Improved understanding of this interaction may provide new and valuable clinical targets for
cancer management, as well as risk assessment and prevention. Non-malignant cells and secreted
proteins from tumor and stromal cells are active participants in cancer progression.
Conclusions—Monitoring the change in the tumor micro-environment via molecular and
cellular profiles as tumor progresses would be vital for identifying cell or protein targets for cancer
prevention and therapy.

Keywords
NIH-PA Author Manuscript

Tumor; Microenvironment; Secreted proteins; Extracellular matrix; Molecular targets; Therapy

Introduction
Cancer is the number one cause of death in the United States for people less than 75 years
old. Every year, more than 11 million people are diagnosed with cancer throughout the
world and it may likely increase to 16 million by 2020. In 2005, cancer accounted for 7.6
million deaths from a total of 58 million deaths worldwide [1]. As a result, a re-evaluation of
our basic assumptions concerning the nature of cancer and how to better assess risk, prevent,
and medically manage is a high priority.

Cancer development has been defined as a multistep process in which somatic cells first
undergo an initiating event (i.e., environmental insult) and then a second or promoting event.
Both events accumulate genetic modifications. The fact that cancer cells have mutated
genomes is well established [2]. A carcinogen or mutagen, for instance from tobacco smoke,
when inhaled in sufficient quantity and duration may act as an adduct forming an unwanted
Mbeunkui and Johann Page 2

bond on DNA and potentially mutating a gene. However, many cancers will develop as a
result of a chronic inflammatory state due to infections.
NIH-PA Author Manuscript

For instance, this is commonly seen with Hepatitis B and C, which can be a harbinger for
Hepatocellular Carcinoma. Gastric infection from Helicobacter pylori, will increase gastric
cancer risk by 75%, and is the second most common type of cancer globally [3]. A few other
clinical examples illustrating the association of chronic inflammation and increased cancer
risk include: inflammatory bowel disease (ulcerative colitis, Crohn's disease) and colon
cancer; cervical infection (human papillomavirus) and cervical cancer, and chronic reflux
esophagitis resulting in Barrett's esophagus that is high risk for esophageal carcinoma. In all
cases, these chronic inflammatory conditions help to establish a tumor microenvironment
full of deranged proliferative signaling networks, which is largely orchestrated by
inflammatory cells and is an indispensable participant in the neoplastic process [4].

The tumor microenvironment was lately recognized as the product of a developing crosstalk
between different cells types. For instance, in epithelial tumors these cells include the
invasive carcinoma and its stromal elements. Critical stromal elements include cancer-
associated fibroblasts, which provide an essential communication network via secretion of
growth factors and chemokines, inducing an altered ECM thus providing additional
oncogenic signals enhancing cancer-cell proliferation and invasion [5]. Active contribution
of tumor-associated stromal cells to cancer progression has been recognized [2,6]. Stromal
NIH-PA Author Manuscript

elements consists of the extracellular matrix (ECM) as well as fibroblasts of various


phenotypes, and a scaffold composed of immune and inflammatory cells, blood and lymph
vessels, and nerves.

For tumors to progress and develop into life threatening entities, they must develop four
critical abilities. First, the ability to move, second the capacity to degrade tissue matrix
(ECM), third the aptitude to survive in blood and finally the physical quality of being able to
establish itself in a new tissue environment. But how exactly do cancer cells acquire these
traits? Recent scientific evidence points towards cancer cells using activated transcription
factors from development/embryology programs, thus gaining pleiotrophic abilities. The
microenvironment is of critical importance for success in this process [7].

The microenvironment of cancer cells provide the necessary signals that turn on the
transcription factors. Thus, it is the stromal or non-malignant cells that induce the requisite
transcription programs allowing the necessary mesenchymal phenotypes to invade distant
tissues and establish a new environment. The cancer cells must then shut down the
transcription factor programs and reconvert from mesenchymal to epithelial cells, thus
recreating themselves from the core of primary tumor cells. It is felt that the role of the
NIH-PA Author Manuscript

microenvironment is essential in all of these steps [7].

Tumor cells directly secrete a variety of proteins that include growth factors and ECM-
degrading proteinases or induce the host to elaborate biomolecules that are able to degrade
the matrix and its component adhesion molecules (Fig. 1). The matrix degradation takes
place in a region close to the tumor cell surface, where the amount of the active degradative
enzymes outbalances the natural proteinase inhibitors present in the matrix or that secreted
by normal cells [8]. Proteins secreted by tumor cell into the ECM microenvironment are
therefore involved in cell adhesion, motility, intercellular communication and invasion. The
knowledge and control of the direct microenvironment within a growing tumor is as
important as the corresponding knowledge and control of the abnormal behavior of
epithelial cells within that tumor.

Secreted proteins are of particular importance because they play a key role in a disease state
or the biological pathway leading to disease, and their identification and characterization

Cancer Chemother Pharmacol. Author manuscript; available in PMC 2010 April 22.
Mbeunkui and Johann Page 3

may lead to the understanding of disease patterns. In cancer, the understanding of the
interactions between tumor and stroma is needed for the development of more effective
therapies. The analysis of secreted proteins for proteomics studies is usually a challenge
NIH-PA Author Manuscript

because proteins are generally secreted at low concentration in the culture media, which
makes their recovery difficult due to dynamic range (concentration) constraints.
Accordingly, very few studies have been published to date analyzing this important class of
proteins that may help in the better understanding of tumor progression.

Although some recent reviews have focused on the contribution of cancer development for
different cell types present in the tumor microenvironment [9-11], the identification and
targeting of proteins secreted by cancer cells into the tumor microenvironment remains
relatively unexplored. Secreted protein signatures in cancer provide important information
that may be an aid to earlier diagnosis, improved disease monitoring as well as enhanced
assessment of the efficacy of therapy along with rationale for required revisions of therapy.
This review focuses on: (1) the approaches of preparing and analyzing secreted proteins, (2)
the contribution of secreted proteins in tumor progression, and (3) the identification of
secreted biomarkers for targets in tumor therapy.

Methods for preparing secreted proteins


The subset of proteins occurring in the conditioned media from cultured cells was previously
NIH-PA Author Manuscript

called “secretome” [12]. A concrete examination of the composition of this subproteome


reveals that the term secretome does not comprise only those proteins secreted through the
classical pathways (i.e., Golgi and endoplasmic reticulum). The secretome in an extensive
sense consists of proteins released through various mechanisms including classical secretion,
secretion through non-classical pathways, and released through secretion of exosomes.
Hitherto, proteomic approaches to analyze the secretome detected only a fraction of proteins
secreted from the cell because many secreted proteins are expressed only by specialized cell
types, have an induced expression during particular cellular response or are expressed during
specific stages of development.

Detection and identification of protein secreted by cells into the ECM, has turned out to be a
challenge for several reasons. In vitro, cells are grown in media rich in salts and
supplemented with serum proteins. Furthermore, a very small amount of proteins are
secreted by cells into the extra-cellular microenvironment. To detect this category of
proteins, the growth culture media should be free of any contaminant proteins.
Consequently, cells are usually grown in serum-free media with the eventual consequence of
cell death. Prior to cell incubation in serum-free media, the washing step may be adjusted so
that the remaining cytosolic proteins and serum are washed away without cell stressing
NIH-PA Author Manuscript

[13-17]. The confluence of cells grown in flask or on plate are then controlled to keep
enough space for cell growth, thus avoiding cell deaths due to saturation (Table 1).

Even a very small number of dead cells are able to release an amount of proteins that far
exceeds the amount of proteins really secreted. Cell viability in serum-free media is assessed
prior to secreted protein preparation either by trypane blue exclusion method [12,15] or by
assessing the presence of the most abundant cytosolic proteins in the culture media [14].
Due to the high concentration of serum protein (5–10%) supplemented in the culture media
prior to starvation; serum-free medium is not always completely free of serum [14]. Hence,
cells easily tolerate the transition from serum supplemented to serum-free media and the
expression of stress proteins is minimized. The incubation time in serum-free media depends
on each cell type and growth rate (Table 1).

Cancer Chemother Pharmacol. Author manuscript; available in PMC 2010 April 22.
Mbeunkui and Johann Page 4

Once cell growth conditions and incubation times in serum-free media are optimized, the
conditioned media are recovered free of any floating cells and debris using various methods.
Floating cells are removed from the conditioned media by low speed centrifugation
NIH-PA Author Manuscript

[12,15,18], by filtration [14,19,20] or by combination of both methods [13,17]. The


concentration of secreted proteins in the conditioned media can go down to the ng/ml range
and to concentrate to an amount detectable by proteomic methods is problematic. Methods
such as precipitation, ultrafiltration using a molecular weigh cutoff filter, adsorption to a
resin, phase extraction and speed vacuum are commonly used.

Precipitation
Precipitation is a method widely used for the recovery of biomolecules like proteins. It is
generally induced by adding salts or organic solvents to the sample, or by altering its pH
leading to the change of protein conformation. Ammonium sulfate is a salt commonly used
for precipitation because of its high solubility and is rather inexpensive. It is an efficient
method for quantitative protein recovery but it is accompanied by loss of many proteins
[21]. This method is recommended for enzyme purification because the extraction media can
be buffered or stabilizing agents can be added to maintain the maximum enzyme activity.
The organic solvent, acetone is also frequently used for protein precipitation. It is an easy to
perform concentration and desalting method that results in a good recovery. However, not
all proteins precipitate and non-proteins like DNA/ RNA and glycans also precipitate,
therefore the pellet is sometimes hard to resolubilize. This shortcoming can be overcome by
NIH-PA Author Manuscript

combining trichloroacetic acid (TCA) and acetone precipitation in which proteins are first
precipitated with TCA and traces of TCA removed with acetone [21].

Ultrafiltration and adsorption


Ultrafiltration is a membrane filtration process that separates particles based on their
molecular weight, while adsorption is a more convenient method for salt-free concentration
of small quantities of proteins for mass spectrometry and 2-DE analyses. Ultrafiltration is
extensively used for secreted proteins concentration [12,19,22] because salts and low
molecular weigh particles have been removed. It performs well in terms of purity and
protein quantity but it is labor demanding to get the concentrated volume. The filters are
always blocked and low molecular weigh proteins and polypeptides are lost. Adsorption is
valuable for the concentration of large volume of conditioned media from cell grown in
vitro. Solid-phase extraction (SPE) is the most cost-effective and versatile method for
protein concentration from plasma, urine or conditioned media. The range of chemistries for
SPE includes reversed phase, normal phase, ion exchange and other sorbents for special
applications.
NIH-PA Author Manuscript

Following the methods described above for secreted protein preparation, low molecular
weigh peptides are removed. Even if they are still present in the preparation, they are
dismissed as noise, biological trash or too small and unstable to be biologically relevant
[23]. The analysis of this class of peptides called peptidome is a rich source of cancer-
specific diagnostic information because it is a recording of the extracellular enzymatic
events taking place in the tumor microenvironment. Peptidomics analysis may lead to the
identification of the parent protein, the determination of the fragment sizes and cleavage
ends, posttranslational modifications, the quantification of the peptide and the nature of the
carrier protein in which it is bound. It is a relatively new field in proteomics analysis and
secreted biomarkers discovery; and hopefully it should be more sensitive and specific than
conventional approaches [24].

Cancer Chemother Pharmacol. Author manuscript; available in PMC 2010 April 22.
Mbeunkui and Johann Page 5

Heavy isotope labeling


An alternative method for secreted protein preparation without worry about contamination
from dead cells is to metabolically label proteins synthesized during a limited period of
NIH-PA Author Manuscript

incubation [20]. In fact, cells are incubated in serum-free media supplemented with isotope
labeled amino acids for a short time, and then washed and re-incubated in serum-free media.
The conditioned media is then collected and secreted protein concentrated as described
above. Proteins are run on 2D gel and those detected by autoradiography are synthesized by
living cells during the period of metabolic labeling [25]. Secreted proteins are then identified
by comparison with controls. This method is indeed intelligible but it suffers from its
potential applicability in the secretome analysis. Metabolic turnover and the protein
expression time depend on each cell type and the physiological status of cells. Moreover
protein released into the medium via classical or non-classical (not signal peptide triggered
protein secretion, e.g, estimated from SecretomeP 2.0 Server) pathways need a longer
incubation time to be detected incorporating the isotopic labeled amino acids.

Methods for analyzing secreted protein


A series of separation, chromatography, electrophoresis and mass spectrometry-based
methods, are available to analyze and elucidate secreted proteins. The 2D gel electrophoresis
(2DE) has become a standard method for the analysis of the proteome in which protein
mixtures are separated according to their charge, by isoelectric focusing (IEF) in the first
NIH-PA Author Manuscript

dimension, and their molecular weight with SDS-PAGE in the second dimension. This
approach coupled to mass spectrometry, is a widely used method for differential protein
expression study in cancers [26]. However, performing 2DE involves many steps ranging
from cell culture up to the stained gel and identification of the protein spot. Moreover, the
main difficulty associated with this approach is protein precipitation at their isoelectric point
during IEF, and reproducibility.

Additionally, 2DE was conceived more than 30 years ago. This technology has been useful
for low-complexity protein mixtures but never matured into a comprehensive and accurate
proteomics technology. The introduction of high-sensitivity protein identification by MS at
first seemed to help 2D gel analysis, but subsequently revealed that the thousands of spots
seen in the gel maps are actually variants of a few hundred of the most abundant proteins.
Recently, it has also become clear that quantitation of even these proteins is far from
accurate because of spot overlap. Accordingly, “biomarkers” found by these technologies
tend to be the same regardless of the system under investigation [27].

Shotgun methods are thus a powerful alternative to gels. In these methods, proteins are
digested with proteases into a more complex peptide mixture and then analyzed by LC–MS/
NIH-PA Author Manuscript

MS. LC–MS/MS couples peptide fractionation with mass spectrometry to generate a


complete record of the proteome of fluid samples [28,29] and secreted proteins [14].

Electrospray ionization (ESI) and matrix-assisted laser desorption/ionization (MALDI), are


the common techniques used to volatilize and ionize proteins or peptides for mass
spectrometry analysis [30]. The main difference between these two techniques is the nature
state of the analytes, which is liquid for the ESI and solid for MALDI. Therefore, MALDI–
MS is generally used for the analysis of simple peptide mixtures, while ESI–MS is desired
for the analysis of complex samples. Ion trap, time-of-flight (TOF), quadrupole and Fourier
transform cyclotron (FT–MS) analyzers are the basic types of mass analyzers commonly
used in proteomics analysis.

MS technology with low resolving power, especially in the form of the so-called surface
enhanced laser desorption and ionization (SELDI) method, caught the imagination of

Cancer Chemother Pharmacol. Author manuscript; available in PMC 2010 April 22.
Mbeunkui and Johann Page 6

clinicians a few years ago. This approach involves measuring a MALDI spectrum of
proteins from the body fluid of a patient and then employs machine-learning-based
computation to differentiate disease and healthy states. However, from a mass-spectrometric
NIH-PA Author Manuscript

point of view, SELDI boils down to simple MALDI spectra of very complex mixtures and
would be expected to only yield a subset of the most abundant low-mass peptides and
protein fragments. Such species could still have proven sufficient to classify patient samples.
However, as the scientific community demanded identification of the peaks comprising the
SELDI patterns, these usually turned out to belong to non-specific proteins unlikely to be
directly associated with the disease [27].

Even though SELDI and LC–MS/MS are technically competent for secreted proteins
analysis, they remain basically qualitative approaches for secreted biomarkers discovery
because peptide peaks are identified solely as present or absent in normal and cancerous
specimens. In addition, very small quantities of many proteins or polypeptides are probably
present in body fluids and cannot routinely be detected by SELDI or LC–MS/MS, which
almost all immunoassays can detect easily. Quantitative proteomics approaches for the
identification of secreted biomarkers can be coupled to immunoassays such as the traditional
ELISA-based and Western blot approaches for validation [15]. Described technologies such
as isotope-coded affinity tagging (ICAT) [31,32] and stable isotope labeling with amino
acids in cell culture (SILAC) [33,34] can be combined with mass spectrometry approaches
to produce efficient procedures to quantitatively analyze cancer secretomes.
NIH-PA Author Manuscript

Role of secreted proteins in cancer progression


Cell–cell interactions and cell–ECM interactions are known to be important during
oncogenesis, and the characterization of associated proteins is essential to illuminating the
primary molecular mechanism of the disease [35]. Likewise, extracellular proteins related to
pre-malignancy transformation offer potential targets for early detection of cancer. The
ECM is extensively modified and remodeled by proteases either secreted by non-neoplastic
and neoplastic cells or localized at the cell surface. Thus, important changes in cell–cell and
cell–ECM interactions arise, and new warning signs are created from the cells surface.
These signals have an effect on gene expression and eventually influence critical cell
behaviors such as proliferation, survival, differentiation and motility.

Proteases expressed in the extracellular milieu, and particularly matrix metalloproteinases


can target ECM proteins and non-ECM proteins such as growth factors, cytokines, cell-
associated molecules and growth factor receptors. For that reason, the activity of these
proteases in cancer is very complex and includes tumor promoting as well as tumor
suppressive effects. The balance between proteases and protease inhibitors is a vital
NIH-PA Author Manuscript

determinant in cancer development because both an absence and an excess of proteolysis


could have a negative effect on angiogenesis [36].

At the time of diagnosis, cancer has already metastasized beyond the size of initiation in
more than 34% of breast cancer patients. Unfortunately, patient survival rates drop about
fivefold for those diagnosed with distal spread of the disease, as compared with localized
diseases [37]. The role of secreted proteins and proteases in cancer progression is very
complex and diverse. Wnt proteins, for example, are a large family of secreted glycoproteins
involved in the genesis of breast cancer [38]. Adipokines are another family of proteins
secreted by adipose tissue and their participation in the development of breast cancers, for
which obesity has been established as a risk factor, has been investigated [39,40]. Agents
targeting some secreted molecules are currently in clinical trials for cancer or have been
approved by the food and drug administration (Table 2).

Cancer Chemother Pharmacol. Author manuscript; available in PMC 2010 April 22.
Mbeunkui and Johann Page 7

Osteopontin
Osteopontin (OPN) is a secreted glycophosphoprotein that is involved in physiologic and
pathologic processes. It has been shown to play a role in various developmental processes
NIH-PA Author Manuscript

and tissue differentiation [41,42] and in wound repair [43]. OPN is over expressed in several
human cancers and has been associated with metastasis and poor prognosis in breast cancer
patients [44]. The mechanisms by which OPN can affect tumor aggressiveness are not yet
completely established, but several evidences show that it is involved in different processes
associated with malignancy such as increased cellular migratory and invasion behaviors,
increased metastasis, protection from apoptosis, and induction of tumor-associated
inflammatory cells [45-47]. Experimental studies have shown that OPN can interact with a
diverse range of factors such as cell surface receptors like integrins and CD44 [48], secreted
metalloproteinase inducer [49], and growth factor/receptor pathways [50,51]. The
interactions of OPN with these factors are evidence of its role in malignancy.

Galectin-3
The galectins consist of a family of beta-galactoside-binding proteins, characterized by their
affinity for beta-galactosides and by a conserved sequence in the carbohydrate recognition
domain that bind to the carbohydrate portion of cell surface glycoproteins or glycolipids.
Galectin-3 is a multifunctional protein that is localized and functions in the cytoplasm, cell
membrane, nucleus, and the extracellular milieu. Galectin-3 expression is related to
NIH-PA Author Manuscript

neoplastic transformation and progression toward metastasis in breast [52], colon, stomach
and thyroid [53]. Additionally, down-regulation of galectin-3 expression in human breast
and colon carcinoma resulted in a considerable decrease of the meta-static potential [54].
Consistently, galectin-3 binding protein (90 K) was found to be strongly expressed in the
aggressive cell lines (MCF10DCIS.com and MCF10CA cl. D) and not or less expressed in
the non-tumor cell line (MCF10A) of the breast cancer series MCF10 [15]. Its expression
was also significant in the secretome of three unrelated cancer cell lines, MA11 in breast
cancer, WM266-4 in melanoma and OHS in osteosarcoma [14].

Transforming growth factor-β (TGF-β)


Transforming growth factor-β is a secreted polypeptide expressed through receptor serine/
threonine kinases and the intracellular Smad. TGF-β was discovered as a secreted protein
that plays a dual role in regulating cell proliferation and eliciting normal transformation of
fibroblasts [55,56]. It is a pluripotent cytokine that is known to inhibit tumor growth at the
early stage but it can also promote advanced tumor cell invasiveness and metastasis at the
later stage [56]. TGF-β inhibits proliferation and induces apoptosis in diverse cell types, and
the accumulation of loss-of-function mutations in the TGF-β receptor or Smad genes in
various human cancers classify its pathway as a tumor suppressor in humans [57]. TGF-β
NIH-PA Author Manuscript

promotes tumor cell survival, invasiveness and metastasis by targeting fibroblasts,


myofibroblasts, and immune cells in the tumor microenvironment. The mechanisms by
which TGF-β regulates the function of numerous cells that participate in tumor progression
have been investigated [58].

Matrix metalloproteinases
The ECM is regarded as a barrier to tumor progression. Cleavage of its components by
proteases is assumed to remove the physical obstruction and allow cell migration and
invasion. The tumor microenvironment is expansively modified and remodeled by proteases
and as a result of this activity, important changes in cell–cell and cell–ECM interactions
occur and new signals are generated from the cell surface. A correlation between increased
MMP expression and a tumor cell's ability to invade neighboring tissue has been established
[59]. MMPs can target many other non-ECM proteins such as growth factors, growth factor

Cancer Chemother Pharmacol. Author manuscript; available in PMC 2010 April 22.
Mbeunkui and Johann Page 8

receptors, cytokines and cell-associated molecules. The participation of these proteases in


tumor progression is far more complex than primarily anticipated.
NIH-PA Author Manuscript

For instance, MMP-7 expressed by malignant breast epithelial cells not only can cleave
matrix components in the tumor microenvironment resulting in basement membrane
structures disruption [60], but can also cleave the cell adhesion molecule E-cadherin,
resulting in the disruption of breast epithelial cell-cell junctions [61]. In addition, MMPs are
expressed in the extracellular milieu as inactive proforms (zymogen forms) that become
activated throughout a variety of mechanisms that regularly involve collaboration among a
number of MMPs families. Thus, overexpression of these proteases documented in tumors
by immunohistochemistry does not essentially mean an increase in proteolitic activity,
because most antibodies available to detect these proteases do not distinguish between their
active and inactive forms [62].

Tissue inhibitor of metalloproteinases (TIMPs), endogenous inhibitors of MMPs, provides


control of MMP activity in vivo under normal physiological circumstances. Therefore, the
balance between MMPs and TIMPs is a critical determinant in cancer progression and
metastasis. Actually, the concept that proteases are necessary to degrade the ECM and allow
endothelial cells to invade neighboring tissues appears to be more complex than initially
predicted. In angiogenesis, both an absence and an excess of proteolytic activity could have
a negative outcome [36]. Moreover, an association between a poor prognosis in cancer
NIH-PA Author Manuscript

patients and high levels of TIMPs like TIMP-2 and plasminogen activator inhibitor-1 has
been reported in many cancer types [63]. Increasing evidence suggests that TIMP-2 might
be a multifunctional protein harboring an inhibitory and stimulatory effect on cancer
progression.

Targeting the tumor microenvironment for cancer therapy


Recent data indicate that primary dysfunction in the tumor microenvironment, in addition to
epithelial malfunction, can be critical for carcinogenesis and formation of metastasis. As
discussed earlier, a chronic inflammatory state can serve to promote cancer initiation, in a
wide variety of organs. Additionally, anti-inflammatory medications in the form of non-
steroidal anti-inflammatory (NSAIDs) or aspirin have been shown to dramatically lower the
risk of cancer for patients with certain chronic inflammatory conditions [64,65]. These
findings imply a convincing case for targeting the tumor microenvironment for cancer
therapy and possible preventive strategies.

Tumor cells are surrounded by non-tumor/stromal cells that can contribute both positive and
negative signals to the tumor. Non-tumor cells are more likely to be genetically stable
compared to tumor cells, which are known to be genetically unstable. Thus, given their
NIH-PA Author Manuscript

inherent genetic instability, tumor cells are more likely to mutate and acquire or develop
resistance to medication. Accordingly, non-tumor cells may serve as therapy targets in the
tumor microenvironment but, a drawback is the delicate balance between their tumor-
inhibitory and tumor-promotion activities, as well as the normal function of the non-cancer
related stromal cells. Ideally, therapeutic approaches should remove the cancer promoting
properties while retaining the normal physiologic role of these specialized stromal cells.

To date, there is a wealth of information about specific targets for cancer therapy in the
tumor microenvironment [10,11]. The remodeling of the ECM is arbitrated in an
orchestrated way by several families of matrix-degrading enzymes such as MMPs, proteases
of cysteine and serine, as well as endoglycosidases [66]. The inhibition of enzymes such as
cysteine proteases, cathepsins and heparanase, offers the possibility to block multiple
processes in the tumor microenvironment [67,68]. However, some MMP enzymes can also
release proteins such as tumastin, endostatin, and angiostatin which inhibit tumorigenesis

Cancer Chemother Pharmacol. Author manuscript; available in PMC 2010 April 22.
Mbeunkui and Johann Page 9

[69]. Certain criteria for instance, cytostatic and cytotoxic effects of drugs, have to be
considered when targeting the tumor microenvironment for cancer therapy. Integrins, a
family of heterodimeric receptors are also therapeutic targets in the extracellular milieu.
NIH-PA Author Manuscript

Several of them are upregulated in cancer, and are expressed on endothelial and tumor cells
[70]. β1-integrin appears to be one of the promising therapeutic targets because β1-integrin
blocking antibodies have been shown to annul cell adhesion-mediated drug resistance
(CAM-DR) [71], and during tumorigenesis ECM remodeling could contribute to CAM-DR
by changing the integrin repertoire and affecting the local ECM composition [72].

Fibroblasts are another key component of the tissue stroma and play a primary role in cancer
development and progression at all stages. Carcinoma-associated fibroblasts (CAFs) are
activated fibroblasts that are allied with malignant tumors and often express α-smooth
muscle actin as a maker. Critical functions of CAFs include: deposition of ECM, regulation
of epithelial differentiation, regulation of inflammation, and wound healing. Fibroblasts are
also an important source of ECM-degrading proteases (MMPs), therefore highlighting their
critical role in the regulation of ECM turnover. Through the secretion of growth factors (i.e.,
hepatocyte growth factor (HGF), insulin-like GF (IGF), nerve GF, EGF, FEF2, WNT1)
proliferative signals are induced within adjacent epithelial cells. Thus, fibroblasts are
involved with direct mesenchymal-epithelial cell interactions. Fibroblasts also serve roles in
immune system modulation, following tissue injury, through secretion of cytokines (IL-1)
and chemokines (monocyte chemostatic protein 1 (MCP1)) [5]. Transforming growth factor
NIH-PA Author Manuscript

β is one of the fibroblast-supplied factors involved in the suppression of epithelial cell


transformation [58]. Molecules enriched in the process of fibroblast activation such as the
fibroblast activation protein (FAP) [73], the hepatocyte growth factor [58] and cathepsin K
[74], can be good selective targets in the tumor microenvironment with some recent
evidences in preclinical models and a phase 1 clinical trial for an anti-FAP antibody [75].

Signal transducers and activators of transcription (STATs), nuclear transcription factor


(NFκB) and hypoxiainducible factor 1α (HIFIα) are attractive therapeutic targets in the
microenvironment, as they are known to be closely involved in regulating inflammation,
wound healing and angiogenesis. STATs phosphorylation, which is required for
transcriptional activity, is regulated by kinases, phosphatases and binding proteins, which
are good targets for cancer therapy [76]. STAT3 is able to increase the expression of
fibrinogen genes in lung cancer cells and fibrinogen overexpression can enhance tumor cell
growth, metastasis and invasion [77]. Moreover, the transcriptional activity of STAT3 is
upregulated during chronic inflammation in mouse lung in vivo and the STAT3 activation is
found in many human lung cancers [77].

NFκB is also an important target for chemoprevention directed at the tumor


NIH-PA Author Manuscript

microenvironment for its critical role and associated proteins role in many aspects of
inflammation [78]. New cancer drug development directed at the regulation of NFκB
function is a promising field of investigation and will certainly generate newer agents for
chemoprevention. HIF1 is a key regulator in the control of tissue homeostasis and is a key
target for cancer prevention and treatment. HIF1α subunit is actually the target for drug
development as the chemopreventive agent sulphoraphane for instance controls the HIF1
role in endothelial cells by inhibiting the expression of HIF1α and HIF1-regulated genes
[9,79].

Challenges and perspectives


The microenvironment is essential for the ultimate goal of this predator we call cancer,
which is the invasion metastasis cascade. If cancer did not spread, successful medical
management would have been achieved many years ago. The invasion metastasis cascade

Cancer Chemother Pharmacol. Author manuscript; available in PMC 2010 April 22.
Mbeunkui and Johann Page 10

consists of the following critical steps: (1) local invasion, (2) intravasation or the invasion of
blood vessels, (3) transport of the cancer cell through the blood to a distant site, (4)
extravasation or escape of the cancer cell from the blood to tissue, and (5) adaptation of the
NIH-PA Author Manuscript

relocated cancer cell to the new local environment, continued growth, and thus becoming
life threatening. The tumor microenvironment is critical in achieving these cascading steps
[7].

Thus, the microenvironment is an essential intrinsic part of the tumor itself. These findings
raise the hope that therapeutic targeting of cellular and molecular components involved in
events occurring in the tumor microenvironment might be relevant. The first step toward this
aim is the identification of molecular elements and signaling pathways in the tumor
microenvironment for each cancer type. In vitro identification of molecular components in
the tumor microenvironment without contamination from the cell cytosol has been a
challenge and techniques for enrichment and analysis have continuously been improved.
Monitoring changes of the microenvironment molecular profile through tumor progression
maybe helpful for identifying cell or protein targets that have undergone modification.
Additional questions relate to how the tumor microenvironment varies across cancer types.
Scientific studies summarized in this review illustrate how the tumor microenvironment
contributes to cancer development and progression, as well as the role of secreted proteins
and their unique technical preparations, to enhance possible biomarker discovery or drug
target elucidation.
NIH-PA Author Manuscript

References
1. World Health Organization. 2006
2. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100(1):57–70. [PubMed: 10647931]
3. Kuper H, Adami HO, Trichopoulos D. Infections as a major preventable cause of human cancer. J
Intern Med 2000;248(3):171–183. [PubMed: 10971784]
4. Coussens LM, Werb Z. Inflammation and cancer. Nature 2002;420(6917):860–867. [PubMed:
12490959]
5. Kalluri R, Zeisberg M. Fibroblasts in cancer. Nat Rev Cancer 2006;6(5):392–401. [PubMed:
16572188]
6. Liotta LA, Kohn EC. The microenvironment of the tumour-host interface. Nature 2001;411(6835):
375–379. [PubMed: 11357145]
7. Weinberg, RA. Garland Science. Taylor & Francis Group, LLC; 2007. The biology of cancer.
8. Handsley MM, Edwards DR. Metalloproteinases and their inhibitors in tumor angiogenesis. Int J
Cancer 2005;115(6):849–860. [PubMed: 15729716]
9. Albini A, Sporn MB. The tumour microenvironment as a target for chemoprevention. Nat Rev
Cancer 2007;7(2):139–147. [PubMed: 17218951]
NIH-PA Author Manuscript

10. Joyce JA. Therapeutic targeting of the tumor microenvironment. Cancer Cell 2005;7(6):513–520.
[PubMed: 15950901]
11. Mueller MM, Fusenig NE. Friends or foes—bipolar effects of the tumour stroma in cancer. Nat
Rev Cancer 2004;4(11):839–849. [PubMed: 15516957]
12. Volmer MW, Radacz Y, Hahn SA, et al. Tumor suppressor Smad4 mediates downregulation of the
anti-adhesive invasion-promoting matricellular protein SPARC: landscaping activity of Smad4 as
revealed by a “secretome” analysis. Proteomics 2004;4(5):1324–1334. [PubMed: 15188399]
13. Chevallet M, Diemer H, Van Dorssealer A, Villiers C, Rabilloud T. Toward a better analysis of
secreted proteins: the example of the myeloid cells secretome. Proteomics 2007;7(11):1757–1770.
[PubMed: 17464941]
14. Mbeunkui F, Fodstad O, Pannell LK. Secretory protein enrichment and analysis: an optimized
approach applied on cancer cell lines using 2D LC–MS/MS. J Proteome Res 2006;5(4):899–906.
[PubMed: 16602697]

Cancer Chemother Pharmacol. Author manuscript; available in PMC 2010 April 22.
Mbeunkui and Johann Page 11

15. Mbeunkui F, Metge BJ, Shevde LA, Pannell LK. Identification of differentially secreted
biomarkers using LC-MS/MS in isogenic cell lines representing a progression of breast cancer. J
Proteome Res 2007;6(8):2993–3002. [PubMed: 17608509]
NIH-PA Author Manuscript

16. Pardo M, Garcia A, Antrobus R, Blanco MJ, Dwek RA, Zitzmann N. Biomarker discovery from
uveal melanoma secretomes: identification of gp100 and cathepsin D in patient serum. J Proteome
Res 2007;6(7):2802–2811. [PubMed: 17539671]
17. Volmer MW, Stuhler K, Zapatka M, et al. Differential proteome analysis of conditioned media to
detect Smad4 regulated secreted biomarkers in colon cancer. Proteomics 2005;5(10):2587–2601.
[PubMed: 15912508]
18. Khwaja FW, Svoboda P, Reed M, Pohl J, Pyrzynska B, Van Meir EG. Proteomic identification of
the wt-p53-regulated tumor cell secretome. Oncogene 2006;25(58):7650–7661. [PubMed:
17043663]
19. Gronborg M, Kristiansen TZ, Iwahori A, et al. Biomarker discovery from pancreatic cancer
secretome using a differential proteomic approach. Mol Cell Proteomics 2006;5(1):157–171.
[PubMed: 16215274]
20. Zwickl H, Traxler E, Staettner S, et al. A novel technique to specifically analyze the secretome of
cells and tissues. Electrophoresis 2005;26(14):2779–2785. [PubMed: 15966010]
21. Jiang L, He L, Fountoulakis M. Comparison of protein precipitation methods for sample
preparation prior to proteomic analysis. J Chromatogr A 2004;1023(2):317–320. [PubMed:
14753699]
22. Schwarz K, Fiedler T, Fischer RJ, Bahl H. A standard operating procedure (SOP) for the
preparation of intra- and extra-cellular proteins of Clostridium acetobutylicum for proteome
NIH-PA Author Manuscript

analysis. J Microbiol Methods 2007;68(2):396–402. [PubMed: 17098314]


23. Diamandis EP. Point: Proteomic patterns in biological fluids: do they represent the future of cancer
diagnostics? Clin Chem 2003;49(8):1272–1275. [PubMed: 12881441]
24. Villanueva J, Shaffer DR, Philip J, et al. Differential exoprotease activities confer tumor-specific
serum peptidome patterns. J Clin Invest 2006;116(1):271–284. [PubMed: 16395409]
25. Gerner C, Vejda S, Gelbmann D, et al. Concomitant determination of absolute values of cellular
protein amounts, synthesis rates, and turnover rates by quantitative proteome profiling. Mol Cell
Proteomics 2002;1(7):528–537. [PubMed: 12239281]
26. Hanash S, Brichory F, Beer D. A proteomic approach to the identification of lung cancer markers.
Dis Markers 2001;17(4):295–300. [PubMed: 11790896]
27. Mann M, Kelleher NL. Special feature: precision proteomics: the case for high resolution and high
mass accuracy. Proc Natl Acad Sci USA. 2008
28. Gronborg M, Bunkenborg J, Kristiansen TZ, et al. Comprehensive proteomic analysis of human
pancreatic juice. J Proteome Res 2004;3(5):1042–1055. [PubMed: 15473694]
29. Kristiansen TZ, Bunkenborg J, Gronborg M, et al. A proteomic analysis of human bile. Mol Cell
Proteomics 2004;3(7):715–728. [PubMed: 15084671]
30. Fenn JB, Mann M, Meng CK, Wong SF, Whitehouse CM. Electrospray ionization for mass
NIH-PA Author Manuscript

spectrometry of large biomolecules. Science 1989;246(4926):64–71. [PubMed: 2675315]


31. Gygi SP, Rist B, Gerber SA, Turecek F, Gelb MH, Aebersold R. Quantitative analysis of complex
protein mixtures using isotope-coded affinity tags. Nat Biotechnol 1999;17(10):994–999.
[PubMed: 10504701]
32. Gygi SP, Rist B, Griffin TJ, Eng J, Aebersold R. Proteome analysis of low-abundance proteins
using multidimensional chromatography and isotope-coded affinity tags. J Proteome Res
2002;1(1):47–54. [PubMed: 12643526]
33. Amanchy R, Kalume DE, Pandey A. Stable isotope labeling with amino acids in cell culture
(SILAC) for studying dynamics of protein abundance and posttranslational modifications. Sci
STKE 2005;2005(267):l2.
34. Ong SE, Blagoev B, Kratchmarova I, et al. Stable isotope labeling by amino acids in cell culture,
SILAC, as a simple and accurate approach to expression proteomics. Mol Cell Proteomics
2002;1(5):376–386. [PubMed: 12118079]
35. DeClerck YA, Mercurio AM, Stack MS, et al. Proteases, extracellular matrix, and cancer: a
workshop of the path B study section. Am J Pathol 2004;164(4):1131–1139. [PubMed: 15039201]

Cancer Chemother Pharmacol. Author manuscript; available in PMC 2010 April 22.
Mbeunkui and Johann Page 12

36. Devy L, Blacher S, Grignet-Debrus C, et al. The pro- or antiangiogenic effect of plasminogen
activator inhibitor 1 is dose dependent. FASEB J 2002;16(2):147–154. [PubMed: 11818362]
37. Etzioni R, Urban N, Ramsey S, et al. The case for early detection. Nat Rev Cancer 2003;3(4):243–
NIH-PA Author Manuscript

252. [PubMed: 12671663]


38. Li Y, Hively WP, Varmus HE. Use of MMTV-Wnt-1 transgenic mice for studying the genetic
basis of breast cancer. Oncogene 2000;19(8):1002–1009. [PubMed: 10713683]
39. Harvie M, Hooper L, Howell AH. Central obesity and breast cancer risk: a systematic review.
Obes Rev 2003;4(3):157–173. [PubMed: 12916817]
40. Rose DP, Komninou D, Stephenson GD. Obesity, adipocytokines, and insulin resistance in breast
cancer. Obes Rev 2004;5(3):153–165. [PubMed: 15245384]
41. Nemir M, Bhattacharyya D, Li X, Singh K, Mukherjee AB, Mukherjee BB. Targeted inhibition of
osteopontin expression in the mammary gland causes abnormal morphogenesis and lactation
deficiency. J Biol Chem 2000;275(2):969–976. [PubMed: 10625634]
42. Rittling SR, Matsumoto HN, McKee MD, et al. Mice lacking osteopontin show normal
development and bone structure but display altered osteoclast formation in vitro. J Bone Miner
Res 1998;13(7):1101–1111. [PubMed: 9661074]
43. Liaw L, Birk DE, Ballas CB, Whitsitt JS, Davidson JM, Hogan BL. Altered wound healing in mice
lacking a functional osteopontin gene (spp1). J Clin Invest 1998;101(7):1468–1478. [PubMed:
9525990]
44. Rudland PS, Platt-Higgins A, El Tanani M, et al. Prognostic significance of the metastasis-
associated protein osteopontin in human breast cancer. Cancer Res 2002;62(12):3417–3427.
NIH-PA Author Manuscript

[PubMed: 12067984]
45. El Tanani MK, Campbell FC, Kurisetty V, Jin D, McCann M, Rudland PS. The regulation and role
of osteopontin in malignant transformation and cancer. Cytokine Growth Factor Rev 2006;17(6):
463–474. [PubMed: 17113338]
46. Tuck AB, Chambers AF. The role of osteopontin in breast cancer: clinical and experimental
studies. J Mammary Gland Biol Neoplasia 2001;6(4):419–429. [PubMed: 12013531]
47. Weber GF. The metastasis gene osteopontin: a candidate target for cancer therapy. Biochim
Biophys Acta 2001;1552(2):61–85. [PubMed: 11825687]
48. Mangala LS, Fok JY, Zorrilla-Calancha IR, Verma A, Mehta K. Tissue transglutaminase
expression promotes cell attachment, invasion and survival in breast cancer cells. Oncogene
2007;26(17):2459–2470. [PubMed: 17043648]
49. Mi Z, Oliver T, Guo H, Gao C, Kuo PC. Thrombin-cleaved COOH(−) terminal osteopontin
peptide binds with cyclophilin C to CD147 in murine breast cancer cells. Cancer Res 2007;67(9):
4088–4097. [PubMed: 17483319]
50. Tuck AB, Elliott BE, Hota C, Tremblay E, Chambers AF. Osteopontin-induced, integrin-
dependent migration of human mammary epithelial cells involves activation of the hepatocyte
growth factor receptor (Met). J Cell Biochem 2000;78(3):465–475. [PubMed: 10861844]
51. Tuck AB, Hota C, Wilson SM, Chambers AF. Osteopontin-induced migration of human mammary
NIH-PA Author Manuscript

epithelial cells involves activation of EGF receptor and multiple signal transduction pathways.
Oncogene 2003;22(8):1198–1205. [PubMed: 12606946]
52. Matarrese P, Fusco O, Tinari N, et al. Galectin-3 overexpression protects from apoptosis by
improving cell adhesion properties. Int J Cancer 2000;85(4):545–554. [PubMed: 10699929]
53. Inohara H, Honjo Y, Yoshii T, et al. Expression of galectin-3 in fine-needle aspirates as a
diagnostic marker differentiating benign from malignant thyroid neoplasms. Cancer 1999;85(11):
2475–2484. [PubMed: 10357421]
54. Honjo Y, Nangia-Makker P, Inohara H, Raz A. Down-regulation of galectin-3 suppresses
tumorigenicity of human breast carcinoma cells. Clin Cancer Res 2001;7(3):661–668. [PubMed:
11297262]
55. Moses HL, Branum EL, Proper JA, Robinson RA. Transforming growth factor production by
chemically transformed cells. Cancer Res 1981;41(7):2842–2848. [PubMed: 6265069]
56. Roberts AB, Wakefield LM. The two faces of transforming growth factor beta in carcinogenesis.
Proc Natl Acad Sci USA 2003;100(15):8621–8623. [PubMed: 12861075]

Cancer Chemother Pharmacol. Author manuscript; available in PMC 2010 April 22.
Mbeunkui and Johann Page 13

57. Levy L, Hill CS. Alterations in components of the TGF-beta superfamily signaling pathways in
human cancer. Cytokine Growth Factor Rev 2006;17(1–2):41–58. [PubMed: 16310402]
58. Bhowmick NA, Chytil A, Plieth D, et al. TGF-beta signaling in fibroblasts modulates the
NIH-PA Author Manuscript

oncogenic potential of adjacent epithelia. Science 2004;303(5659):848–851. [PubMed: 14764882]


59. Nakajima M, Welch DR, Belloni PN, Nicolson GL. Degradation of basement membrane type IV
collagen and lung subendothelial matrix by rat mammary adenocarcinoma cell clones of differing
metastatic potentials. Cancer Res 1987;47(18):4869–4876. [PubMed: 3621180]
60. Fingleton B, Vargo-Gogola T, Crawford HC, Matrisian LM. Matrilysin [MMP-7] expression
selects for cells with reduced sensitivity to apoptosis. Neoplasia 2001;3(6):459–468. [PubMed:
11774028]
61. Noe V, Fingleton B, Jacobs K, et al. Release of an invasion promoter E-cadherin fragment by
matrilysin and stromelysin-1. J Cell Sci 2001;114(Pt 1):111–118. [PubMed: 11112695]
62. Coussens LM, Fingleton B, Matrisian LM. Matrix metalloproteinase inhibitors and cancer: trials
and tribulations. Science 2002;295(5564):2387–2392. [PubMed: 11923519]
63. Remacle A, McCarthy K, Noel A, et al. High levels of TIMP-2 correlate with adverse prognosis in
breast cancer. Int J Cancer 2000;89(2):118–121. [PubMed: 10756061]
64. Garcia-Rodriguez LA, Huerta-Alvarez C. Reduced risk of colorectal cancer among long-term users
of aspirin and nonaspirin nonsteroidal antiinflammatory drugs. Epidemiology 2001;12(1):88–93.
[PubMed: 11138826]
65. Koki AT, Masferrer JL. Celecoxib: a specific COX-2 inhibitor with anticancer properties. Cancer
Control 2002;9(2 Suppl):28–35. [PubMed: 11965228]
NIH-PA Author Manuscript

66. Carmeliet P. Angiogenesis in health and disease. Nat Med 2003;9(6):653–660. [PubMed:
12778163]
67. Joyce JA, Baruch A, Chehade K, et al. Cathepsin cysteine proteases are effectors of invasive
growth and angiogenesis during multistage tumorigenesis. Cancer Cell 2004;5(5):443–453.
[PubMed: 15144952]
68. Joyce JA, Freeman C, Meyer-Morse N, Parish CR, Hanahan D. A functional heparan sulfate
mimetic implicates both heparanase and heparan sulfate in tumor angiogenesis and invasion in a
mouse model of multistage cancer. Oncogene 2005;24(25):4037–4051. [PubMed: 15806157]
69. Hamano Y, Zeisberg M, Sugimoto H, et al. Physiological levels of tumstatin, a fragment of
collagen IV alpha3 chain, are generated by MMP-9 proteolysis and suppress angiogenesis via
alphaV beta3 integrin. Cancer Cell 2003;3(6):589–601. [PubMed: 12842087]
70. Guo W, Giancotti FG. Integrin signalling during tumour progression. Nat Rev Mol Cell Biol
2004;5(10):816–826. [PubMed: 15459662]
71. Sethi T, Rintoul RC, Moore SM, et al. Extracellular matrix proteins protect small cell lung cancer
cells against apoptosis: a mechanism for small cell lung cancer growth and drug resistance in vivo.
Nat Med 1999;5(6):662–668. [PubMed: 10371505]
72. Morin PJ. Drug resistance and the microenvironment: nature and nurture. Drug Resist Updat
2003;6(4):169–172. [PubMed: 12962682]
NIH-PA Author Manuscript

73. Park JE, Lenter MC, Zimmermann RN, Garin-Chesa P, Old LJ, Rettig WJ. Fibroblast activation
protein, a dual specificity serine protease expressed in reactive human tumor stromal fibroblasts. J
Biol Chem 1999;274(51):36505–36512. [PubMed: 10593948]
74. Allinen M, Beroukhim R, Cai L, et al. Molecular characterization of the tumor microenvironment
in breast cancer. Cancer Cell 2004;6(1):17–32. [PubMed: 15261139]
75. Scott AM, Wiseman G, Welt S, et al. A Phase I dose-escalation study of sibrotuzumab in patients
with advanced or meta-static fibroblast activation protein-positive cancer. Clin Cancer Res
2003;9(5):1639–1647. [PubMed: 12738716]
76. Yu H, Jove R. The STATs of cancer-new molecular targets come of age. Nat Rev Cancer
2004;4(2):97–105. [PubMed: 14964307]
77. Dauer DJ, Ferraro B, Song L, et al. Stat3 regulates genes common to both wound healing and
cancer. Oncogene 2005;24(21):3397–3408. [PubMed: 15735721]
78. Karin M. NF-kappaB and cancer: mechanisms and targets. Mol Carcinog 2006;45(6):355–361.
[PubMed: 16673382]

Cancer Chemother Pharmacol. Author manuscript; available in PMC 2010 April 22.
Mbeunkui and Johann Page 14

79. Bertl E, Bartsch H, Gerhauser C. Inhibition of angiogenesis and endothelial cell functions are
novel sulforaphane-mediated mechanisms in chemoprevention. Mol Cancer Ther 2006;5(3):575–
585. [PubMed: 16546971]
NIH-PA Author Manuscript

80. Martin DB, Gifford DR, Wright ME, et al. Quantitative proteomic analysis of proteins released by
neoplastic prostate epithelium. Cancer Res 2004;64(1):347–355. [PubMed: 14729644]
81. Kerbel R, Folkman J. Clinical translation of angiogenesis inhibitors. Nat Rev Cancer 2002;2(10):
727–739. [PubMed: 12360276]
82. Young SL, Chaplin DJ. Combretastatin A4 phosphate: background and current clinical status.
Expert Opin Investig Drugs 2004;13(9):1171–1182.
83. Jimeno A, Carducci M. Atrasentan: targeting the endothelin axis in prostate cancer. Expert Opin
Investig Drugs 2004;13(12):1631–1640.
84. Mooberry SL. New insights into 2-methoxyestradiol, a promising antiangiogenic and antitumor
agent. Curr Opin Oncol 2003;15(6):425–430. [PubMed: 14624224]
85. Jin H, Varner J. Integrins: roles in cancer development and as treatment targets. Br J Cancer
2004;90(3):561–565. [PubMed: 14760364]
86. Gutheil JC, Campbell TN, Pierce PR, et al. Targeted anti-angiogenic therapy for cancer using
Vitaxin: a humanized monoclonal antibody to the integrin alphavbeta3. Clin Cancer Res
2000;6(8):3056–3061. [PubMed: 10955784]
87. Burke PA, DeNardo SJ, Miers LA, Lamborn KR, Matzku S, DeNardo GL. Cilengitide targeting of
alpha(v)beta(3) integrin receptor synergizes with radioimmunotherapy to increase efficacy and
apoptosis in breast cancer xenografts. Cancer Res 2002;62(15):4263–4272. [PubMed: 12154028]
NIH-PA Author Manuscript

88. Gingras D, Boivin D, Deckers C, Gendron S, Barthomeuf C, Beliveau R. Neovastat-a novel


antiangiogenic drug for cancer therapy. Anticancer Drugs 2003;14(2):91–96. [PubMed: 12569294]
89. Bartlett JB, Dredge K, Dalgleish AG. The evolution of thalidomide and its IMiD derivatives as
anticancer agents. Nat Rev Cancer 2004;4(4):314–322. [PubMed: 15057291]
90. Sleijfer S, Kruit WH, Stoter G. Thalidomide in solid tumours: the resurrection of an old drug. Eur J
Cancer 2004;40(16):2377–2382. [PubMed: 15519508]
91. Druker BJ. Imatinib as a paradigm of targeted therapies. Adv Cancer Res 2004;91:1–30. [PubMed:
15327887]
92. Pietras K, Sjoblom T, Rubin K, Heldin CH, Ostman A. PDGF receptors as cancer drug targets.
Cancer Cell 2003;3(5):439–443. [PubMed: 12781361]
93. Graff JR, McNulty AM, Hanna KR, et al. The protein kinase Cbeta-selective inhibitor, Enzastaurin
(LY317615.HCl), suppresses signaling through the AKT pathway, induces apoptosis, and
suppresses growth of human colon cancer and glioblastoma xenografts. Cancer Res 2005;65(16):
7462–7469. [PubMed: 16103100]
94. Mendel DB, Laird AD, Xin X, et al. In vivo antitumor activity of SU11248, a novel tyrosine kinase
inhibitor targeting vascular endothelial growth factor and platelet-derived growth factor receptors:
determination of a pharmacokinetic/pharmacodynamic relationship. Clin Cancer Res 2003;9(1):
327–337. [PubMed: 12538485]
NIH-PA Author Manuscript

95. Pietras K, Hanahan D. A multitargeted, metronomic, and maximum-tolerated dose “chemo-switch”


regimen is antiangiogenic, producing objective responses and survival benefit in a mouse model of
cancer. J Clin Oncol 2005;23(5):939–952. [PubMed: 15557593]
96. Ferrara N, Hillan KJ, Gerber HP, Novotny W. Discovery and development of bevacizumab, an
anti-VEGF antibody for treating cancer. Nat Rev Drug Discov 2004;3(5):391–400. [PubMed:
15136787]
97. Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus irinotecan, fluorouracil, and
leucovorin for metastatic colorectal cancer. N Engl J Med 2004;350(23):2335–2342. [PubMed:
15175435]
98. Holash J, Davis S, Papadopoulos N, et al. VEGF-Trap: a VEGF blocker with potent antitumor
effects. Proc Natl Acad Sci USA 2002;99(17):11393–11398. [PubMed: 12177445]
99. Manley PW, Bold G, Bruggen J, et al. Advances in the structural biology, design and clinical
development of VEGF-R kinase inhibitors for the treatment of angiogenesis. Biochim Biophys
Acta 2004;1697(1–2):17–27. [PubMed: 15023347]

Cancer Chemother Pharmacol. Author manuscript; available in PMC 2010 April 22.
Mbeunkui and Johann Page 15

100. Morgan B, Thomas AL, Drevs J, et al. Dynamic contrast-enhanced magnetic resonance imaging
as a biomarker for the pharmacological response of PTK787/ZK 222584, an inhibitor of the
vascular endothelial growth factor receptor tyrosine kinases, in patients with advanced colorectal
NIH-PA Author Manuscript

cancer and liver metastases: results from two phase I studies. J Clin Oncol 2003;21(21):3955–
3964. [PubMed: 14517187]
101. Wedge SR, Ogilvie DJ, Dukes M, et al. ZD6474 inhibits vascular endothelial growth factor
signaling, angiogenesis, and tumor growth following oral administration. Cancer Res
2002;62(16):4645–4655. [PubMed: 12183421]
NIH-PA Author Manuscript
NIH-PA Author Manuscript

Cancer Chemother Pharmacol. Author manuscript; available in PMC 2010 April 22.
Mbeunkui and Johann Page 16
NIH-PA Author Manuscript
NIH-PA Author Manuscript
NIH-PA Author Manuscript

Fig. 1. Carcinogenesis
a A well-differentiated stratified epithelium is separated from the stromal compartment by a
well delineated basement membrane. The stromal compartment essentially consists of
fibroblasts, leukocytes (monocytes and macrophages), collagen bundles and mature blood
vessels. b During carcinogenesis, both epithelial and stromal components initially undergo
modifications that promote epithelial cell propagation and mutation. Fibroblast becomes
activated while the number of macrophages increases. Transient angiogenesis occurs with
new vasculature similar to the one with normal epithelia. c Proliferation of epithelial cells
with the development of an activated stroma is associated with the progression to carcinoma.
Hence, extracellular-matrix components such as collagen are degraded, the number of

Cancer Chemother Pharmacol. Author manuscript; available in PMC 2010 April 22.
Mbeunkui and Johann Page 17

inflammatory cells increases and fibroblasts differentiate to myofibroblasts resulting in the


expression of growth factors, matrix components and degrading proteases. Angiogenesis is
maintained resulting in the development of new blood vessels. Cells and molecular
NIH-PA Author Manuscript

components of the microenvironment are targets for chemopreventive or therapeutic agents


at all steps shown
NIH-PA Author Manuscript
NIH-PA Author Manuscript

Cancer Chemother Pharmacol. Author manuscript; available in PMC 2010 April 22.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

Table 1
Preparation and analysis of secreted proteins

Mediuma Incubation Conf.b Protein Protein concentrationd Analyzing method Cell type References
time (%) Recoveryc
(hours)

Argine and lysine-free 24 80 Filtration MWCO filter SILAC-SDS-PAGE-LC-MS/MS Pancreatic duct epithelial ceUs Gronborg et al.
(HPDE) [19]
Mbeunkui and Johann

Serum-free Pancreatic ductal adenocarcinoma cells


(Panl)
Methionine and cysteine-free 2 NA Filtration Precipitation (ethanol) LC-MS/MS Hepathocarcinoma cells (HepG2) Zwickl et al.
[20]
Serum and phenol-free 24–18 NA Centrifugation Ultrafiltration 2DE-MALDI/MS Murine myeloid cells (J774) Chevallet et al.
[13]
Adsorption 2DE-LC-MS/MS
Phase extraction (phenol)
Precipitation (TCA)
Serum and phenol-free 72 75 Centrifugation MWCO filter ICAT-LC-MS/MS Prostate cancer cells (LNCaP) Martin et al.
[80]
Serum-free 48 60–70 Centrifugation MWCO filter 2DE-MALDI/MS Colon carcinoma cells (SW480) Volmer et al.
[12]
Filtration
Serum-free 48 NA Centrifugation Precipitation (TCA) 2DE-MALDI/MS Glioblastoma cells (LN-Z308) Khwaja et al.
[18]
ICAT-2DE-LC-MS/MS
Serum-free 36 60-70 Centrifugation MWCO filter SDS-PAGE-LC-MS/MS Melanoma cells (UM) Pardo et al.
[16]
Filtration Precipitation (TCA/acetone)
Serum-free 30 60-70 Filtration SPE (tC2) LC-MS/MS Melanoma cells (WM 266-4) Mbeunkui et al.
[14]

Cancer Chemother Pharmacol. Author manuscript; available in PMC 2010 April 22.
Speed vacuum Osteosarcoma cells (OHS)
Breast cancer cells (MA11)
Serum-free 16 60 Centrifugation SPE (tC2) LC-MS/MS Breast cancer cells (MCF10A, Mbeunkui et al.
MCF10AT, MCF10 DCIS.com, [15]
MCF10CA cl. D)
Filtration Speed vacuum SDS-PAGE-Immunoassay Human tumor xenografts (MT-1)
MWCO filter

a
Modification of cell growth medium. Components added or removed from the medium
Page 18
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
b
Cell confluence in the growth medium prior to harvesting
c
Methods for recovering secreted proteins free of floating or dead cells
d
Methods for concentrating proteins from the growth medium
Mbeunkui and Johann

Cancer Chemother Pharmacol. Author manuscript; available in PMC 2010 April 22.
Page 19
Mbeunkui and Johann Page 20

Table 2
Therapeutic targeted molecules in the tumor microenvironment
NIH-PA Author Manuscript

Molecular target Drug name Clinical triala References

Basic Wbroblast growth factor Interferon-alpha II and III Kerbel and Folkman [81]
Endothelial microtubules Combretastatin A4 phosphate II Young and Chaplin [82]
Endothelin A receptor Atrasentan II Jimeno and Carducci [83]
Fibroblast activation protein Sibrotuzumab I Scott et al. [75]
Hypoxia-inducible factor 1α 2-Methoxyestradiol (2-ME2) I Mooberry [84]
Integrin α5β1 Volociximab (M200) II Jin and Varner [85]
Integrin ανβ3 Vitaxin II Gutheil et al. [86]; Jin and Varner [85]
Integrin ανβ3 and ανβ5 Cilengitide (EMD121974) I and II Burke et al. [87]
MMP-2, MMP-9 and MMP-12 Neovastat (AE-941) III Gingras et al. [88]
NFκB, TNFα, IL-6 and VEGF Thalidomid (Thalidomide) III Bartlett et al. [89], Sleijfer et al. [90]
NFκB, TNFα, IL-6 and VEGF CC-5013 (Revimid) II and III Bartlett et al. [89], Sleijfer et al. [90]
PDGFR, c-Kit and Tyrosine kinases Gleevec (Imatinib Mesylate) FDA-approved Druker et al. [91], Pietras et al. [92]
Protein kinase cβ Enzastaurin (LY-317615) II GraV et al. [93]
NIH-PA Author Manuscript

Tyrosine kinase, VEGFR, and CSF-1R SU11248 II Mendel et al. [94], Pietras and Hanahan
[95]
Vascular endothelial growth factor Avastin (Bevacizumab) FDA-approved Ferrara et al. [96], Hurwitz et al. [97]
Vascular endothelial growth factor Neovastat (AE-941) III Gingras et al. [88]
Vascular endothelial growth factor A VEGF-Trap I and II Holash et al. [98]
Vascular endothelial growth factor receptor Vatalanib (PTK787/ZK 222584) II and III Manley et al. [99]; Morgan et al. [100]
Vascular endothelial growth factor receptor Vandetanib (ZD6474) I and II Wedge et al. [101]
2

MMP matrix metalloproteinase, VEGF vascular endothelial growth factor, VEGFR vascular endothelial growth factor receptor, NFκB nuclear
factor of κB, TNF tumor necrosis factor, IL interleukin, PDGFR platelet-derived growth factor receptor, CSF-1R colony-stimulating factor receptor
a
Phase of therapeutic agents in clinical trial or approved by FDA
NIH-PA Author Manuscript

Cancer Chemother Pharmacol. Author manuscript; available in PMC 2010 April 22.

You might also like