Next Article in Journal
Plasticity of the 340-Loop in Influenza Neuraminidase Offers New Insight for Antiviral Drug Development
Previous Article in Journal
Tuning of Molecular Electrostatic Potential Enables Efficient Charge Transport in Crystalline Azaacenes: A Computational Study
Previous Article in Special Issue
Pancreatic Cancer and Its Microenvironment—Recent Advances and Current Controversies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

CD36 and CD97 in Pancreatic Cancer versus Other Malignancies

by
Cristiana Tanase
1,2,*,
Ancuta-Augustina Gheorghisan-Galateanu
3,4,
Ionela Daniela Popescu
1,
Simona Mihai
1,
Elena Codrici
1,
Radu Albulescu
1,5 and
Mihail Eugen Hinescu
1,3
1
Victor Babeș National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania
2
Faculty of Medicine, Titu Maiorescu University, 001863 Bucharest, Romania
3
Department of Cellular and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 8 Eroilor Sanitari Str., 050474 Bucharest, Romania
4
‘C.I. Parhon’ National Institute of Endocrinology, 001863 Bucharest, Romania
5
National Institute for Chemical Pharmaceutical R&D, 001863 Bucharest, Romania
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(16), 5656; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21165656
Submission received: 23 July 2020 / Revised: 31 July 2020 / Accepted: 4 August 2020 / Published: 6 August 2020

Abstract

:
Starting from the recent identification of CD36 and CD97 as a novel marker combination of fibroblast quiescence in lung during fibrosis, we aimed to survey the literature in search for facts about the separate (or concomitant) expression of clusters of differentiation CD36 and CD97 in either tumor- or pancreatic-cancer-associated cells. Here, we provide an account of the current knowledge on the diversity of the cellular functions of CD36 and CD97 and explore their potential (common) contributions to key cellular events in oncogenesis or metastasis development. Emphasis is placed on quiescence as an underexplored mechanism and/or potential target in therapy. Furthermore, we discuss intricate signaling mechanisms and networks involving CD36 and CD97 that may regulate different subpopulations of tumor-associated cells, such as cancer-associated fibroblasts, adipocyte-associated fibroblasts, tumor-associated macrophages, or neutrophils, during aggressive pancreatic cancer. The coexistence of quiescence and activated states in cancer-associated cell subtypes during pancreatic cancer should be better documented, in different histological forms. Remodeling of the local microenvironment may also change the balance between growth and dormant state. Taking advantage of the reported data in different other tissue types, we explore the possibility to induce quiescence (similar to that observed in normal cells), as a therapeutic option to delay the currently observed clinical outcome.

1. Introduction

Pancreatic cancer is a very lethal disease, with a five-year survival rate of 8% and very slow advances in its treatment [1,2]. Pancreatic ductal adenocarcinoma (PDAC) represents the seventh leading cause of cancer-related deaths worldwide. It is characterized by a high mortality rate, largely because of late diagnosis, early metastasis, and limited reaction to chemotherapy or radiotherapy. Unfortunately, most of the patients with pancreatic cancer fail to develop important symptoms before reaching the advanced stage of the disease. Moreover, the CA19-9 antigen test, currently in use, is not sufficient to diagnose pancreatic cancer with high sensitivity and specificity [3]. According to GLOBOCAN data, 458,918 new cases of pancreatic cancer and 432,242 new deaths were recorded in 2018. By 2040, 355,317 new cases are estimated to occur. The five-year survival rate still stands at 9%. By 2030, pancreatic cancer is projected to be the third cause of cancer-related death. The main therapeutic approach for this malignancy is surgical resection, followed by adjuvant chemotherapy that includes 5-fluorouracil/leucovorin with irinotecan and oxaliplatin (FOLFIRINOX) and gemcitabine/nab-paclitaxel. However, the development of chemoresistance among PDAC patients leads to poor clinical outcomes. Studies focused on this disease suggested that PDAC chemoresistance is a result of the interaction between pancreatic cancer cells, cancer stem cells, and the tumor microenvironment [4,5,6,7,8,9].
Several risk factors have been associated with PDAC, including tobacco smoking, diabetes mellitus, obesity, dietary factors, alcohol abuse, age, ethnicity, family history and genetic factors, Helicobacter pylori infection, belonging to non-0 blood group, and chronic pancreatitis [6,10,11,12]. The inflammation and immunosuppression caused by microbiome changes are other factors involved in the development of PDAC, and they are able to affect the metabolism of chemotherapy [13].
Besides PDAC, which represents the most fatal tumoral disease of the pancreas (covering about 90% of the total cases), several other cancers are present in the pancreatic environment. For PDAC, there is the need of molecular subtyping, thus advancing the need of a framework of molecular taxonomy. Several ductal lesions are considered tumor precursors, and a standard was adopted recently for the classification of pancreatic intraepithelial neoplasia (panIN). Molecular investigation demonstrated that PanIN-2 and -3 represent distinct steps toward invasive carcinoma. Several advances were made in further immunocytochemical and molecular characterization of other pancreatic neoplasms—mucinous noncystic carcinoma, undifferentiated mucinous cystic neoplasm, intraductal papillary mucinous neoplasm, medullary carcinoma, and other rare tumors of the pancreas [14] (see Figure 1).
In this review, we examine the recent literature, in order to explore the hypothesis that the induction of quiescence in pancreatic cancer, either in tumor cells or in tumor-associated cells, could be a putative valid therapeutic strategy. The recent recognition (in other types of tissues) of CD36 and CD97 as markers of quiescence compelled us to examine if our hypothesis could be supported by experimental facts available in the literature.

2. CD36 in Pancreatic Cancer vs. CD36 in Normal Tissues: Where Do We Stand?

2.1. CD36 in Normal Tissues

CD36, a scavenger receptor class B type 2 (SR-B2), is a transmembrane glycoprotein that is expressed on the cell surface in multiple cell types, including dendritic cells, microvascular endothelial cells (MVECs), retinal epithelial cells, platelets, monocytes/macrophages, erythrocytes, adipocytes, microglial cells, podocytes, skeletal muscle cells, mammary epithelial cells, taste receptor cells, hepatocytes, Kupffer cells, enterocytes, and serous ovarian epithelial cells. CD36 molecule was examined during several diseases, including cancer, where it seems to support development of metastasis.
In the pancreas, CD36 was found in the plasma membrane, as well as intracellularly and co-localized with insulin granules. CD36 activity appears important for the uptake of fatty acids (FAs) into β-cells, as well as for mediating their modulatory effects on insulin secretion [17]. In a comparative study, exploring pancreatic cancer versus normal pancreatic tissue, CD36 was found to be significantly lower in cancer than in corresponding non-tumor normal tissues [18].
Exposure to the ligand determinates CD36 to dimerize. In some membrane microdomains, such as caveolae, a special type of lipid rafts that are rich in proteins and lipids, CD36 can copolymerize with caveolin-1, suggesting the participation of the two molecules together in the activation of the signaling pathways [19].
Furthermore, CD36 may associate with other transmembrane proteins, such as integrins (β1, β2, and β5) and four-transmembrane proteins named tetraspanins (CD9 and CD81), which jointly mediate ligand binding and signal transduction [20]. CD36 intracellular domains, one single short cytoplasmic tail at each terminal (N and C), associate with members of the Src family of tyrosine kinases. A molecular interaction is most probably mediated by lipids in the context of lipid rafts [21]. Having a wide distribution in membrane-bound and cytoplasm organelles, such as mitochondria, endosomes, and endoplasmic reticulum (ER), CD36 promotes FA oxidation by itself or in cooperation with carnitine palmitoyltransferase-1 (CPT1) in mitochondria, along with maturation and ubiquitylation-mediated inactivation of CD36 in the ER [22]. CD36 can be transported to organelles and cell membrane by intracellular and extracellular vesicles.
CD36 transport to the cell membrane can be facilitated by several physiological stimuli, the most potent of which are (a) insulin—by activating the phosphatidylinositol 3-kinase (PI3K)/AKT signaling axis; (b) muscle contraction—by activating adenosine 5ʹ monophosphate-activated protein kinase (AMPK); and (c) inflammation [23,24].
CD36 is also known as a fatty acid translocase (FAT), because it imports long-chain fatty acids (LCFAs) in cells. The miscellaneous lipid and protein related ligands of CD36 contribute to its versatile functionality. Regarding the lipid-related ligands category that binds CD36, we can include LCFAs, anionic phospholipids, and oxidized lipids such as low- and high-density lipoprotein (ox-LDL and ox-HDL) and oxidized phospholipids (ox-PLs). CD36 has been involved in FAs transfer to cytosolic FA binding protein (FABPc) that mediates its passage to mitochondria. The lipid binding process described above has a major role on the cell’s energy metabolism [25].
Furthermore, CD36 has protein-related ligands with variated functions, among which we list the following: amyloid proteins, thrombospondins (TSP) 1 and 2, advanced glycation end products (AGEs), and advanced oxidation protein products (AOPPs). CD36 expresses on MVECs a specific domain for TSP-1 ligand (the CLESH domain) that generates Src-family pathway activation and promotes endothelial cells apoptosis [26,27].
Expression of CD36 on immune system cells, like dendritic cells and macrophages, promotes recognition and binding of apoptotic cells, respectively, β -amyloid peptides, AGEs, and AOPPs [28,29,30,31,32].

2.2. CD36 Promotes Tumor Metastasis in Pancreatic Cancer

Numerous studies confirmed the relationship between CD36 and metastasis. Investigation of CD36 in cancer revealed the role of CD36 in tumor metabolism, as well as in tumor immuno-editing, anti-angiogenic processes, metastasis, or therapy resistance. By associating with different ligands, CD36 is involved in cancer development [33].
Metastasis requires cellular changes related to cell-to-cell and cell-to-matrix adhesion, immune surveillance, activation of growth and survival signaling pathways, and epigenetic modifications. To be effective, these changes must occur in a time-dependent manner, modifying the cell phenotype for survival in new microenvironments [34].
CD36 contributes to the progression and metastatic potential of cancer by several mechanisms, such as activation of cancer stem cells, epithelial-to-mesenchymal transition, and chemoresistance [35,36].
CD36 can be a prognostic marker for different cancers, most often of epithelial origins, such as breast cancer, ovary cancer, prostate cancer, or hepatocellular carcinoma. It was proposed as an “early prognostic marker” for metastasis in gastric, ovary, and breast cancer; oral squamous cell carcinoma, esophageal squamous cell carcinoma, or hepatocellular carcinoma; or as “an unfavorable prognostic factor” in lung, bladder, breast, and PDAC [36,37,38]. In PDAC, the decreased expression of CD36 is associated with large tumor size and reduced survival rate, and less associated with TNM staging [18].
Out of a study on more than 2500 cases of different cancers came the confirmation of a role of CD36 in metastasis by investigating genes implicated in metabolic reconnection to aerobic glycolysis and fatty acids synthesis in metastatic vs. primary tumors. It was found that the CD36 gene appeared to be frequently amplified in metastatic tumors. Moreover, survival rates were reduced in the high-copy number group, as compared to the low copy group [39].
CD36 is expressed in tumor tissues, not only by tumor cells, but also by stromal, immune cells, and MVECs, and depends on tumor stage and cell type. Experimental data suggest that CD36 has a minor role in the initiation of the primary tumor, but its implication is significant in starting metastasis process. In tumor microvessels that support tumor development and metastasis, expression is generally downregulated. In the tumor stroma, CD36 expression is also deficient; the lower the CD36 level in the stroma, the more aggressive the tumor [40,41,42,43].
It has been shown that, in some forms of cancer, such as colon, breast, and ovarian, a low-CD36 expression in the primary tumor is associated with higher metastasis grade and poor prognosis. It was demonstrated that CD36 has significantly lower expression in pancreatic cancer cells’ lines and tumor tissues [18]. It was suggested that low expression of CD36 might reduce tumor cell adhesion to the extracellular matrix, followed by an increase of cell mobility due to decreased ability of CD36 to bind collagen [44].
Experimental data suggest that CD36 is involved only in metastasis initiation and proliferation of metastatic cells. The uncontrolled division of tumor cells requires high energy. The cellular metabolic pathway that provides the most energy is β-oxidation of FAs. Thus, a large number of FAs’ molecules via CD36 supports cancer cell proliferation [45]. It can be concluded that lipid metabolism is involved in the survival of migrated tumor cells in a new microenvironment, and increasing the expression of CD36 in these cells may be a marker that supports their proliferation.
There is evidence suggesting that, in some forms of cancer, such as hepatocellular carcinoma, fatty acid uptake through CD36 may promote cancer cell metastasis and distant proliferation [46].
The tumor metastasis-initiating cells derive from the primary tumor and contribute to seeding metastases in other organs [47]. Involvement of CD36 in the metastatic process is related to the three components of any tumor niche: the tumor cells, the stromal cells, and the endothelial cells. In cancer studies, CD36 is investigated mostly in relationship with thrombospondins (TSPs) and, to a lesser extent, with transforming growth factor-β (TGF-ß).
CD36 protein expression can be modified in metastasis via epigenetic modifications and post-transcriptional interference of non-coding RNA, as was recently suggested. As such, in certain cell types, regulation of CD36 expression involved DNA methylation or histone tails or miRNA interference [48].
CD36-induced potential of metastasis was reported to depend on lipid metabolism in cancer cells. CD36 mediates the FA uptake, key nutrients for tumor metabolism. In the case of gastric cancer, uptake of palmitic acid, mediated by CD36, was demonstrated to activate AKT phosphorylation and inhibit the degradation of glycogen synthase kinase 3β (GSK-3)/β-catenin, thus promoting metastasis [17].
Tumor-associated adipocytes provide enough fatty acids to the tumor cells and support proliferation and metastasis. For instance, gastric cancer often metastasizes in the greater omentum, rich in adipocytes. Adipocytes induce CD36 expression in metastatic ovarian tumors [40]. In the oxidation of fatty acids, the rate-limiting step is their transport to mitochondria, a process having as key enzymes CPT1 and CPT2. In human skeletal muscle cells, CD36 on mitochondria is able to bind CPT1, and upregulation of mitochondrial CD36 correlates with increased oxidation of fatty acids. In the case of oral squamous cell carcinoma cells, CD36 blockade generates intense intracellular lipid accumulation, leading to lipotoxic cell death and vicious metastasis [40,45].
Tumor-associated neutrophils are studied to a lesser extent, in pancreatic cancer; Zhang et al. found that in neuroendocrine tumors their presence predicts a poor survival [49].
Sano et al. documented a shift in immune-inflammatory microenvironment in a mouse model of PDAC, supporting the idea that tumor-stromal interaction could be a therapeutic target [50].
Apparently, even in pancreatic cancers, tumor associated neutrophils contribute to maintenance of a “permissive tumor microenvironment” [51].
The epithelial–mesenchymal transition promotes cancer-cell metastasis; a large number of studies regarding epithelial–mesenchymal transition (EMT) in all cancers were published [52,53]. Existing data suggest that EMT has a significant role in the development of the tumor budding, which contains one to five highly aggressive non-proliferating neoplastic cells at the infiltrative front of the tumor. Tumor budding becomes responsible for the invasion of the peritumoral connective tissue, and the infiltration of the lymphatic and blood vessels [54]. Thus, tumor budding is considered as an indicator of cancer invasiveness, including PDAC. Many processes are involved in EMT, including the reorganization of cell-surface and cytoskeletal proteins, low expression of E-cadherin, the activation of the zinc finger transcription factors that repress genes responsible for the epithelial phenotype (e.g., ZEB1, SNAIL, Slugand, and Twist), acquisition of mesenchymal markers (e.g., N-cadherin, Vimentin, and Fibronectin), increased production of extracellular matrix components, and changes in the expression of specific microRNAs [55]. Thus, epithelial cells lose their polarity and adhesion, acquire migratory and invasive capabilities, and become resistant to apoptosis.
In PDAC, tumor-budding cells and adjacent stromal cells showed increased levels of the E-cadherin repressors ZEB1, ZEB2, and SNAIL1. Moreover, tumor-budding cells lose expression for membrane adhesion molecule E-cadherin and β-catenin, without detectable nuclear β-catenin, and favorize tumor budding detachment from the primary tumor [56]. High expression of ZEB1 in tumor-budding and stromal cells was correlated with high peritumoral invasion. It was demonstrated that ZEB1- and ZEB2-positive stromal cells are cancer-associated fibroblasts, and it was suggested the existence of many subtypes of stromal cells phenotypically distinct [57]. A high level of miR-21 and a low level of miR-200c expression were associated with pancreatic cancers [54].
CD36 attenuates angiogenesis by binding to TSP-1 and thereby inducing apoptosis or blocking the vascular endothelial growth factor receptor 2 pathway in tumor microvascular endothelial cells [58].
Despite its anti-angiogenic action, TSP-1 might have a contrary effect suggested by promotion of metastatic behavior (increased production of cancerous emboli and enhanced adhesion of cancer cells) [59]. Activated TGFβ, released by a RFK/WXXW–TSP-1 interaction, is involved in tumor cells’ expansion mechanism by enhancing matrix production and altering expression of integrins, and it promotes upregulation of plasminogen activator, its receptor, and plasminogen activator inhibitor-1 [60,61].
In some cancers, such as breast, prostate, gastric, and lung, a partial link has been observed between the metastatic effect and the concentration of TGFβ at the tumor site.
The opportunity of using TSP-1 as an anti-angiogenic therapeutic target in cancer treatment is overshadowed by the divergent effect of TSP-1 (pro-angiogenic, on the one hand, by the TGFβ release, and the anti-angiogenic effect described above, on the other hand).

2.3. CD36—A Mediator of the Engulfment of Pancreatic Tumor Microvesicles

In the blood of patients with metastatic cancer circulate extracellular vesicles named tumor microvesicles, which could play a major role in intercellular communications and have been suggested as an early tumor-detection marker [62]. These vesicles act as carriers for various RNA species. They are different from exosomes in what concerns biogenesis, composition, and biological functions and participate in the progression of several types of cancer (prostate, colorectal, and pancreatic cancer) [63]. They transfer bioactive cargoes to both adjacent and distant sites, and they orchestrate carcinogenesis and malignant progression [64]. Microvesicles play an important role in mediating immune system response in metastasis progression and can also influence tumor cells phenotype (transition from a weak to a highly invasive phenotype) through the transfer of the epidermal growth factor receptor variant EGFRvIII [65].
This mechanism has been broadly researched in metastatic pancreatic tumors, following the trajectories taken by pancreatic tumor microvesicles in the liver microcirculation, the major site of pancreatic cancer metastasis [66]. A fraction of the tumor microvesicles present in the liver microcirculation have the ability to cross liver sinusoids endothelial layer via CD36 receptor and relocate in perivascular Ly6C2 macrophages for at most two weeks. Ly6C2 macrophages, which are different from Kupffer cells, can originate from recruited Ly6C2 patrolling monocytes or from extravasated inflammatory monocytes [67]. Thus, the microvesicles are increasingly integrated into CD36-induced premetastatic cell clusters and enhance development of liver metastasis. The persistent infiltration of perivascular macrophages with tumor microvesicles was associated with an augmented survival of extravasated tumor cells [66]. In vivo mice liver macrophages and in vitro myeloid immune cells studies confirmed the important role of CD36 in tumor microvesicles mediation [66,67]. Intravesicular cargo of microvesicles transferred to immune cells via CD36 can persist in these cells for extended time periods, and, thus, CD36 could potentially support the long-term reprogramming of cellular phenotypes relevant for tumor metastasis [66].

2.4. CD36 Can Regulate Chemoresistance in Pancreatic Cancer

In medical practice, cancer raises two major problems: early diagnosis and resistance to therapy. A possible correlation between CD36 expression and chemotherapy resistance has been studied in patients with PDAC treated with Gemcitabine. Many of them exhibited resistance to treatment after a short time, with consequently poor prognosis. The Gemcitabine resistance and the poor outcome in these patients were related to a CD36 strong expression correlated with a significant microinvasion to the venous system [38]. Moreover, patients expressing high levels of CD36 showed a worse prognosis in survival statistics. The unfavorable outcome could be explained by a more severe clinical–pathological picture, with microinvasions of the venous system also significantly correlated to CD36 expression [38].
It has been suggested that CD36 expression influences gemcitabine resistance by regulating anti-apoptosis proteins such as B-cell lymphoma-2 (Bcl-2), B-cell lymphoma extra-large (Bcl-xL), and myeloid cell leukemia-1 (Mcl-1). The results indicated that CD36 could enhance anti-apoptosis protein expression, which contributed to gemcitabine resistance by protecting cancer cells from drug-induced cell death. Thus, high CD36 expression is an unfavorable prognostic factor in PDAC [38,68].
Alone or with gemcitabine, quercetin administered orally in the diet has been reported to inhibit pancreatic cancer. Although the mechanisms have not been elucidated and the results are divergent, CD36 was suggested as a possible target for quercetin because this flavonoid promotes the cell adhesion, regulates the thrombospondin-1 activity, and increases FAs uptake and oxidation by activating glutathione transferases [69].
Based on the data presented, one may consider that there are just a few studies on CD36 expression in normal pancreatic tissue and cancer.

3. CD97 During Pancreatic Cancer vs. CD97 in Normal Tissue

3.1. Distribution and Functions of CD 97

CD97 is a member of the seven-trans-membrane subfamily of the class B G protein-coupled receptor (GPCR) group of the epidermal growth factor (EGF) and is present on the surface of lymphocytes, monocytes, macrophages, dendritic cells, granulocytes, and smooth muscle, being a dimeric glycoprotein with a 75–90 kDa intracellular domain and a 28 kDa extracellular domain [70,71]. CD97 is expressed on T-cells, but rarely on B cells. CD97 is intended to be an important part in cell adhesion, migration, and regulation of intercellular junctions. Various studies reported CD97 as being induced or upregulated, and/or biochemically modified in various malignancies, including of those of the thyroid, stomach, colon, prostate, pancreas, and brain, as compared to the normal tissues in question [72,73,74,75,76,77,78,79]. Moreover, CD97 was related to an invasive phenotype, correlated with tumor grade, invasion of the lymph node, metastatic spread, and overall prognosis [73,76,78,80]. The association of CD97 with human cancers represents an emergent subject of research in recent years.
In normal tissues, excessive CD97 expression was found only in macrophages and dendritic cells, excepting neuroglia, and some T and B cells [81].
Based on its unique expression pattern and structure, CD97 might play key roles in cellular adhesion, through connections with other proteins of the cell surface and of the extracellular matrix. In humans, three ligands for CD97 were identified. The first is CD55, also known as decay-accelerating factor, which interacts with EGF domains, a negative regulator of the complement cascade. The second is glycosaminoglycan chondroitin sulfate, which binds specifically to the large isoform(s) of CD97 and affects cell attachment [82,83]. The third category of ligands includes integrins, such as α5β1 and αvβ3, which bind a (arginine-glycine-aspartic acid) RGD motif in the stalk region of the CD97 α-chain 19 [84,85,86].
It was revealed that, during chronic inflammatory processes, soluble forms of CD97 protein were expressed in various body fluids. Based on the fact that chronic inflammation accompanies all the malignancies, it was observed that certain types of cancer (e.g., gastric cancer) display an overexpression of CD97 in comparison to normal individuals [87,88,89,90,91,92].

3.2. Expression of CD97 in Pancreatic Cancer

Discrimination between pancreatitis (PT) and PDAC represents a significant problem concerning the perioperative assessment of pancreatic tissue frozen sections.
The dynamics of the expression of CD97, CD95, and Fas-L in pancreatic tissues, using immunohistochemical evaluation, could be a potential diagnostic marker for the separation of inflammatory syndromes versus malignant neoplasms in the perioperative evaluation of pancreatic cryo-cut sections. In human pancreatic ductal adenocarcinomas, it could be a possible marker for dedifferentiation, invasiveness, or aggressive activity. This study revealed that CD97 expression was strong only in PT; its expression is weak in poorly differentiated PDAC. Since CD97 was not expressed by normal pancreatic tissue, it was concluded that CD97 could be considered a useful marker for PT and undifferentiated carcinomas. Thereby, in cryo-cut sections, CD97, CD95, and Fas-L can be used as additional markers to differentiate between PT and well-differentiated PDAC [90].
Even though CD97 and CD55 were considered to be defensive mechanisms in relation to the complement immune system, the presence of a small population that significantly expresses CD97 and CD55 appears to be correlated with a poor prognosis in some malignancies. Several studies have shown that both CD97 and CD55 play important roles in dedifferentiation, migration, invasiveness, and metastasis of tumors [71,93]. The association between expression of CD97 and CD55 in pancreatic cancer was not sufficiently investigated yet.
He et al. observed by immunohistochemistry analysis that, as the expression of CD97 and CD55 increases, a deterioration in cancer prognosis occurs, closely associated with lymph node involvement, metastasis, and vascular invasion [75].
Furthermore, Vogl et al. [94] revealed that the levels of CD97, CD274, and CD276 assessed by ELISA could serve as readily measurable prognostic or predictive markers in patients with advanced disease or metastatic breast, colon, or pancreatic cancer, being at baseline before cytotoxic treatment, and during the course of the chemotherapy, as well.
Consequently, it has been demonstrated that CD97 levels expressed considerable variability during the course of chemotherapy [94]. No correlations were found between CD97 expression, clinical infection, or C-Reactive Protein level, being hypothesized that infection could activate CD97 through upregulation of its ligand, CD55. No significant correlation was found between CD97 expression and tumor response, as well, in colorectal or pancreatic cancer.
However, CD97 levels are prognostic for overall survival, and thus they predefine the disease’s aggressiveness. Chemotherapy also affects certain cancer cell clusters which express the membranous form of the molecule CD97 [94].
In spite of the fact that CD97 was not or was vaguely expressed in the corresponding normal tissues of various analyzed tumors, its expression was positive in pancreatic ducts, the origin of progenitor cells, and for the majority of pancreatic adenocarcinomas.
Aust et al. have demonstrated the presence of CD97 in gastric, pancreatic, and esophageal tumors, revealing the implication of CD97 in the invasion of tumor cells, possibly as a differentiation-dependent or adhesion molecule. It should be noted that these studies used RT-PCR and flow cytometry for determination of CD 97 and EMR2 expression at the messenger RNA and protein levels, followed by immunohistochemical methods [89].
Given all these aspects, it remains questionable whether CD97 acts in an analogous manner in pancreatic cancer or whether CD97 exerts potential roles in the differentiation processes involving pancreatic progenitor cells.

4. Why Examine Concomitant Expression of CD36 and CD97s? (Why Bother with CD36 and CD97 in Pancreatic Cancer?)

Advances in surface proteome analysis and CD markers’ discovery might offer valuable insights concerning the metastatic niche in pancreatic cancer. There are few studies in literature that tackle the surface proteome profiling in different organs, to key cellular events in oncogenesis or metastasis development. Facts with reference to a potential common contribution of CD36 and CD97 were revealed firstly by Heinzelmann et al. [95]. They studied expression changes in different CD markers profile in lung fibrosis, with a major emphasis on specific phenotypes during fibroblast–myofibroblast activation by TGFβ, known to express αSMA (α-smooth muscle actin). Thus, it is accomplished the phenotype switch into a highly proliferative and migrating one, with impacts on extracellular matrix (ECM)-producing cell types in the lung. Moreover, it was reported the presence of a minor population exhibiting a strong expression of both CD36 and CD97 in remodeled areas of idiopathic pulmonary fibrosis tissue, but αSMA-negative (by immunofluorescent staining), suggesting that they were not activated fibroblasts, but more likely being considered as indicator of a quiescent, non-proliferative fibroblast background. It was also observed that CD36- and CD97-positive population decreased upon TGFβ stimulation and was part of a senescent population, as well, being significantly increased in high passages. The simultaneous presence of quiescent and activated fibroblasts could be mirrored by dynamic changes in surface markers; thus, different fibroblast phenotypes are characterized by various combinations of CD expression [95].
Taking into consideration that these two markers were separately described in different studies [35,53,71,79] as conferring an invasive phenotype, silencing both their expression could affect the physiopathology of the disease.
It is hypothesized that cellular senescence could negatively impact cancer development, by shaping its surroundings toward a pro-carcinogenic microenvironment, with the accumulation of mutations over time [96]. Since fibroblasts are thought to be key players in the tumor microenvironment, deciphering their cancer-specific features by their different expression in CD markers on activated fibroblasts could open new avenues in the evolving concept of cell identity [97].
Due to the unique versatility of cancer-associated fibroblasts (CAFs) (which are particularly heterogeneous and highly plastic), the distinction between a CAF and a normal one within the tumor microenvironment is considered to be functional, is and less defined by specific biomarkers expression or other features [98].
The lack of specific markers that can be used in order to identify CAF populations derives from their own heterogeneity, which reflects a similar situation encountered in cancer stem cells. Both populations express distinct markers which show great variation during the disease and are defined more with reference to a specific cell state, rather than to a distinct cell type. This may be hypothesized that CAFs could behave as a dynamic state of fibroblasts [98].
In light of these findings, the question whether CD36 and CD97 would complement each other remains open. Further studies are needed to gain insight into the concomitant impact of CD36 and CD97 on modulating fibroblasts phenotypes under different conditions [95], thus offering potential innovative therapeutic strategies to inactivate CAF and prevent aberrant tumor-stroma crosstalk in pancreatic cancer.

5. Heterogeneity of Pancreatic-Cancer-Associated Fibroblasts

5.1. Tumor Microenvironment

The unique tumor microenvironment emerges as a result of complex interactions between tumor cells, a wide range of stromal cell types, which refer to the non-malignant cells in the tumor microenvironment (mostly fibroblasts, endothelial cells, and immune cells—T cells, neutrophils, and macrophages), blood vessels, inflammatory cells, and a wide diversity of associated tissue cells. The dense extracellular matrix is present in various tumors, acting as a barrier to drug delivery, or as a nutrient supply for tumors [99,100]. A specific characteristic of pancreatic cancer resides in its tumor composition, being represented in a percentage of 90% of stroma cells, and only a minority of them are cancer cells [100,101].

5.2. Normal Fibroblast

Among the most studied cells, fibroblasts remain “enigmatic and mysterious”, particularly due to the lack of a unique/specific marker; hence, they are characterized based on their morphology, tissue position, and lack of lineage markers specific for epithelial, endothelial cells, and leukocytes.
Fibroblasts are usually quiescent/resting cells and are reversibly activated in response to tissue injury, being involved in synthesizing extracellular matrix (ECM) proteins, the production of cytokines/chemokines, the enrolling immune cells, and modifying tissue architecture, and thus participating in wound healing process [102,103].
Markers for fibroblast subtypes have been identified, including Vimentin and platelet-derived growth factor receptor-α (PDGFRα), but together with other standards, like cell site or cellular shape, α-smooth muscle actin (αSMA), and fibroblast activation protein (FAP), assigning important roles in bone and fat homeostasis [104,105,106].
In pancreatic tissue was observed a distinctive type of fibroblast—quiescent pancreatic stellate cells (PSCs), which accumulate lipid droplets of vitamin A [107]. In their activated form, PSCs become proliferative and attain an expansive secretome, expressing αSMA marker, and losing the lipid droplets [108]. It was observed that the equilibrium between quiescence and activation cells is mediated by the vitamin D receptor; in its absence, spontaneous pancreas fibrosis is generated. Moreover, other studies [109,110,111] have assigned an important role in metabolic homeostasis to these stellate cells, indicating that fibroblasts are no longer simply producers of ECM, but are involved in a complex networking with different other cell types, playing important roles in both normal tissue homeostasis and repair [106].

5.3. Cancer-Associated Fibroblasts (CAFs)

It was revealed that fibroblasts become irreversibly activated within tumors, being epigenetically modified, and represent a key player of the tumor microenvironment, having a diversity of functions, including matrix production and remodeling, extensive reciprocal signaling interactions with cancer cells, and crosstalk with infiltrating leukocytes, both metabolically activated and proliferative [106].
In clinical practice, when analyzing a tissue biopsy, CAFs cells are identified by using both exclusion and inclusion criteria. Thus, exclusion criteria are negative staining for epithelial, endothelial, and leukocyte markers; an elongated morphology; and the absence of mutations found within cancer cells. Usually, these exclusion criteria are associated with positivity for a mesenchymal marker, like vimentin (which does not exclude other mesenchymal lineages, such as pericytes or adipocytes) [106]. In conclusion, is difficult to define CAFs cells, partially because of the lack of precision in defining its specific markers.
Based on the results of several experimental studies, in which it was observed that such cells exhibit distinctive characteristics compared to normal fibroblasts, it was highlighted that CAFs are extremely heterogeneous and highly plastic than was previously believed [112,113].
CAFs play an essential role in the multistep processes of promoting tumor initiation, progression, invasion, and metastasis, having a dual action: first as a barrier to immune surveillance and drug delivery, and second by secreting survival factors [100,114,115,116,117].
Multiple mechanisms of CAFs’ activation, following cancer cells–fibroblasts contact, have been proposed, and they are summarized in Table 1.
Within the tumor microenvironment, CAFs could be activated by different signals and stimuli from other cells, including macrophages inducing granulin, which promotes the activation of a fibrotic environment [126].
It was revealed that CAFs could be initiated from different cell types: activated tissue fibroblasts, transdifferentiated epithelial cells, or pericytes; and mesenchymal progenitor cells recruited into the tumor, stem cells [127,128,129], or pluripotent-adipose-tissue-derived stromal cells [102,130].
DeFilippis et al. found that the transmembrane receptor CD36, normally expressed in all stromal cells, including disease-free fibroblasts, is drastically decreased in CAF population [131]. They noticed that fibroblasts with a low expression of CD36 produced increased amounts of collagens and fibronectin, compared to fibroblasts with high expression of CD36. Similarly, low expression of CD36 in different cell types generates abnormalities, as observed in endothelial cells, which have shown an amplified angiogenesis in preadipocytes, which were incapable of differentiating into adipocytes, and in immune cells, which exert a pro-tumorigenic state (cells in M2), instead of an anti-tumorigenic state (cells in M2). Thus, a decreased expression of CD36 in CAFs cells was associated with disease progression [131,132].

5.4. Cancer-Associated Fibroblasts (CAFs) in PDAC

In the microenvironment of pancreatic cancer, CAFs represent the most abundant and heterogeneous population of stromal cells, originating both from the tumor itself, and from its surroundings, as well, having as mainly precursors the pancreatic stellate cells (PSCs).
Remarkably, in different types of cancers were noticed different CAFs phenotypes, thus generating an extraordinary heterogeneity [127,133]. Based on their complex roles, CAFs could be divided into five categories: tumor-promoting (pCAF), tumor-retarding (rCAF), secretory (sCAF), inflammatory (iCAF), and myofibroblast (myoCAF) [102,129]. In PDAC, the most common and intensively analyzed CAF subtypes are myoCAFs and iCAFs [102,106,134].
A combination of cellular markers is used to identify typical stromal fibroblasts, such as αSMA, chondroitin sulfate proteoglycan (NG2), FAP, fibroblasts specific protein-1 (FSP-1), and PDGFR [127]. While αSMA and FAP are known to be characteristic to myoCAFs, the specific marker for iCAFs is represented by the increased expression of inflammatory cytokine IL-6 [102,135].
The shift into iCAFs or myoCAFs depends on cellular signaling that implies TGFβ and IL-1 [136]. For example, a recent analysis of squamous PDAC has underlined a potential role of p63 in the initiation of the IL-1α-driven conversion of PSCs into iCAFs [102,137].
The involvement of PSCs in the synthesis process of collagens, fibronectin, laminin, and other ECM components, is marked once PSCs are activated, as a consequence of the transdifferentiation into myofibroblasts, thus contributing to the desmoplastic reaction that worsens the outcome of anticancer therapies, being recognized as one of the main hallmarks of PDAC [138]. In addition, the presence of CAFs in distant metastases led to the hypothesis that CAFs play major roles in cancer spread [102,139].
Moreover, the CAFs’ distribution in PDAC is different according with the distance from cancer cells. Thus, CAFs expressing high levels of αSMA (myoCAF phenotype) are in proximity to neoplastic cells, while CAFs expressing higher levels of IL-6 (iCAFs phenotype) are more distantly distributed inside the tumor [134]. The two distinct phenotypes could be a result of TGFβ-mediated suppression of the IL-1 receptor, further implied in NF-κB signaling and subsequent IL-6 expression [106,136].
CAF heterogeneity raised new questions, such as whether CAF subtypes might interconvert with each other or not. CAFs isolated from a PDAC mouse model can be reversible from the αSMA-high and IL-6-producing states through modification of TGFβ and IL-1 tumor-secreted ligands, claiming for remarkable plasticity in fibroblast phenotypes [106,136].

5.5. Secretome

CAFs possess an active and dynamic secretome, releasing a multitude of soluble factors, and induce numerous phenotypes in adjacent cells [132,140]. There are various molecular mediators which exert their actions at different levels, and they can be categorized according to factors that are highlighted in Table 2.

5.6. Metabolism

The metabolites exchange between CAFs and neoplastic cells is an important path by which stromal fibroblasts are connected with cancer cells [155,156,157,158]. Autophagy in stromal fibroblasts could produce alanine, which is later used by PDAC cells, especially PSCs, and incorporated in the tricarboxylic acid cycle [155,159].
Recent studies have illustrated that glutaminase expression was increased in CAFs compared to pancreatic tumoral cells, being the reason for which CAFs were more sensitive to glutamine removal than tumoral cells [100,160]. It was also observed that CAFs secrete greater levels of glutamate and glutamine in culture, sustaining the growth of pancreatic tumoral cells, compared to normal PSCs [161]. Nevertheless, future studies are needed to elucidate the role of glutamine metabolism in fibroblasts [100].
A study on patients with PDAC, conducted by Shan et al., revealed that the loss of stromal caveolin-1 is linked with poor clinical outcomes, affecting tumor development through metabolism transformation [162]. Moreover, downregulation of caveolin-1 and up-regulation of monocarboxylate transporter 4 was detected in CAFs and tumor cells, transferring lactate into neoplastic cells to growth ATP formation [160,163].
Until now, few things are known regarding heterogeneity in CAF metabolism and how it is associated with different CAF subtypes; nevertheless, it is likely that CAFs display a wide range of metabolic profiles, depending on the different metabolites’ availability. Deciphering the mechanisms involved in heterogenous cancer cell–CAF metabolic coupling will open novel therapeutic strategies in PDAC.

5.7. Challenges to Studying Metabolic Interactions in the Tumor Microenvironment

The application of common in vitro culture systems in metabolic studies comes with many disadvantages, due to the tumor heterogeneity and stromal content. Thus, new approaches are needed; one of them could be the 3D cell culture models/organoid cultures that mimic aspects presented in neoplastic and stromal cells [164,165,166,167].
Moreover, tissue slice cultures can be used in metabolic studies, mainly because they still maintain the metabolic features and cellular diversity of tumors [100,168].
In conclusion, metabolic studies have to take advantage of using organoid systems due to their enhanced mimicking of tumor heterogeneity and nutrient availability, unravelling through aspects in the metabolism of tumor stromal cell types [100].

5.8. Targeting CAFs Could Create New Therapeutic Avenues in Pancreatic Cancer Therapy

Numerous research groups made great efforts in targeting CAFs for clinical benefit. Novel therapeutic approaches targeted mainly direct CAFs cells, through reprogramming of CAFs into a quiescent fibroblast, or tried to block the crosstalk between CAFs and adjacent cells. Although some of the results were quite promising, CAF targeting was confronting with many difficulties, mainly due to heterogeneity of CAF or the lack of CAF-specific markers, as previously emphasized [113].
Complete depletion of CAFs in GEMM of PDAC models by using CAF-related cell surface markers (selective depletion of α-SMA positive cells) generated surprisingly results, respectively, increased metastatic spread, enhanced intratumoral infiltration of immunosuppressive regulatory T cells, and reduced survival [169]. In contrary, Feig C. demonstrated that the depletion of FAP-expressing cells, by conditional ablation of FAP+ cells using diphtheria toxin led to an increase in anti-tumorigenic cytotoxic CD8+ T cells, slowing the pancreatic tumor growth [102,113,170].
Based on these findings, novel therapeutic strategies could be explored, addressing, on one hand, the reprogramming CAFs into quiescent fibroblasts, which could be achieved by using vitamin A metabolism/all-trans retinoic acid (ATRA), which switches both PSCs and CAFs into more quiescent fibroblast [171,172], or using vitamin D receptor or calcipotriol [173]. On the other hand, targeting interaction between CAFs and their surrounding microenvironment could be achieved by addressing the following signaling molecules:
-
TGFβ and interleukin signaling—blocking antibodies inhibitors;
-
NFkB and TNFα signaling, to reduce perlecan secretion;
-
Cancer cells–ECM interaction: Hedgehog signaling through IPI-926 (sonidegib and vismodegib) or hyaluronic acid through enzymes (PEGPH20) blocking antibodies inhibitors
-
Immunosuppression in the tumor microenvironment [113].

5.9. Future Directions

Considering the abovementioned aspects, we believe that further large studies are needed to explore the roles of CAFs in the development and progression of PDAC. Nevertheless, the lack of definitive markers in order to better characterize the dynamic CAF phenotype and the extreme heterogeneity of CAF and its impact on pancreatic cancer physiopathology represent research hot topics for further investigations [102]. Even less information is available about some other cancer-associated cell types.

6. Signaling Side: TGFβ, CD36, and CD97—Signaling in Pancreatic Cancer

The most frequent form of pancreatic cancer is PDAC, and there are multiple levels of modification in the cell environment, involving several most frequent extracellular molecules. An analysis using the Reactome Pathway Browser [174] revealed multiple pathways that appear deregulated in cancers, (Figure 2). Certain molecular key points outlined in pancreatic cancer, and mostly in PDAC, are connected to the TGFbeta pathway, including CD36.
The superfamily of TGFβ consists of more than forty members, including TGFβs, bone morphogenic proteins (BMPs), Activin, etc. [175,176]. The signaling pathway TGFβ/SMAD4 controls transduction from plasma membrane to nucleus and affects a large number of processes, such as proliferation, differentiation, apoptosis, migration, and cancer initiation and progression. TGFβ has dual actions toward tumorigenesis, exhibiting a suppressive role in early stages of tumor development, by inducing cell cycle arrest and apoptosis, while on later stages of tumor progression, cells become insensitive, and the secreted TGFβ enhances immunosuppression and promotes angiogenesis, invasion, and metastasis [175]. As the main mediator of the TGFβ pathway, SMAD4 plays a key role in switching TGFβ function on tumorigenesis. TGFβ/SMAD4 can be extensively regulated by other pathways, MAPK, PI3K/AKT, and WNT/β-catenin, forming a complex network [175,177,178,179].
If an ectopic overexpression of SMAD4 is induced in SMAD4-negative cells, this can bind the p21 promotor and enhance its transcription [180]. TGFβ is not able to induce p21 expression in pancreatic cell lines lacking SMAD4, resulting in out-of-control cell growth [181].
It was also found that TGFβ/SMAD4 may enhance p27 expression, while p27 inhibition using siRNA blocks cell growth arrest related to TGFβ/SMAD4 [182].
It was demonstrated that the TGFβ-inducible early response gene (TIEG) provoked apoptosis of pancreatic epithelial cells [183].

6.1. Crosstalk with Other Pathways

In the last twenty years, reports have been published on an increased number of pathways interacting with the canonical TGFβ/SMAD4. Such crosslinks appear along the whole chain of TGFβ/SMAD4 signal transduction, from the phosphorylation of SMAD2/3, formation of SMAD complex, and translocation to the nucleus.
A summary of the main interactions between the TGFβ/SMAD4 pathway and other pathways, such as MAPK (mitogen-activated protein kinase), PI3K/AKT (phosphatidylinositol-3 kinase/AKT), and WNT/β-catenin pathways, is illustrated in Figure 2.
Numerous studies proved that alteration of SMAD4 is closely associated with pancreatic cancer. In about 60% of human pancreatic cancers, the loss of heterozygosity occurs, and about 50% are presenting homozygous deletion or inactivating mutations [185,186]. The deletion of SMAD4 (both heterozygous and homozygous deletion) was initially discovered in pancreatic duct adenocarcinoma, followed by its detection in other cancers, like gastric, prostate, or colorectal cancers, albeit at reduced frequency compared to PDAC [1,2]. Another study demonstrated that SMAD4 mutation was associated with pancreatic tumor stages; the degree of inactivation was 31% in high-grade stage neoplasms (Pan IN-3), while none was found in the low-grade lesions (Pan IN-1-2) [187]. Knockout of SMAD4 by PDX1-Cre or P48 did not trigger cancer in mice [188,189], but it could facilitate tumor progression due to activation of KRASG12D [190] or inactivation of PTEN [191]. These studies suggest a tumor suppressive role of SMAD4 in progressive stages.
Of similar importance is, mainly in PDAC, the Hedgehog signaling. A schematic of the biogenesis and of the Hedgehog (Hh) “on” and “off” signaling is presented in Figure 3, which illustrates both the biogenesis of Hh and its externalization, a process involving participation of ER and Golgi (note the presence of palmitoylation and of cholesterol on the Hh molecule). The externalization is achieved by the association of Hh–Np to DISP2 and to SCUBE2, with the latter serving as an externalization factor. Without the Hh, cytosolic Gli undergo proteolytic cleavage, resulting in a form that is able to translocate to the nucleus, where it represses the transcription of target genes. Binding Hh to the cell surface receptor Patched (Ptc) stabilizes integral Gli proteins in their transcriptional activator form, thus stimulating Hh-dependent gene expression [192,193,194,195].
Hedgehog signaling has an important relationship to tumorigenesis [196,197]. Overexpression of Hh and Gli1 associates with the start of pancreatic ductal neoplasia [198,199]. In pancreatic cancer was reported a ligand-dependent activation of Hedgehog signaling, while in other cancers, genomic mutations were reported. The Sonic Hedgehog (Shh) overexpression seems to trigger the onset of pancreatic cancer [196]. Shh, along with that of Smo and Gli, is present on stromal-derived PSCs. Meanwhile, the ligands Shh and Ihh are expressed only in pancreatic tumor cells. The genetic ablation of Smo did not affect the development of PDAC tumors, suggesting the involvement of paracrine signaling [200,201]. Treating PSCs with Shh and Ihh resulted in the upregulation of the Hedgehog pathway and induced PSC proliferation. The Hedgehog signal ligands released by cancer cells induced the secretion by PCDs of cancer cells stimulating factors [202].
The involvement of both TGFβ/SMAD and of Hedgehog pathways was demonstrated in several studies regarding the oncogenesis processes [116,203], although without clear evidence of a real crosstalk between the two pathways.

6.2. Involvement of CD36 and CD 97 in Signaling Pathways

CD36 and CD97 interfere with complex signaling network, finetuning the cellular responses to this devastating disease. The ability of CD36 to bind different ligands provides its functional diversity. As a lipid translocase, CD36 can facilitate the transfer of lipid molecules, including LCFAs (171), ox-LDL (172), anionic phospholipids (173), and oxidized phospholipids (174). Membrane-bound CD36 is able to transfer fatty acids to the fatty acids binding protein from the cytosol and further transport them into mitochondria, thus providing energy to the cell. CD36 is able to bind other ligands, for instance, amyloid proteins, AOPPs, TSP-1, TSP-2, advanced glycation end products (AGEs), and AOPPs (175) and (176). TSP-1 is capable of binding to the CLESH domain on CD36, present on MVECs, followed by subsequent activation of Src pathway, thus influencing apoptosis of the endothelial cells. Acting as scavenger receptor, CD36 binds to other transduction proteins on the cell surface, like integrins and CD9 or CD91, mediating binding and transduction of signal. Acting as a modulator of the Toll-like receptors 4 and 6, CD36 may moderate the transduction of inflammatory signals, when meeting ox-LDL and exogenous stimuli [204] (see Figure 4).
Several studies connect CD36 and TSP-1 to the TGFβ1 signaling, linking this to pancreatic cancer [205], providing information on another molecule involved in the process, PAI-1, which is upregulated in pancreatic cancer cells. Other proofs of the involvement of CD36 and TSP-1 were provided for the decrease of protein expression of CD36 in colon cancer (with progression of the decrease from adenoma to carcinoma) and provided insights on the roles of CD36 as suppressor of the β-catenin/c-myc signaling by promotion of proteasome-dependent ubiquitination of GPC4 [206]. Such details did not appear so far for pancreatic cancer. However, for PC was reported that CD36 is required on immune cells, to allow extravasation of tumor microvesicles from premetastatic foci, while it seems that CD36 may also act as a tumor-suppressive protein, since it appears downregulated on PC cells [18,66].
The presence of CD97 in cancers like pancreatic, gastric esophageal, or thyroid suggests that its expression might be a common characteristic of such tumors. The results were confirmed on a number of other carcinomas [81]. The potential interaction of CD97 with CD55 situated in the extracellular matrix and its importance for tumor invasion are supported by results regarding enhanced CD97 in scattered tumor cells present in the invasion front. Such cells in the invasion front displayed a modification of expression or function of the adhesive E-cadherin–catenin complex [207,208]. Beta-catenin, which is usually expressed on the cell membrane, presented abnormal accumulation in the cell nuclei, with the process having a major role in acquiring an invasive phenotype [209]. CD97 has a distinct pattern of expression in the pancreas, where it is present in higher amounts than in other tumoral and peritumoral tissues, where it usually shows a very low presence [81,210,211]. In the pancreatic ducts, where most pancreatic adenocarcinoma originates, the pancreatic progenitor cells are CD97+. The pattern of CD97 expression in normal tissue is parallel with that of Ep-CAM. Epithelial growth by budding from ductal cells induces the upregulation of Ep-CAM, while the differentiation into endocrine cells generates a downregulation of the same molecule. This shows characteristics of a molecule acting in a morpho-regulatory and differentiation-dependent manner. However, it is still left to speculation whether CD97 is acting by a similar pattern on pancreatic progenitor cells or if it plays a more general role in the process of differentiation.
Exploration of signaling pathways, both of the TGF-beta dependent and Hedgehog and of the crosstalk with CD36 and CD97, or of these with other intracellular signaling molecules offers potential solutions for stratification of pancreatic cancers and optimization of therapies, based on particular aspects of each subset.

7. Favoring Quiescence (Cell Dormancy)—A Valid Therapeutic Strategy in Pancreatic Cancer?

Cell quiescence can be defined as a cellular state/phenotype that cells can enter or exit, acting as a switch and allowing tuning (during health or disease) of regenerative/proliferative mechanisms. Quiescence is a versatile resource cells actually exploit, be they hematopoietic stem cells [212,213], satellite cells in skeletal muscle [214], or subpopulations of neoplastic cells [215].
In this case, as a “dormant cellular state, which dictates the distinct tumorigenic aggressiveness” [216], beside mechanisms operating in normal cells, there are probably complementary mechanisms that are less-explored and possible double-edged sword.
Recently, systematic information about quiescence in different cell types has been constantly accumulating, e.g., neurons [216], hair follicle stem cells [217], muscle stem cells [217], vessels (“During quiescence, the angiogenic switch is ‘off’”) [218], vessels during diabetes [219], pancreatic stem cells [109], beta-cells during diabetes [220], mesenchymal stem cells [221], or pancreatic progenitor-like acinar cells [222].
It was found reasonable to inquire about the possibility to search (and find) factor(s) responsible for quiescence or senescence of pancreatic beta-cells (i.e., “the failure of cyclin-dependent kinases (CDKs) and cyclins to access the nucleus”), in order to reverse the clinical status by favoring exit quiescence [220].
On the opposite, one may put the question if to induce a type of quiescence similar with that observed in normal cells could be, as well, a reasonable therapeutic strategy in pancreatic cancer.
During neoplasia, a series of dis-equilibria concerning complex intricate mechanisms were revealed, and each of them was, at its turn (more or less), examined in terms of neoplastic (or metastatic) progression.
For tumor cells, the balance between quiescence and activation of different cell subtypes may favor tumor development or therapy resistance. Poles apart, from a therapeutical point of view, altering this equilibrium (favoring a quiescence similar with that operating in stem cells or neurons) could be, a priori, an effective strategy.
In another type of cancer, human mesenchymal stem cells (hMSC) were shown contribute to quiescence and therapy resistance of persistent acute myeloid leukemia (AML cells) [223]. In this study, quiescence is related to therapy resistance, and thus with an unwanted result. However, what facts are currently documented for pancreatic cancer that explicitly explore the relationship between quiescence and therapeutically outcome? As a pure speculation, inducing quiescence obviously does not cure the disease, but, again, a priori, if effective, manipulating quiescence may gain time in a disease with a dramatic evolution in a short time.
Different markers associated with quiescence were identified, in different normal and tumoral tissues. Leucine-rich repeats and immunoglobulin-like domains 1 (Lrig1) regulate cultured neural stem cell quiescence. The marker was identified in multiple organs in mice [224], but there is only a paper examining Lrig1 in pancreatic cancer [225].
Quiescence and long-term maintenance of different cell populations’ subtypes were documented in different (including pancreatic) cancer types [109,215,226]. Table 3 presents some examples of facts/or proposed mechanisms contributing to the understanding of quiescence of different pancreatic cancer cell types.
Facts (experimental findings) presented in this paper suggest that a long-term induced quiescence of specific cell types in the pancreas may turn down the evolution of pancreatic cancer, through mechanisms that are currently still controversial and speculative. However, the hypothesis deserves a more thorough examination, taking into account the slow progress in improving prognosis in this disease, despite the extensive new approaches.
The relative proportion of cell types required to block or delay progression remains to be established. One may speculate that it is possible that one single cell type reconverted to quiescence to divert evolution to a better stage, but it is also possible to achieve quiescence only when several cellular types act together in a coordinated manner.

8. Conclusions

Considering the abovementioned aspects, further large studies are needed to explore the roles and behavior of different cancer-associated cells in the development and progression of PDAC. Nevertheless, the lack of definitive markers in order to accurately characterize the dynamics of cancer-associated cells’ phenotypes, the extreme heterogeneity of these cells, and its impact on pancreatic cancer pathophysiology represent hot research topics for further investigations.
Exploration of signaling pathways, both of the TGF-beta dependent and of the crosstalk with CD36 and CD97, or with other intracellular signaling molecules, offers potential hints for identification and stratification of pancreatic cancer cell subtypes, cell cooperation in tumor microenvironment, and optimization of therapies, based on particular aspects of each subset.
Reversal or induction of a quiescence state in cancer-associated target cells deserves further exploration in pancreatic cancer, and there is an acute need of new biomarkers able to identify not only specific cell subtypes, but also states of specific cellular subtypes.
Quiescence is a cellular state that is long-lasting in normal cells, and understanding how to induce such a state in aggressively proliferating pancreatic neoplasia could, a priori, provide the means to delay evolution and improve outcome.
At least two recent observations support this view:
(a)
“Therefore, reversal of activated fibroblasts to the quiescence state is an important area of investigation that may help the therapeutic management of a number of diseases including pancreatic cancer” [227].
(b)
“Thus, targeting the CAFs at this stage with molecules that can revert the back to “quiescent” state can be considered an attractive therapeutic strategy, as this will disrupt the tumor–stroma crosstalk and inhibit the tumor growth and progression” [228].

Author Contributions

All authors contributed equally to the manuscript, as follows: conceptualization C.T., A.-A.G.-G., I.D.P., S.M., E.C., R.A., and M.E.H.; writing—original draft preparation C.T., A.-A.G.-G., I.D.P., S.M., E.C., R.A., and M.E.H.; writing—review and editing C.T., A.-A.G.-G., I.D.P., S.M., E.C., R.A., and M.E.H. All authors have read and agree to the published version of the manuscript.

Funding

This research was funded by the Romanian Ministry of Education and Research, UEFISCDI, in the framework of the following: Program 1—The Improvement of the National System of Research and Development; Subprogram 1.2—Institutional Excellence—Projects of Excellence Funding in RDI, Contract No. 7PFE/16.10.2018, PN 19.29.01.04, POC-A.1.1.4-E (2016–2020) - Dia-Cure P_37_794.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

CDcluster of differentiation
PDACpancreatic ductal adenocarcinoma
panINpancreatic intraepithelial neoplasia
SR-B2receptor class B type 2
MVECsmicrovascular endothelial cells
ERendoplasmic reticulum
FAOmitochondrial FA oxidation
CPT1carnitine palmitoyltransferase-1
PI3Kphosphatidylinositol 3-kinase
AMPK5′ adenosine monophosphate-activated protein kinase
LCFAslong-chain fatty acids
ox-LDLoxidized low-density lipoprotein
ox-HDLoxidized high-density lipoprotein
ox-PLsoxidized phospholipids
FABPccytosolic FA binding protein
TSPthrombospondins
AOPPsadvanced oxidation protein products
AGEsadvanced glycation end products
EMTepithelial–mesenchymal transition
TSP-1thrombospondin-1
TGFβtransforming growth factor-β
EGFsepidermal growth factors
PTpancreatitis
CAFcancer-associated fibroblast
ECMextracellular matrix
PDGFRαplatelet-derived growth factor receptor-α
αSMAα-smooth muscle actin
FAPfibroblast activation protein
PSCspancreatic stellate cells

References

  1. Croce, C.M. Oncogenes and cancer. N. Engl. J. Med. 2008, 358, 502–511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Chen, W.; Zheng, R.; Baade, P.D.; Zhang, S.; Zeng, H.; Bray, F.; Jemal, A.; Yu, X.Q.; He, J. Cancer statistics in China, 2015. CA Cancer J. Clin. 2016, 66, 115–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Lan, B.; Zeng, S.; Grutzmann, R.; Pilarsky, C. The Role of Exosomes in Pancreatic Cancer. Int. J. Mol. Sci. 2019, 20, 4332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Zeng, S.; Pottler, M.; Lan, B.; Grutzmann, R.; Pilarsky, C.; Yang, H. Chemoresistance in Pancreatic Cancer. Int. J. Mol. Sci. 2019, 20, 4504. [Google Scholar] [CrossRef] [Green Version]
  5. Rahib, L.; Smith, B.D.; Aizenberg, R.; Rosenzweig, A.B.; Fleshman, J.M.; Matrisian, L.M. Projecting cancer incidence and deaths to 2030: The unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014, 74, 2913–2921. [Google Scholar] [CrossRef] [Green Version]
  6. Rawla, P.; Sunkara, T.; Gaduputi, V. Epidemiology of Pancreatic Cancer: Global Trends, Etiology and Risk Factors. World J. Oncol. 2019, 10, 10–27. [Google Scholar] [CrossRef]
  7. Caravia, L.; Dudau, M.; Gherghiceanu, M.; Tanase, C.; Enciu, A.M. Could caveolae be acting as warnings of mitochondrial ageing? Mech. Ageing Dev. 2015, 146, 81–87. [Google Scholar] [CrossRef]
  8. Pistol-Tanase, C.; Raducan, E.; Dima, S.O.; Albulescu, L.; Alina, I.; Marius, P.; Cruceru, L.M.; Codorean, E.; Neagu, T.M.; Popescu, I. Assessment of soluble angiogenic markers in pancreatic cancer. Biomark. Med. 2008, 2, 447–455. [Google Scholar] [CrossRef] [Green Version]
  9. Tanase, C.P.; Neagu, M.; Albulescu, R.; Hinescu, M.E. Advances in pancreatic cancer detection. Adv. Clin. Chem. 2010, 51, 145–180. [Google Scholar] [CrossRef]
  10. Tanase, C.P.; Neagu, A.I.; Necula, L.G.; Mambet, C.; Enciu, A.M.; Calenic, B.; Cruceru, M.L.; Albulescu, R. Cancer stem cells: Involvement in pancreatic cancer pathogenesis and perspectives on cancer therapeutics. World J. Gastroenterol. 2014, 20, 10790–10801. [Google Scholar] [CrossRef]
  11. Albulescu, R.; Neagu, M.; Albulescu, L.; Tanase, C. Tissular and soluble miRNAs for diagnostic and therapy improvement in digestive tract cancers. Expert Rev. Mol. Diagn. 2011, 11, 101–120. [Google Scholar] [CrossRef] [PubMed]
  12. Dima, S.O.; Tanase, C.; Albulescu, R.; Herlea, V.; Chivu-Economescu, M.; Purnichescu-Purtan, R.; Dumitrascu, T.; Duda, D.G.; Popescu, I. An exploratory study of inflammatory cytokines as prognostic biomarkers in patients with ductal pancreatic adenocarcinoma. Pancreas 2012, 41, 1001–1007. [Google Scholar] [CrossRef]
  13. Wang, Y.; Yang, G.; You, L.; Yang, J.; Feng, M.; Qiu, J.; Zhao, F.; Liu, Y.; Cao, Z.; Zheng, L.; et al. Role of the microbiome in occurrence, development and treatment of pancreatic cancer. Mol. Cancer 2019, 18, 173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Collisson, E.A.; Bailey, P.; Chang, D.K.; Biankin, A.V. Molecular subtypes of pancreatic cancer. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 207–220. [Google Scholar] [CrossRef] [PubMed]
  15. Gordon-Dseagu, V.L.; Devesa, S.S.; Goggins, M.; Stolzenberg-Solomon, R. Pancreatic cancer incidence trends: Evidence from the Surveillance, Epidemiology and End Results (SEER) population-based data. Int. J. Epidemiol. 2018, 47, 427–439. [Google Scholar] [CrossRef]
  16. Haeberle, L.; Esposito, I. Pathology of pancreatic cancer. Transl. Gastroenterol. Hepatol. 2019, 4, 50. [Google Scholar] [CrossRef]
  17. Noushmehr, H.; D’Amico, E.; Farilla, L.; Hui, H.; Wawrowsky, K.A.; Mlynarski, W.; Doria, A.; Abumrad, N.A.; Perfetti, R. Fatty acid translocase (FAT/CD36) is localized on insulin-containing granules in human pancreatic beta-cells and mediates fatty acid effects on insulin secretion. Diabetes 2005, 54, 472–481. [Google Scholar] [CrossRef] [Green Version]
  18. Jia, S.; Zhou, L.; Shen, T.; Zhou, S.; Ding, G.; Cao, L. Down-expression of CD36 in pancreatic adenocarcinoma and its correlation with clinicopathological features and prognosis. J. Cancer 2018, 9, 578–583. [Google Scholar] [CrossRef] [Green Version]
  19. Lamaze, C.; Tardif, N.; Dewulf, M.; Vassilopoulos, S.; Blouin, C.M. The caveolae dress code: Structure and signaling. Curr. Opin. Cell Biol. 2017, 47, 117–125. [Google Scholar] [CrossRef]
  20. Pepino, M.Y.; Kuda, O.; Samovski, D.; Abumrad, N.A. Structure-function of CD36 and importance of fatty acid signal transduction in fat metabolism. Annu. Rev. Nutr. 2014, 34, 281–303. [Google Scholar] [CrossRef] [Green Version]
  21. Thorne, R.F.; Law, E.G.; Elith, C.A.; Ralston, K.J.; Bates, R.C.; Burns, G.F. The association between CD36 and Lyn protein tyrosine kinase is mediated by lipid. Biochem. Biophys. Res. Commun. 2006, 351, 51–56. [Google Scholar] [CrossRef] [PubMed]
  22. Yoshida, Y.; Jain, S.S.; McFarlan, J.T.; Snook, L.A.; Chabowski, A.; Bonen, A. Exercise-and training-induced upregulation of skeletal muscle fatty acid oxidation are not solely dependent on mitochondrial machinery and biogenesis. J. Physiol. 2013, 591, 4415–4426. [Google Scholar] [CrossRef] [PubMed]
  23. Luiken, J.J.; Koonen, D.P.; Willems, J.; Zorzano, A.; Becker, C.; Fischer, Y.; Tandon, N.N.; Van Der Vusse, G.J.; Bonen, A.; Glatz, J.F. Insulin stimulates long-chain fatty acid utilization by rat cardiac myocytes through cellular redistribution of FAT/CD36. Diabetes 2002, 51, 3113–3119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Luiken, J.J.; Coort, S.L.; Willems, J.; Coumans, W.A.; Bonen, A.; van der Vusse, G.J.; Glatz, J.F. Contraction-induced fatty acid translocase/CD36 translocation in rat cardiac myocytes is mediated through AMP-activated protein kinase signaling. Diabetes 2003, 52, 1627–1634. [Google Scholar] [CrossRef] [Green Version]
  25. Nieva, C.; Marro, M.; Santana-Codina, N.; Rao, S.; Petrov, D.; Sierra, A. The lipid phenotype of breast cancer cells characterized by Raman microspectroscopy: Towards a stratification of malignancy. PLoS ONE 2012, 7, e46456. [Google Scholar] [CrossRef]
  26. Iwao, Y.; Nakajou, K.; Nagai, R.; Kitamura, K.; Anraku, M.; Maruyama, T.; Otagiri, M. CD36 is one of important receptors promoting renal tubular injury by advanced oxidation protein products. Am. J. Physiol. Ren. Physiol. 2008, 295, F1871–F1880. [Google Scholar] [CrossRef] [Green Version]
  27. Zhu, W.; Li, W.; Silverstein, R.L. Advanced glycation end products induce a prothrombotic phenotype in mice via interaction with platelet CD36. Blood 2012, 119, 6136–6144. [Google Scholar] [CrossRef] [Green Version]
  28. Albert, M.L.; Pearce, S.F.; Francisco, L.M.; Sauter, B.; Roy, P.; Silverstein, R.L.; Bhardwaj, N. Immature dendritic cells phagocytose apoptotic cells via alphavbeta5 and CD36, and cross-present antigens to cytotoxic T lymphocytes. J. Exp. Med. 1998, 188, 1359–1368. [Google Scholar] [CrossRef]
  29. Doens, D.; Valiente, P.A.; Mfuh, A.M.; Vo, A.X.T.; Tristan, A.; Carreno, L.; Quijada, M.; Nguyen, V.T.; Perry, G.; Larionov, O.V.; et al. Identification of Inhibitors of CD36-Amyloid Beta Binding as Potential Agents for Alzheimer’s Disease. Acs Chem. Neurosci. 2017, 8, 1232–1241. [Google Scholar] [CrossRef]
  30. Pennathur, S.; Pasichnyk, K.; Bahrami, N.M.; Zeng, L.; Febbraio, M.; Yamaguchi, I.; Okamura, D.M. The macrophage phagocytic receptor CD36 promotes fibrogenic pathways on removal of apoptotic cells during chronic kidney injury. Am. J. Pathol. 2015, 185, 2232–2245. [Google Scholar] [CrossRef] [Green Version]
  31. Ping, M.; Xiao, W.; Mo, L.; Xiao, X.; Song, S.; Tang, W.; Yang, X. Paeonol attenuates advanced oxidation protein product-induced oxidative stress injury in THP-1 macrophages. Pharmacology 2014, 93, 286–295. [Google Scholar] [CrossRef] [PubMed]
  32. Ohgami, N.; Nagai, R.; Ikemoto, M.; Arai, H.; Miyazaki, A.; Hakamata, H.; Horiuchi, S.; Nakayama, H. CD36, serves as a receptor for advanced glycation endproducts (AGE). J. Diabetes Complicat. 2002, 16, 56–59. [Google Scholar] [CrossRef]
  33. Yang, P.; Su, C.; Luo, X.; Zeng, H.; Zhao, L.; Wei, L.; Zhang, X.; Varghese, Z.; Moorhead, J.F.; Chen, Y.; et al. Dietary oleic acid-induced CD36 promotes cervical cancer cell growth and metastasis via up-regulation Src/ERK pathway. Cancer Lett. 2018, 438, 76–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Enciu, A.M.; Radu, E.; Popescu, I.D.; Hinescu, M.E.; Ceafalan, L.C. Targeting CD36 as Biomarker for Metastasis Prognostic: How Far from Translation into Clinical Practice? Biomed Res. Int. 2018, 2018, 7801202. [Google Scholar] [CrossRef] [Green Version]
  35. Deng, M.; Cai, X.; Long, L.; Xie, L.; Ma, H.; Zhou, Y.; Liu, S.; Zeng, C. CD36 promotes the epithelial-mesenchymal transition and metastasis in cervical cancer by interacting with TGF-beta. J. Transl. Med. 2019, 17, 352. [Google Scholar] [CrossRef] [PubMed]
  36. Yang, G.; Addai, J.; Tian, W.H.; Frolov, A.; Wheeler, T.M.; Thompson, T.C. Reduced infiltration of class A scavenger receptor positive antigen-presenting cells is associated with prostate cancer progression. Cancer Res. 2004, 64, 2076–2082. [Google Scholar] [CrossRef] [Green Version]
  37. Kubo, M.; Eguchi, H. ASO Author Reflections: Regulation of Chemoresistance in Pancreatic Ductal Adenocarcinoma by Scavenger Receptor CD36. Ann. Surg. Oncol. 2020, 27, 620–621. [Google Scholar] [CrossRef] [PubMed]
  38. Kubo, M.; Gotoh, K.; Eguchi, H.; Kobayashi, S.; Iwagami, Y.; Tomimaru, Y.; Akita, H.; Asaoka, T.; Noda, T.; Takeda, Y.; et al. Impact of CD36 on Chemoresistance in Pancreatic Ductal Adenocarcinoma. Ann. Surg. Oncol. 2020, 27, 610–619. [Google Scholar] [CrossRef] [PubMed]
  39. Nath, A.; Chan, C. Genetic alterations in fatty acid transport and metabolism genes are associated with metastatic progression and poor prognosis of human cancers. Sci. Rep. 2016, 6, 18669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Ladanyi, A.; Mukherjee, A.; Kenny, H.A.; Johnson, A.; Mitra, A.K.; Sundaresan, S.; Nieman, K.M.; Pascual, G.; Benitah, S.A.; Montag, A.; et al. Adipocyte-induced CD36 expression drives ovarian cancer progression and metastasis. Oncogene 2018, 37, 2285–2301. [Google Scholar] [CrossRef]
  41. Pan, J.; Fan, Z.; Wang, Z.; Dai, Q.; Xiang, Z.; Yuan, F.; Yan, M.; Zhu, Z.; Liu, B.; Li, C. CD36 mediates palmitate acid-induced metastasis of gastric cancer via AKT/GSK-3beta/beta-catenin pathway. J. Exp. Clin. Cancer Res. Cr. 2019, 38, 52. [Google Scholar] [CrossRef] [Green Version]
  42. Hale, J.S.; Otvos, B.; Sinyuk, M.; Alvarado, A.G.; Hitomi, M.; Stoltz, K.; Wu, Q.; Flavahan, W.; Levison, B.; Johansen, M.L.; et al. Cancer stem cell-specific scavenger receptor CD36 drives glioblastoma progression. Stem Cells 2014, 32, 1746–1758. [Google Scholar] [CrossRef] [Green Version]
  43. Pascual, G.; Avgustinova, A.; Mejetta, S.; Martin, M.; Castellanos, A.; Attolini, C.S.; Berenguer, A.; Prats, N.; Toll, A.; Hueto, J.A.; et al. Targeting metastasis-initiating cells through the fatty acid receptor CD36. Nature 2017, 541, 41–45. [Google Scholar] [CrossRef] [PubMed]
  44. Yu, X.; Guo, C.; Fisher, P.B.; Subjeck, J.R.; Wang, X.Y. Scavenger Receptors: Emerging Roles in Cancer Biology and Immunology. Adv. Cancer Res. 2015, 128, 309–364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Li, Z.; Kang, Y. Lipid Metabolism Fuels Cancer’s Spread. Cell Metab. 2017, 25, 228–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Nath, A.; Li, I.; Roberts, L.R.; Chan, C. Elevated free fatty acid uptake via CD36 promotes epithelial-mesenchymal transition in hepatocellular carcinoma. Sci. Rep. 2015, 5, 14752. [Google Scholar] [CrossRef] [Green Version]
  47. Celia-Terrassa, T.; Kang, Y. Distinctive properties of metastasis-initiating cells. Genes Dev. 2016, 30, 892–908. [Google Scholar] [CrossRef] [Green Version]
  48. Niculite, C.M.; Enciu, A.M.; Hinescu, M.E. CD 36: Focus on Epigenetic and Post-Transcriptional Regulation. Front. Genet. 2019, 10, 680. [Google Scholar] [CrossRef]
  49. Zhang, W.H.; Wang, W.Q.; Gao, H.L.; Xu, S.S.; Li, S.; Li, T.J.; Han, X.; Xu, H.X.; Li, H.; Jiang, W.; et al. Tumor-Infiltrating Neutrophils Predict Poor Survival of Non-Functional Pancreatic Neuroendocrine Tumor. J. Clin. Endocrinol. Metab. 2020, 105. [Google Scholar] [CrossRef]
  50. Sano, M.; Ijichi, H.; Takahashi, R.; Miyabayashi, K.; Fujiwara, H.; Yamada, T.; Kato, H.; Nakatsuka, T.; Tanaka, Y.; Tateishi, K.; et al. Blocking CXCLs-CXCR2 axis in tumor-stromal interactions contributes to survival in a mouse model of pancreatic ductal adenocarcinoma through reduced cell invasion/migration and a shift of immune-inflammatory microenvironment. Oncogenesis 2019, 8, 8. [Google Scholar] [CrossRef]
  51. Roufas, C.; Chasiotis, D.; Makris, A.; Efstathiades, C.; Dimopoulos, C.; Zaravinos, A. The Expression and Prognostic Impact of Immune Cytolytic Activity-Related Markers in Human Malignancies: A Comprehensive Meta-analysis. Front. Oncol. 2018, 8, 27. [Google Scholar] [CrossRef] [PubMed]
  52. Ma, Z.; Xin, Z.; Hu, W.; Jiang, S.; Yang, Z.; Yan, X.; Li, X.; Yang, Y.; Chen, F. Forkhead box O proteins: Crucial regulators of cancer EMT. Semin. Cancer Biol. 2018, 50, 21–31. [Google Scholar] [CrossRef] [PubMed]
  53. Wang, J.; Li, Y. CD36 tango in cancer: Signaling pathways and functions. Theranostics 2019, 9, 4893–4908. [Google Scholar] [CrossRef] [PubMed]
  54. Karamitopoulou, E. Role of epithelial-mesenchymal transition in pancreatic ductal adenocarcinoma: Is tumor budding the missing link? Front. Oncol. 2013, 3, 221. [Google Scholar] [CrossRef] [Green Version]
  55. Miyashita, H.; Watanabe, H.; Ohe, H.; Itakura, Y.; Ohnishi, K.; Hayami, H.; Watanabe, M. [An application of 2D-Doppler color flow mapping to the prostate]. Nihon Hinyokika Gakkai Zasshi. Jpn. J. Urol. 1988, 79, 235–238. [Google Scholar] [CrossRef] [Green Version]
  56. Lawlor, R.T.; Veronese, N.; Nottegar, A.; Malleo, G.; Smith, L.; Demurtas, J.; Cheng, L.; Wood, L.D.; Silvestris, N.; Salvia, R.; et al. Prognostic Role of High-Grade Tumor Budding in Pancreatic Ductal Adenocarcinoma: A Systematic Review and Meta-Analysis with a Focus on Epithelial to Mesenchymal Transition. Cancers 2019, 11, 113. [Google Scholar] [CrossRef] [Green Version]
  57. Galvan, J.A.; Zlobec, I.; Wartenberg, M.; Lugli, A.; Gloor, B.; Perren, A.; Karamitopoulou, E. Expression of E-cadherin repressors SNAIL, ZEB1 and ZEB2 by tumour and stromal cells influences tumour-budding phenotype and suggests heterogeneity of stromal cells in pancreatic cancer. Br. J. Cancer 2015, 112, 1944–1950. [Google Scholar] [CrossRef] [Green Version]
  58. Chu, L.Y.; Ramakrishnan, D.P.; Silverstein, R.L. Thrombospondin-1 modulates VEGF signaling via CD36 by recruiting SHP-1 to VEGFR2 complex in microvascular endothelial cells. Blood 2013, 122, 1822–1832. [Google Scholar] [CrossRef] [Green Version]
  59. Murphy-Ullrich, J.E.; Poczatek, M. Activation of latent TGF-beta by thrombospondin-1: Mechanisms and physiology. Cytokine Growth Factor Rev. 2000, 11, 59–69. [Google Scholar] [CrossRef]
  60. Simonian, S.J.; Stuart, F.P.; Hill, J.L.; Mahajan, S.K. Conversion of a Scribner shunt to an arteriovenous fistula for chronic dialysis. Surgery 1977, 82, 448–451. [Google Scholar]
  61. Katsuno, Y.; Lamouille, S.; Derynck, R. TGF-beta signaling and epithelial-mesenchymal transition in cancer progression. Curr. Opin. Oncol. 2013, 25, 76–84. [Google Scholar] [CrossRef]
  62. Tkach, M.; Thery, C. Communication by Extracellular Vesicles: Where We Are and Where We Need to Go. Cell 2016, 164, 1226–1232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Soung, Y.H.; Ford, S.; Zhang, V.; Chung, J. Exosomes in Cancer Diagnostics. Cancers 2017, 9, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Han, L.; Lam, E.W.; Sun, Y. Extracellular vesicles in the tumor microenvironment: Old stories, but new tales. Mol. Cancer 2019, 18, 59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Al-Nedawi, K.; Meehan, B.; Micallef, J.; Lhotak, V.; May, L.; Guha, A.; Rak, J. Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat. Cell Biol. 2008, 10, 619–624. [Google Scholar] [CrossRef]
  66. Pfeiler, S.; Thakur, M.; Grunauer, P.; Megens, R.T.A.; Joshi, U.; Coletti, R.; Samara, V.; Muller-Stoy, G.; Ishikawa-Ankerhold, H.; Stark, K.; et al. CD36-triggered cell invasion and persistent tissue colonization by tumor microvesicles during metastasis. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2019, 33, 1860–1872. [Google Scholar] [CrossRef] [Green Version]
  67. Krenkel, O.; Tacke, F. Liver macrophages in tissue homeostasis and disease. Nat. Rev. Immunol. 2017, 17, 306–321. [Google Scholar] [CrossRef]
  68. Iwagami, Y.; Eguchi, H.; Nagano, H.; Akita, H.; Hama, N.; Wada, H.; Kawamoto, K.; Kobayashi, S.; Tomokuni, A.; Tomimaru, Y.; et al. miR-320c regulates gemcitabine-resistance in pancreatic cancer via SMARCC1. Br. J. Cancer 2013, 109, 502–511. [Google Scholar] [CrossRef] [Green Version]
  69. Pang, B.; Xu, X.; Lu, Y.; Jin, H.; Yang, R.; Jiang, C.; Shao, D.; Liu, Y.; Shi, J. Prediction of new targets and mechanisms for quercetin in the treatment of pancreatic cancer, colon cancer, and rectal cancer. Food Funct. 2019, 10, 5339–5349. [Google Scholar] [CrossRef]
  70. Eichler, W.; Aust, G.; Hamann, D. Characterization of an early activation-dependent antigen on lymphocytes defined by the monoclonal antibody BL-Ac(F2). Scand. J. Immunol. 1994, 39, 111–115. [Google Scholar] [CrossRef]
  71. Safaee, M.; Clark, A.J.; Ivan, M.E.; Oh, M.C.; Bloch, O.; Sun, M.Z.; Oh, T.; Parsa, A.T. CD97 is a multifunctional leukocyte receptor with distinct roles in human cancers (Review). Int. J. Oncol. 2013, 43, 1343–1350. [Google Scholar] [CrossRef] [PubMed]
  72. Wang, T.; Ward, Y.; Tian, L.; Lake, R.; Guedez, L.; Stetler-Stevenson, W.G.; Kelly, K. CD97, an adhesion receptor on inflammatory cells, stimulates angiogenesis through binding integrin counterreceptors on endothelial cells. Blood 2005, 105, 2836–2844. [Google Scholar] [CrossRef] [PubMed]
  73. Ward, Y.; Lake, R.; Martin, P.L.; Killian, K.; Salerno, P.; Wang, T.; Meltzer, P.; Merino, M.; Cheng, S.Y.; Santoro, M.; et al. CD97 amplifies LPA receptor signaling and promotes thyroid cancer progression in a mouse model. Oncogene 2013, 32, 2726–2738. [Google Scholar] [CrossRef] [Green Version]
  74. Han, S.L.; Xu, C.; Wu, X.L.; Li, J.L.; Liu, Z.; Zeng, Q.Q. The impact of expressions of CD97 and its ligand CD55 at the invasion front on prognosis of rectal adenocarcinoma. Int. J. Colorectal Dis. 2010, 25, 695–702. [Google Scholar] [CrossRef] [PubMed]
  75. He, Z.; Wu, H.; Jiao, Y.; Zheng, J. Expression and prognostic value of CD97 and its ligand CD55 in pancreatic cancer. Oncol. Lett. 2015, 9, 793–797. [Google Scholar] [CrossRef] [Green Version]
  76. Liu, D.; Trojanowicz, B.; Ye, L.; Li, C.; Zhang, L.; Li, X.; Li, G.; Zheng, Y.; Chen, L. The invasion and metastasis promotion role of CD97 small isoform in gastric carcinoma. PLoS ONE 2012, 7, e39989. [Google Scholar] [CrossRef]
  77. Somasundaram, A.; Ardanowski, N.; Opalak, C.F.; Fillmore, H.L.; Chidambaram, A.; Broaddus, W.C. Wilms tumor 1 gene, CD97, and the emerging biogenetic profile of glioblastoma. Neurosurg. Focus 2014, 37, E14. [Google Scholar] [CrossRef] [Green Version]
  78. Ward, Y.; Lake, R.; Yin, J.J.; Heger, C.D.; Raffeld, M.; Goldsmith, P.K.; Merino, M.; Kelly, K. LPA receptor heterodimerizes with CD97 to amplify LPA-initiated RHO-dependent signaling and invasion in prostate cancer cells. Cancer Res. 2011, 71, 7301–7311. [Google Scholar] [CrossRef] [Green Version]
  79. Yin, Y.; Xu, X.; Tang, J.; Zhang, W.; Zhangyuan, G.; Ji, J.; Deng, L.; Lu, S.; Zhuo, H.; Sun, B. CD97 Promotes Tumor Aggressiveness Through the Traditional G Protein-Coupled Receptor-Mediated Signaling in Hepatocellular Carcinoma. Hepatology 2018, 68, 1865–1878. [Google Scholar] [CrossRef] [Green Version]
  80. Yokoyama, K.; Reynolds, J.C.; Paik, C.H.; Sood, V.K.; Maloney, P.J.; Larson, S.M.; Reba, R.C. Immunoreactivity affects the biodistribution and tumor targeting of radiolabeled anti-P97 Fab fragment. J. Nucl. Med. Off. Publ. Soc. Nucl. Med. 1990, 31, 202–210. [Google Scholar]
  81. Jaspars, L.H.; Vos, W.; Aust, G.; Van Lier, R.A.; Hamann, J. Tissue distribution of the human CD97 EGF-TM7 receptor. Tissue Antigens 2001, 57, 325–331. [Google Scholar] [CrossRef] [PubMed]
  82. Kwakkenbos, M.J.; Pouwels, W.; Matmati, M.; Stacey, M.; Lin, H.H.; Gordon, S.; van Lier, R.A.; Hamann, J. Expression of the largest CD97 and EMR2 isoforms on leukocytes facilitates a specific interaction with chondroitin sulfate on B cells. J. Leukoc. Biol. 2005, 77, 112–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Kwakkenbos, M.J.; Matmati, M.; Madsen, O.; Pouwels, W.; Wang, Y.; Bontrop, R.E.; Heidt, P.J.; Hoek, R.M.; Hamann, J. An unusual mode of concerted evolution of the EGF-TM7 receptor chimera EMR2. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2006, 20, 2582–2584. [Google Scholar] [CrossRef] [Green Version]
  84. Veninga, H.; Becker, S.; Hoek, R.M.; Wobus, M.; Wandel, E.; van der Kaa, J.; van der Valk, M.; de Vos, A.F.; Haase, H.; Owens, B.; et al. Analysis of CD97 expression and manipulation: Antibody treatment but not gene targeting curtails granulocyte migration. J. Immunol. 2008, 181, 6574–6583. [Google Scholar] [CrossRef] [Green Version]
  85. Yeon Won, H.; Hwan Mun, S.; Shin, B.; Lee, S.K. Contradictory Role of CD97 in Basal and Tumor Necrosis Factor-Induced Osteoclastogenesis In Vivo. Arthritis Rheumatol. 2016, 68, 1301–1313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Tjong, W.Y.; Lin, H.H. The RGD motif is involved in CD97/ADGRE5-promoted cell adhesion and viability of HT1080 cells. Sci. Rep. 2019, 9, 1517. [Google Scholar] [CrossRef] [Green Version]
  87. Gray, J.X.; Haino, M.; Roth, M.J.; Maguire, J.E.; Jensen, P.N.; Yarme, A.; Stetler-Stevenson, M.A.; Siebenlist, U.; Kelly, K. CD97 is a processed, seven-transmembrane, heterodimeric receptor associated with inflammation. J. Immunol. 1996, 157, 5438–5447. [Google Scholar]
  88. Kop, E.N.; Adriaansen, J.; Smeets, T.J.; Vervoordeldonk, M.J.; van Lier, R.A.; Hamann, J.; Tak, P.P. CD97 neutralisation increases resistance to collagen-induced arthritis in mice. Arthritis Res. Ther. 2006, 8, R155. [Google Scholar] [CrossRef] [Green Version]
  89. Aust, G.; Steinert, M.; Schutz, A.; Boltze, C.; Wahlbuhl, M.; Hamann, J.; Wobus, M. CD97, but not its closely related EGF-TM7 family member EMR2, is expressed on gastric, pancreatic, and esophageal carcinomas. Am. J. Clin. Pathol. 2002, 118, 699–707. [Google Scholar] [CrossRef]
  90. Boltze, C.; Schneider-Stock, R.; Aust, G.; Mawrin, C.; Dralle, H.; Roessner, A.; Hoang-Vu, C. CD97, CD95 and Fas-L clearly discriminate between chronic pancreatitis and pancreatic ductal adenocarcinoma in perioperative evaluation of cryocut sections. Pathol. Int. 2002, 52, 83–88. [Google Scholar] [CrossRef]
  91. Steinert, M.; Wobus, M.; Boltze, C.; Schutz, A.; Wahlbuhl, M.; Hamann, J.; Aust, G. Expression and regulation of CD97 in colorectal carcinoma cell lines and tumor tissues. Am. J. Pathol. 2002, 161, 1657–1667. [Google Scholar] [CrossRef] [Green Version]
  92. Hamann, J.; Wishaupt, J.O.; van Lier, R.A.; Smeets, T.J.; Breedveld, F.C.; Tak, P.P. Expression of the activation antigen CD97 and its ligand CD55 in rheumatoid synovial tissue. Arthritis Rheum. 1999, 42, 650–658. [Google Scholar] [CrossRef]
  93. Mikesch, J.H.; Schier, K.; Roetger, A.; Simon, R.; Buerger, H.; Brandt, B. The expression and action of decay-accelerating factor (CD55) in human malignancies and cancer therapy. Cell. Oncol. Off. J. Int. Soc. Cell. Oncol. 2006, 28, 223–232. [Google Scholar] [CrossRef]
  94. Vogl, U.M.; Ohler, L.; Rasic, M.; Frischer, J.M.; Modak, M.; Stockl, J. Evaluation of Prognostic Immune Signatures in Patients with Breast, Colorectal and Pancreatic Cancer Receiving Chemotherapy. Anticancer Res. 2017, 37, 1947–1955. [Google Scholar] [CrossRef] [Green Version]
  95. Heinzelmann, K.; Lehmann, M.; Gerckens, M.; Noskovicova, N.; Frankenberger, M.; Lindner, M.; Hatz, R.; Behr, J.; Hilgendorff, A.; Konigshoff, M.; et al. Cell-surface phenotyping identifies CD36 and CD97 as novel markers of fibroblast quiescence in lung fibrosis. Am. J. Physiol. Lung Cell. Mol. Physiol. 2018, 315, L682–L696. [Google Scholar] [CrossRef]
  96. Schosserer, M.; Grillari, J.; Breitenbach, M. The Dual Role of Cellular Senescence in Developing Tumors and Their Response to Cancer Therapy. Front. Oncol. 2017, 7, 278. [Google Scholar] [CrossRef] [Green Version]
  97. Morris, S.A. The evolving concept of cell identity in the single cell era. Development 2019, 146, dev.169748. [Google Scholar] [CrossRef] [Green Version]
  98. Nurmik, M.; Ullmann, P.; Rodriguez, F.; Haan, S.; Letellier, E. In search of definitions: Cancer-associated fibroblasts and their markers. Int. J. Cancer 2020, 146, 895–905. [Google Scholar] [CrossRef] [Green Version]
  99. Codrici, E.; Enciu, A.M.; Popescu, I.D.; Mihai, S.; Tanase, C. Glioma Stem Cells and Their Microenvironments: Providers of Challenging Therapeutic Targets. Stem Cells Int. 2016, 2016, 5728438. [Google Scholar] [CrossRef] [Green Version]
  100. Lau, A.N.; Heiden, M.G.V. Metabolism in the Tumor Microenvironment. Annu. Rev. Cancer Biol. 2020, 4, 17–40. [Google Scholar] [CrossRef] [Green Version]
  101. Feig, C.; Gopinathan, A.; Neesse, A.; Chan, D.S.; Cook, N.; Tuveson, D.A. The pancreas cancer microenvironment. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2012, 18, 4266–4276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Stopa, K.B.; Kusiak, A.A.; Szopa, M.D.; Ferdek, P.E.; Jakubowska, M.A. Pancreatic Cancer and Its Microenvironment-Recent Advances and Current Controversies. Int. J. Mol. Sci. 2020, 21, 3218. [Google Scholar] [CrossRef] [PubMed]
  103. Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 2016, 16, 582–598. [Google Scholar] [CrossRef] [PubMed]
  104. Roberts, E.W.; Deonarine, A.; Jones, J.O.; Denton, A.E.; Feig, C.; Lyons, S.K.; Espeli, M.; Kraman, M.; McKenna, B.; Wells, R.J.; et al. Depletion of stromal cells expressing fibroblast activation protein-alpha from skeletal muscle and bone marrow results in cachexia and anemia. J. Exp. Med. 2013, 210, 1137–1151. [Google Scholar] [CrossRef] [PubMed]
  105. Tomasek, J.J.; Gabbiani, G.; Hinz, B.; Chaponnier, C.; Brown, R.A. Myofibroblasts and mechano-regulation of connective tissue remodelling. Nat. Rev. Mol. Cell Biol. 2002, 3, 349–363. [Google Scholar] [CrossRef]
  106. Sahai, E.; Astsaturov, I.; Cukierman, E.; DeNardo, D.G.; Egeblad, M.; Evans, R.M.; Fearon, D.; Greten, F.R.; Hingorani, S.R.; Hunter, T.; et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat. Rev. Cancer 2020, 20, 174–186. [Google Scholar] [CrossRef] [Green Version]
  107. Watari, N.; Hotta, Y.; Mabuchi, Y. Morphological studies on a vitamin A-storing cell and its complex with macrophage observed in mouse pancreatic tissues following excess vitamin A administration. Okajimas Folia Anat. Jpn. 1982, 58, 837–858. [Google Scholar] [CrossRef] [Green Version]
  108. Wehr, A.Y.; Furth, E.E.; Sangar, V.; Blair, I.A.; Yu, K.H. Analysis of the human pancreatic stellate cell secreted proteome. Pancreas 2011, 40, 557–566. [Google Scholar] [CrossRef] [Green Version]
  109. Apte, M.; Pirola, R.C.; Wilson, J.S. Pancreatic stellate cell: Physiologic role, role in fibrosis and cancer. Curr. Opin. Gastroenterol. 2015, 31, 416–423. [Google Scholar] [CrossRef]
  110. Blaner, W.S.; O’Byrne, S.M.; Wongsiriroj, N.; Kluwe, J.; D’Ambrosio, D.M.; Jiang, H.; Schwabe, R.F.; Hillman, E.M.; Piantedosi, R.; Libien, J. Hepatic stellate cell lipid droplets: A specialized lipid droplet for retinoid storage. Biochim. Biophys. Acta 2009, 1791, 467–473. [Google Scholar] [CrossRef] [Green Version]
  111. Sherman, M.H.; Yu, R.T.; Tseng, T.W.; Sousa, C.M.; Liu, S.; Truitt, M.L.; He, N.; Ding, N.; Liddle, C.; Atkins, A.R.; et al. Stromal cues regulate the pancreatic cancer epigenome and metabolome. Proc. Natl. Acad. Sci. USA 2017, 114, 1129–1134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Olumi, A.F.; Grossfeld, G.D.; Hayward, S.W.; Carroll, P.R.; Tlsty, T.D.; Cunha, G.R. Carcinoma-associated fibroblasts direct tumor progression of initiated human prostatic epithelium. Cancer Res. 1999, 59, 5002–5011. [Google Scholar] [CrossRef] [PubMed]
  113. Pereira, B.A.; Vennin, C.; Papanicolaou, M.; Chambers, C.R.; Herrmann, D.; Morton, J.P.; Cox, T.R.; Timpson, P. CAF Subpopulations: A New Reservoir of Stromal Targets in Pancreatic Cancer. Trends Cancer 2019, 5, 724–741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Jacobetz, M.A.; Chan, D.S.; Neesse, A.; Bapiro, T.E.; Cook, N.; Frese, K.K.; Feig, C.; Nakagawa, T.; Caldwell, M.E.; Zecchini, H.I.; et al. Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut 2013, 62, 112–120. [Google Scholar] [CrossRef]
  115. Neesse, A.; Frese, K.K.; Bapiro, T.E.; Nakagawa, T.; Sternlicht, M.D.; Seeley, T.W.; Pilarsky, C.; Jodrell, D.I.; Spong, S.M.; Tuveson, D.A. CTGF antagonism with mAb FG-3019 enhances chemotherapy response without increasing drug delivery in murine ductal pancreas cancer. Proc. Natl. Acad. Sci. USA 2013, 110, 12325–12330. [Google Scholar] [CrossRef] [Green Version]
  116. Olive, K.P.; Jacobetz, M.A.; Davidson, C.J.; Gopinathan, A.; McIntyre, D.; Honess, D.; Madhu, B.; Goldgraben, M.A.; Caldwell, M.E.; Allard, D.; et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 2009, 324, 1457–1461. [Google Scholar] [CrossRef] [Green Version]
  117. Provenzano, P.P.; Cuevas, C.; Chang, A.E.; Goel, V.K.; Von Hoff, D.D.; Hingorani, S.R. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell 2012, 21, 418–429. [Google Scholar] [CrossRef] [Green Version]
  118. Strell, C.; Paulsson, J.; Jin, S.B.; Tobin, N.P.; Mezheyeuski, A.; Roswall, P.; Mutgan, C.; Mitsios, N.; Johansson, H.; Wickberg, S.M.; et al. Impact of Epithelial-Stromal Interactions on Peritumoral Fibroblasts in Ductal Carcinoma in Situ. J. Natl. Cancer Inst. 2019, 111, 983–995. [Google Scholar] [CrossRef]
  119. Albrengues, J.; Bertero, T.; Grasset, E.; Bonan, S.; Maiel, M.; Bourget, I.; Philippe, C.; Herraiz Serrano, C.; Benamar, S.; Croce, O.; et al. Epigenetic switch drives the conversion of fibroblasts into proinvasive cancer-associated fibroblasts. Nat. Commun. 2015, 6, 10204. [Google Scholar] [CrossRef] [Green Version]
  120. Albrengues, J.; Bourget, I.; Pons, C.; Butet, V.; Hofman, P.; Tartare-Deckert, S.; Feral, C.C.; Meneguzzi, G.; Gaggioli, C. LIF mediates proinvasive activation of stromal fibroblasts in cancer. Cell Rep. 2014, 7, 1664–1678. [Google Scholar] [CrossRef] [Green Version]
  121. Avery, D.; Govindaraju, P.; Jacob, M.; Todd, L.; Monslow, J.; Pure, E. Extracellular matrix directs phenotypic heterogeneity of activated fibroblasts. Matrix Biol. J. Int. Soc. Matrix Biol. 2018, 67, 90–106. [Google Scholar] [CrossRef] [PubMed]
  122. Calvo, F.; Ege, N.; Grande-Garcia, A.; Hooper, S.; Jenkins, R.P.; Chaudhry, S.I.; Harrington, K.; Williamson, P.; Moeendarbary, E.; Charras, G.; et al. Mechanotransduction and YAP-dependent matrix remodelling is required for the generation and maintenance of cancer-associated fibroblasts. Nat. Cell Biol. 2013, 15, 637–646. [Google Scholar] [CrossRef] [PubMed]
  123. Amatangelo, M.D.; Bassi, D.E.; Klein-Szanto, A.J.; Cukierman, E. Stroma-derived three-dimensional matrices are necessary and sufficient to promote desmoplastic differentiation of normal fibroblasts. Am. J. Pathol. 2005, 167, 475–488. [Google Scholar] [CrossRef] [Green Version]
  124. Ferrari, N.; Ranftl, R.; Chicherova, I.; Slaven, N.D.; Moeendarbary, E.; Farrugia, A.J.; Lam, M.; Semiannikova, M.; Westergaard, M.C.W.; Tchou, J.; et al. Dickkopf-3 links HSF1 and YAP/TAZ signalling to control aggressive behaviours in cancer-associated fibroblasts. Nat. Commun. 2019, 10, 130. [Google Scholar] [CrossRef]
  125. Fordyce, C.; Fessenden, T.; Pickering, C.; Jung, J.; Singla, V.; Berman, H.; Tlsty, T. DNA damage drives an activin a-dependent induction of cyclooxygenase-2 in premalignant cells and lesions. Cancer Prev. Res. (Phila) 2010, 3, 190–201. [Google Scholar] [CrossRef] [Green Version]
  126. Demaria, M.; O’Leary, M.N.; Chang, J.; Shao, L.; Liu, S.; Alimirah, F.; Koenig, K.; Le, C.; Mitin, N.; Deal, A.M.; et al. Cellular Senescence Promotes Adverse Effects of Chemotherapy and Cancer Relapse. Cancer Discov. 2017, 7, 165–176. [Google Scholar] [CrossRef] [Green Version]
  127. Pietras, K.; Ostman, A. Hallmarks of cancer: Interactions with the tumor stroma. Exp. Cell Res. 2010, 316, 1324–1331. [Google Scholar] [CrossRef]
  128. Sugimoto, H.; Mundel, T.M.; Kieran, M.W.; Kalluri, R. Identification of fibroblast heterogeneity in the tumor microenvironment. Cancer Biol. Ther. 2006, 5, 1640–1646. [Google Scholar] [CrossRef] [Green Version]
  129. Kobayashi, H.; Enomoto, A.; Woods, S.L.; Burt, A.D.; Takahashi, M.; Worthley, D.L. Cancer-associated fibroblasts in gastrointestinal cancer. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 282–295. [Google Scholar] [CrossRef]
  130. Okumura, T.; Ohuchida, K.; Kibe, S.; Iwamoto, C.; Ando, Y.; Takesue, S.; Nakayama, H.; Abe, T.; Endo, S.; Koikawa, K.; et al. Adipose tissue-derived stromal cells are sources of cancer-associated fibroblasts and enhance tumor progression by dense collagen matrix. Int. J. Cancer 2019, 144, 1401–1413. [Google Scholar] [CrossRef]
  131. DeFilippis, R.A.; Chang, H.; Dumont, N.; Rabban, J.T.; Chen, Y.Y.; Fontenay, G.V.; Berman, H.K.; Gauthier, M.L.; Zhao, J.; Hu, D.; et al. CD36 repression activates a multicellular stromal program shared by high mammographic density and tumor tissues. Cancer Discov. 2012, 2, 826–839. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. DeFilippis, R.A.; Fordyce, C.; Patten, K.; Chang, H.; Zhao, J.; Fontenay, G.V.; Kerlikowske, K.; Parvin, B.; Tlsty, T.D. Stress signaling from human mammary epithelial cells contributes to phenotypes of mammographic density. Cancer Res. 2014, 74, 5032–5044. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Costa, A.; Kieffer, Y.; Scholer-Dahirel, A.; Pelon, F.; Bourachot, B.; Cardon, M.; Sirven, P.; Magagna, I.; Fuhrmann, L.; Bernard, C.; et al. Fibroblast Heterogeneity and Immunosuppressive Environment in Human Breast Cancer. Cancer Cell 2018, 33, 463–479.e10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Ohlund, D.; Handly-Santana, A.; Biffi, G.; Elyada, E.; Almeida, A.S.; Ponz-Sarvise, M.; Corbo, V.; Oni, T.E.; Hearn, S.A.; Lee, E.J.; et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J. Exp. Med. 2017, 214, 579–596. [Google Scholar] [CrossRef]
  135. Goulet, C.R.; Champagne, A.; Bernard, G.; Vandal, D.; Chabaud, S.; Pouliot, F.; Bolduc, S. Cancer-associated fibroblasts induce epithelial-mesenchymal transition of bladder cancer cells through paracrine IL-6 signalling. BMC Cancer 2019, 19, 137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Biffi, G.; Oni, T.E.; Spielman, B.; Hao, Y.; Elyada, E.; Park, Y.; Preall, J.; Tuveson, D.A. IL1-Induced JAK/STAT Signaling Is Antagonized by TGFbeta to Shape CAF Heterogeneity in Pancreatic Ductal Adenocarcinoma. Cancer Discov. 2019, 9, 282–301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Somerville, T.D.; Biffi, G.; Dassler-Plenker, J.; Hur, S.K.; He, X.Y.; Vance, K.E.; Miyabayashi, K.; Xu, Y.; Maia-Silva, D.; Klingbeil, O.; et al. Squamous trans-differentiation of pancreatic cancer cells promotes stromal inflammation. ELife 2020, 9, e53381. [Google Scholar] [CrossRef] [Green Version]
  138. Lewis, J.S.; Keshari, K.R. Cancer Metabolism. In Imaging and Metabolism; Springer International Publishing: Berlin/Heidelberg, Germany, 2018. [Google Scholar] [CrossRef]
  139. Nizri, E.; Bar-David, S.; Aizic, A.; Sternbach, N.; Lahat, G.; Wolf, I.; Klausner, J. Desmoplasia in Lymph Node Metastasis of Pancreatic Adenocarcinoma Reveals Activation of Cancer-Associated Fibroblasts Pattern and T-helper 2 Immune Cell Infiltration. Pancreas 2019, 48, 367–373. [Google Scholar] [CrossRef]
  140. Gascard, P.; Tlsty, T.D. Carcinoma-associated fibroblasts: Orchestrating the composition of malignancy. Genes Dev. 2016, 30, 1002–1019. [Google Scholar] [CrossRef]
  141. O’Connell, J.T.; Sugimoto, H.; Cooke, V.G.; MacDonald, B.A.; Mehta, A.I.; LeBleu, V.S.; Dewar, R.; Rocha, R.M.; Brentani, R.R.; Resnick, M.B.; et al. VEGF-A and Tenascin-C produced by S100A4+ stromal cells are important for metastatic colonization. Proc. Natl. Acad. Sci. USA 2011, 108, 16002–16007. [Google Scholar] [CrossRef] [Green Version]
  142. Harper, J.; Sainson, R.C. Regulation of the anti-tumour immune response by cancer-associated fibroblasts. Semin. Cancer Biol. 2014, 25, 69–77. [Google Scholar] [CrossRef] [PubMed]
  143. Yu, Y.; Xiao, C.H.; Tan, L.D.; Wang, Q.S.; Li, X.Q.; Feng, Y.M. Cancer-associated fibroblasts induce epithelial-mesenchymal transition of breast cancer cells through paracrine TGF-beta signalling. Br. J. Cancer 2014, 110, 724–732. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Tyan, S.W.; Kuo, W.H.; Huang, C.K.; Pan, C.C.; Shew, J.Y.; Chang, K.J.; Lee, E.Y.; Lee, W.H. Breast cancer cells induce cancer-associated fibroblasts to secrete hepatocyte growth factor to enhance breast tumorigenesis. PLoS ONE 2011, 6, e15313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Auciello, F.R.; Bulusu, V.; Oon, C.; Tait-Mulder, J.; Berry, M.; Bhattacharyya, S.; Tumanov, S.; Allen-Petersen, B.L.; Link, J.; Kendsersky, N.D.; et al. A Stromal Lysolipid-Autotaxin Signaling Axis Promotes Pancreatic Tumor Progression. Cancer Discov. 2019, 9, 617–627. [Google Scholar] [CrossRef] [Green Version]
  146. Zhuang, J.; Lu, Q.; Shen, B.; Huang, X.; Shen, L.; Zheng, X.; Huang, R.; Yan, J.; Guo, H. TGFbeta1 secreted by cancer-associated fibroblasts induces epithelial-mesenchymal transition of bladder cancer cells through lncRNA-ZEB2NAT. Sci. Rep. 2015, 5, 11924. [Google Scholar] [CrossRef] [Green Version]
  147. Orimo, A.; Gupta, P.B.; Sgroi, D.C.; Arenzana-Seisdedos, F.; Delaunay, T.; Naeem, R.; Carey, V.J.; Richardson, A.L.; Weinberg, R.A. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell 2005, 121, 335–348. [Google Scholar] [CrossRef]
  148. Matsuo, Y.; Ding, Q.; Desaki, R.; Maemura, K.; Mataki, Y.; Shinchi, H.; Natsugoe, S.; Takao, S. Hypoxia inducible factor-1 alpha plays a pivotal role in hepatic metastasis of pancreatic cancer: An immunohistochemical study. J. Hepato Biliary Pancreat. Sci. 2014, 21, 105–112. [Google Scholar] [CrossRef]
  149. Samain, R.; Sanz-Moreno, V. Cancer-associated fibroblasts: Activin A adds another string to their bow. Embo Mol. Med. 2020, 12, e12102. [Google Scholar] [CrossRef]
  150. Sun, Y.; Fan, X.; Zhang, Q.; Shi, X.; Xu, G.; Zou, C. Cancer-associated fibroblasts secrete FGF-1 to promote ovarian proliferation, migration, and invasion through the activation of FGF-1/FGFR4 signaling. Tumour Biol. J. Int. Soc. Oncodev. Biol. Med. 2017, 39, 1010428317712592. [Google Scholar] [CrossRef] [Green Version]
  151. LeBleu, V.S.; Kalluri, R. A peek into cancer-associated fibroblasts: Origins, functions and translational impact. Dis. Models Mech. 2018, 11, dmm029447. [Google Scholar] [CrossRef] [Green Version]
  152. Nissen, N.I.; Karsdal, M.; Willumsen, N. Collagens and Cancer associated fibroblasts in the reactive stroma and its relation to Cancer biology. J. Exp. Clin. Cancer Res. CR 2019, 38, 115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Erdogan, B.; Ao, M.; White, L.M.; Means, A.L.; Brewer, B.M.; Yang, L.; Washington, M.K.; Shi, C.; Franco, O.E.; Weaver, A.M.; et al. Cancer-associated fibroblasts promote directional cancer cell migration by aligning fibronectin. J. Cell Biol. 2017, 216, 3799–3816. [Google Scholar] [CrossRef] [Green Version]
  154. Qin, X.; Yan, M.; Wang, X.; Xu, Q.; Zhu, X.; Shi, J.; Li, Z.; Zhang, J.; Chen, W. Cancer-associated Fibroblast-derived IL-6 Promotes Head and Neck Cancer Progression via the Osteopontin-NF-kappa B Signaling Pathway. Theranostics 2018, 8, 921–940. [Google Scholar] [CrossRef] [PubMed]
  155. Sanford-Crane, H.; Abrego, J.; Sherman, M.H. Fibroblasts as Modulators of Local and Systemic Cancer Metabolism. Cancers 2019, 11, 619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Bertero, T.; Oldham, W.M.; Grasset, E.M.; Bourget, I.; Boulter, E.; Pisano, S.; Hofman, P.; Bellvert, F.; Meneguzzi, G.; Bulavin, D.V.; et al. Tumor-Stroma Mechanics Coordinate Amino Acid Availability to Sustain Tumor Growth and Malignancy. Cell Metab. 2019, 29, 124–140.e10. [Google Scholar] [CrossRef] [Green Version]
  157. Valencia, T.; Kim, J.Y.; Abu-Baker, S.; Moscat-Pardos, J.; Ahn, C.S.; Reina-Campos, M.; Duran, A.; Castilla, E.A.; Metallo, C.M.; Diaz-Meco, M.T.; et al. Metabolic reprogramming of stromal fibroblasts through p62-mTORC1 signaling promotes inflammation and tumorigenesis. Cancer Cell 2014, 26, 121–135. [Google Scholar] [CrossRef] [Green Version]
  158. Martinez-Outschoorn, U.E.; Lisanti, M.P.; Sotgia, F. Catabolic cancer-associated fibroblasts transfer energy and biomass to anabolic cancer cells, fueling tumor growth. Semin. Cancer Biol. 2014, 25, 47–60. [Google Scholar] [CrossRef]
  159. Sousa, C.M.; Biancur, D.E.; Wang, X.; Halbrook, C.J.; Sherman, M.H.; Zhang, L.; Kremer, D.; Hwang, R.F.; Witkiewicz, A.K.; Ying, H.; et al. Pancreatic stellate cells support tumour metabolism through autophagic alanine secretion. Nature 2016, 536, 479–483. [Google Scholar] [CrossRef] [Green Version]
  160. Knudsen, E.S.; Balaji, U.; Freinkman, E.; McCue, P.; Witkiewicz, A.K. Unique metabolic features of pancreatic cancer stroma: Relevance to the tumor compartment, prognosis, and invasive potential. Oncotarget 2016, 7, 78396–78411. [Google Scholar] [CrossRef] [Green Version]
  161. Francescone, R.; Vendramini-Costa, D.B.; Franco-Barraza, J.; Wagner, J.; Muir, A.; Lau, A.N.; Gabitova, L.; Pazina, T.; Gupta, S.; Luong, T.; et al. The NetrinG1/NGL-1 Axis promotes pancreatic tumorigenesis through cancer associated fibroblast driven nutritional support and immunosuppression. BioRxiv 2019, 330209. [Google Scholar] [CrossRef] [Green Version]
  162. Shan, T.; Lu, H.; Ji, H.; Li, Y.; Guo, J.; Chen, X.; Wu, T. Loss of stromal caveolin-1 expression: A novel tumor microenvironment biomarker that can predict poor clinical outcomes for pancreatic cancer. PLoS ONE 2014, 9, e97239. [Google Scholar] [CrossRef]
  163. Codrici, E.; Albulescu, L.; Popescu, I.D.; Mihai, S.; Enciu, A.M.; Albulescu, R.; Tanase, C.; Hinescu, M.E. Caveolin-1-Knockout Mouse as a Model of Inflammatory Diseases. J. Immunol. Res. 2018, 2018, 2498576. [Google Scholar] [CrossRef] [PubMed]
  164. Boj, S.F.; Hwang, C.I.; Baker, L.A.; Chio, I.I.; Engle, D.D.; Corbo, V.; Jager, M.; Ponz-Sarvise, M.; Tiriac, H.; Spector, M.S.; et al. Organoid models of human and mouse ductal pancreatic cancer. Cell 2015, 160, 324–338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Huang, L.; Holtzinger, A.; Jagan, I.; BeGora, M.; Lohse, I.; Ngai, N.; Nostro, C.; Wang, R.; Muthuswamy, L.B.; Crawford, H.C.; et al. Ductal pancreatic cancer modeling and drug screening using human pluripotent stem cell- and patient-derived tumor organoids. Nat. Med. 2015, 21, 1364–1371. [Google Scholar] [CrossRef] [PubMed]
  166. Li, X.; Nadauld, L.; Ootani, A.; Corney, D.C.; Pai, R.K.; Gevaert, O.; Cantrell, M.A.; Rack, P.G.; Neal, J.T.; Chan, C.W.; et al. Oncogenic transformation of diverse gastrointestinal tissues in primary organoid culture. Nat. Med. 2014, 20, 769–777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Walsh, A.J.; Castellanos, J.A.; Nagathihalli, N.S.; Merchant, N.B.; Skala, M.C. Optical Imaging of Drug-Induced Metabolism Changes in Murine and Human Pancreatic Cancer Organoids Reveals Heterogeneous Drug Response. Pancreas 2016, 45, 863–869. [Google Scholar] [CrossRef] [PubMed]
  168. Fan, T.W.; Lane, A.N.; Higashi, R.M. Stable Isotope Resolved Metabolomics Studies in Ex Vivo TIssue Slices. Bio-Protocol 2016, 6, e1730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Ozdemir, B.C.; Pentcheva-Hoang, T.; Carstens, J.L.; Zheng, X.; Wu, C.C.; Simpson, T.R.; Laklai, H.; Sugimoto, H.; Kahlert, C.; Novitskiy, S.V.; et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell 2014, 25, 719–734. [Google Scholar] [CrossRef] [Green Version]
  170. Feig, C.; Jones, J.O.; Kraman, M.; Wells, R.J.; Deonarine, A.; Chan, D.S.; Connell, C.M.; Roberts, E.W.; Zhao, Q.; Caballero, O.L.; et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc. Natl. Acad. Sci. USA 2013, 110, 20212–20217. [Google Scholar] [CrossRef] [Green Version]
  171. Froeling, F.E.; Feig, C.; Chelala, C.; Dobson, R.; Mein, C.E.; Tuveson, D.A.; Clevers, H.; Hart, I.R.; Kocher, H.M. Retinoic acid-induced pancreatic stellate cell quiescence reduces paracrine Wnt-beta-catenin signaling to slow tumor progression. Gastroenterology 2011, 141, 1486–1497. [Google Scholar] [CrossRef]
  172. Ene-Obong, A.; Clear, A.J.; Watt, J.; Wang, J.; Fatah, R.; Riches, J.C.; Marshall, J.F.; Chin-Aleong, J.; Chelala, C.; Gribben, J.G.; et al. Activated pancreatic stellate cells sequester CD8+ T cells to reduce their infiltration of the juxtatumoral compartment of pancreatic ductal adenocarcinoma. Gastroenterology 2013, 145, 1121–1132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Sherman, M.H.; Yu, R.T.; Engle, D.D.; Ding, N.; Atkins, A.R.; Tiriac, H.; Collisson, E.A.; Connor, F.; Van Dyke, T.; Kozlov, S.; et al. Vitamin D receptor-mediated stromal reprogramming suppresses pancreatitis and enhances pancreatic cancer therapy. Cell 2014, 159, 80–93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Available online: https://reactome.org/PathwayBrowser/#/R-HSA-2644603&PATH=R-HSA-1643685,R-HSA-5663202&DTAB=AN&ANALYSIS=MjAyMDA2MTcyMTMxMjlfMzk3 (accessed on 29 July 2020).
  175. Connolly, E.C.; Freimuth, J.; Akhurst, R.J. Complexities of TGF-beta targeted cancer therapy. Int. J. Biol. Sci. 2012, 8, 964–978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Massague, J. TGFbeta signalling in context. Nat. Rev. Mol. Cell Biol. 2012, 13, 616–630. [Google Scholar] [CrossRef]
  177. Zhao, M.; Mishra, L.; Deng, C.X. The role of TGF-beta/SMAD4 signaling in cancer. Int. J. Biol. Sci. 2018, 14, 111–123. [Google Scholar] [CrossRef] [Green Version]
  178. Ellenrieder, V.; Fernandez Zapico, M.E.; Urrutia, R. TGFbeta-mediated signaling and transcriptional regulation in pancreatic development and cancer. Curr. Opin. Gastroenterol. 2001, 17, 434–440. [Google Scholar] [CrossRef]
  179. Derynck, R.; Akhurst, R.J.; Balmain, A. TGF-beta signaling in tumor suppression and cancer progression. Nat. Genet. 2001, 29, 117–129. [Google Scholar] [CrossRef]
  180. Hunt, K.K.; Fleming, J.B.; Abramian, A.; Zhang, L.; Evans, D.B.; Chiao, P.J. Overexpression of the tumor suppressor gene Smad4/DPC4 induces p21waf1 expression and growth inhibition in human carcinoma cells. Cancer Res. 1998, 58, 5656–5661. [Google Scholar]
  181. Grau, A.M.; Zhang, L.; Wang, W.; Ruan, S.; Evans, D.B.; Abbruzzese, J.L.; Zhang, W.; Chiao, P.J. Induction of p21waf1 expression and growth inhibition by transforming growth factor beta involve the tumor suppressor gene DPC4 in human pancreatic adenocarcinoma cells. Cancer Res. 1997, 57, 3929–3934. [Google Scholar]
  182. Lecanda, J.; Ganapathy, V.; D’Aquino-Ardalan, C.; Evans, B.; Cadacio, C.; Ayala, A.; Gold, L.I. TGFbeta prevents proteasomal degradation of the cyclin-dependent kinase inhibitor p27kip1 for cell cycle arrest. Cell Cycle 2009, 8, 742–756. [Google Scholar] [CrossRef] [Green Version]
  183. Tachibana, I.; Imoto, M.; Adjei, P.N.; Gores, G.J.; Subramaniam, M.; Spelsberg, T.C.; Urrutia, R. Overexpression of the TGFbeta-regulated zinc finger encoding gene, TIEG, induces apoptosis in pancreatic epithelial cells. J. Clin. Investig. 1997, 99, 2365–2374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Fabregat, A.; Sidiropoulos, K.; Viteri, G.; Marin-Garcia, P.; Ping, P.; Stein, L.; D’Eustachio, P.; Hermjakob, H. Reactome diagram viewer: Data structures and strategies to boost performance. Bioinformatics 2018, 34, 1208–1214. [Google Scholar] [CrossRef] [PubMed]
  185. Hahn, S.A.; Schutte, M.; Hoque, A.T.; Moskaluk, C.A.; da Costa, L.T.; Rozenblum, E.; Weinstein, C.L.; Fischer, A.; Yeo, C.J.; Hruban, R.H.; et al. DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science 1996, 271, 350–353. [Google Scholar] [CrossRef] [PubMed]
  186. Hahn, S.A.; Seymour, A.B.; Hoque, A.T.; Schutte, M.; da Costa, L.T.; Redston, M.S.; Caldas, C.; Weinstein, C.L.; Fischer, A.; Yeo, C.J.; et al. Allelotype of pancreatic adenocarcinoma using xenograft enrichment. Cancer Res. 1995, 55, 4670–4675. [Google Scholar]
  187. Wilentz, R.E.; Iacobuzio-Donahue, C.A.; Argani, P.; McCarthy, D.M.; Parsons, J.L.; Yeo, C.J.; Kern, S.E.; Hruban, R.H. Loss of expression of Dpc4 in pancreatic intraepithelial neoplasia: Evidence that DPC4 inactivation occurs late in neoplastic progression. Cancer Res. 2000, 60, 2002–2006. [Google Scholar]
  188. Gu, G.; Dubauskaite, J.; Melton, D.A. Direct evidence for the pancreatic lineage: NGN3+ cells are islet progenitors and are distinct from duct progenitors. Development 2002, 129, 2447–2457. [Google Scholar]
  189. Kawaguchi, Y.; Cooper, B.; Gannon, M.; Ray, M.; MacDonald, R.J.; Wright, C.V. The role of the transcriptional regulator Ptf1a in converting intestinal to pancreatic progenitors. Nat. Genet. 2002, 32, 128–134. [Google Scholar] [CrossRef]
  190. Bardeesy, N.; Cheng, K.H.; Berger, J.H.; Chu, G.C.; Pahler, J.; Olson, P.; Hezel, A.F.; Horner, J.; Lauwers, G.Y.; Hanahan, D.; et al. Smad4 is dispensable for normal pancreas development yet critical in progression and tumor biology of pancreas cancer. Genes Dev. 2006, 20, 3130–3146. [Google Scholar] [CrossRef] [Green Version]
  191. Xu, X.; Ehdaie, B.; Ohara, N.; Yoshino, T.; Deng, C.X. Synergistic action of Smad4 and Pten in suppressing pancreatic ductal adenocarcinoma formation in mice. Oncogene 2010, 29, 674–686. [Google Scholar] [CrossRef] [Green Version]
  192. Jiang, J.; Hui, C.C. Hedgehog signaling in development and cancer. Dev. Cell 2008, 15, 801–812. [Google Scholar] [CrossRef] [Green Version]
  193. Hui, C.C.; Angers, S. Gli proteins in development and disease. Annu. Rev. Cell Dev. Biol. 2011, 27, 513–537. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Briscoe, J.; Therond, P.P. The mechanisms of Hedgehog signalling and its roles in development and disease. Nat. Revi. Mol. Cell Biol. 2013, 14, 416–429. [Google Scholar] [CrossRef] [PubMed]
  195. Jeng, K.S.; Chang, C.F.; Lin, S.S. Sonic Hedgehog Signaling in Organogenesis, Tumors, and Tumor Microenvironments. Int. J. Mol. Sci. 2020, 21, 758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Bai, Y.; Dong, J.; Li, Q.; Jin, Y.; Chen, B.; Zhou, M. Hedgehog Signaling in Pancreatic Fibrosis and Cancer. Medicine 2016, 95, e2996. [Google Scholar] [CrossRef]
  197. Gu, J.; Saiyin, H.; Fu, D.; Li, J. Stroma—A Double-Edged Sword in Pancreatic Cancer: A Lesson From Targeting Stroma in Pancreatic Cancer With Hedgehog Signaling Inhibitors. Pancreas 2018, 47, 382–389. [Google Scholar] [CrossRef]
  198. Hezel, A.F.; Kimmelman, A.C.; Stanger, B.Z.; Bardeesy, N.; Depinho, R.A. Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev. 2006, 20, 1218–1249. [Google Scholar] [CrossRef] [Green Version]
  199. Taipale, J.; Beachy, P.A. The Hedgehog and Wnt signalling pathways in cancer. Nature 2001, 411, 349–354. [Google Scholar] [CrossRef]
  200. Tian, H.; Callahan, C.A.; DuPree, K.J.; Darbonne, W.C.; Ahn, C.P.; Scales, S.J.; de Sauvage, F.J. Hedgehog signaling is restricted to the stromal compartment during pancreatic carcinogenesis. Proc. Natl. Acad. Sci. USA 2009, 106, 4254–4259. [Google Scholar] [CrossRef] [Green Version]
  201. Nolan-Stevaux, O.; Lau, J.; Truitt, M.L.; Chu, G.C.; Hebrok, M.; Fernandez-Zapico, M.E.; Hanahan, D. GLI1 is regulated through Smoothened-independent mechanisms in neoplastic pancreatic ducts and mediates PDAC cell survival and transformation. Genes Dev. 2009, 23, 24–36. [Google Scholar] [CrossRef] [Green Version]
  202. Hwang, R.F.; Moore, T.T.; Hattersley, M.M.; Scarpitti, M.; Yang, B.; Devereaux, E.; Ramachandran, V.; Arumugam, T.; Ji, B.; Logsdon, C.D.; et al. Inhibition of the hedgehog pathway targets the tumor-associated stroma in pancreatic cancer. Mol. Cancer Res. Mcr. 2012, 10, 1147–1157. [Google Scholar] [CrossRef] [Green Version]
  203. Steinway, S.N.; Zanudo, J.G.; Ding, W.; Rountree, C.B.; Feith, D.J.; Loughran, T.P., Jr.; Albert, R. Network modeling of TGFbeta signaling in hepatocellular carcinoma epithelial-to-mesenchymal transition reveals joint sonic hedgehog and Wnt pathway activation. Cancer Res. 2014, 74, 5963–5977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Inamoto, S.; Itatani, Y.; Yamamoto, T.; Minamiguchi, S.; Hirai, H.; Iwamoto, M.; Hasegawa, S.; Taketo, M.M.; Sakai, Y.; Kawada, K. Loss of SMAD4 Promotes Colorectal Cancer Progression by Accumulation of Myeloid-Derived Suppressor Cells through the CCL15-CCR1 Chemokine Axis. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2016, 22, 492–501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Albo, D.; Berger, D.H.; Vogel, J.; Tuszynski, G.P. Thrombospondin-1 and transforming growth factor beta-1 upregulate plasminogen activator inhibitor type 1 in pancreatic cancer. J. Gastrointest. Surg. Off. J. Soc. Surg. Aliment. Tract 1999, 3, 411–417. [Google Scholar] [CrossRef]
  206. Fang, Y.; Shen, Z.Y.; Zhan, Y.Z.; Feng, X.C.; Chen, K.L.; Li, Y.S.; Deng, H.J.; Pan, S.M.; Wu, D.H.; Ding, Y. CD36 inhibits beta-catenin/c-myc-mediated glycolysis through ubiquitination of GPC4 to repress colorectal tumorigenesis. Nat. Commun. 2019, 10, 3981. [Google Scholar] [CrossRef] [Green Version]
  207. Huiping, C.; Kristjansdottir, S.; Jonasson, J.G.; Magnusson, J.; Egilsson, V.; Ingvarsson, S. Alterations of E-cadherin and beta-catenin in gastric cancer. BMC Cancer 2001, 1, 16. [Google Scholar] [CrossRef] [Green Version]
  208. Joo, Y.E.; Rew, J.S.; Kim, H.S.; Choi, S.H.; Park, C.S.; Kim, S.J. Changes in the E-cadherin-catenin complex expression in early and advanced gastric cancers. Digestion 2001, 64, 111–119. [Google Scholar] [CrossRef]
  209. Miyazawa, K.; Iwaya, K.; Kuroda, M.; Harada, M.; Serizawa, H.; Koyanagi, Y.; Sato, Y.; Mizokami, Y.; Matsuoka, T.; Mukai, K. Nuclear accumulation of beta-catenin in intestinal-type gastric carcinoma: Correlation with early tumor invasion. Virchows Arch. Int. J. Pathol. 2000, 437, 508–513. [Google Scholar] [CrossRef]
  210. Aust, G.; Eichler, W.; Laue, S.; Lehmann, I.; Heldin, N.E.; Lotz, O.; Scherbaum, W.A.; Dralle, H.; Hoang-Vu, C. CD97: A dedifferentiation marker in human thyroid carcinomas. Cancer Res. 1997, 57, 1798–1806. [Google Scholar]
  211. Brown, R.S.; Bellisario, R.L.; Botero, D.; Fournier, L.; Abrams, C.A.; Cowger, M.L.; David, R.; Fort, P.; Richman, R.A. Incidence of transient congenital hypothyroidism due to maternal thyrotropin receptor-blocking antibodies in over one million babies. J. Clin. Endocrinol. Metab. 1996, 81, 1147–1151. [Google Scholar] [CrossRef] [Green Version]
  212. Post, Y.; Clevers, H. Defining Adult Stem Cell Function at Its Simplest: The Ability to Replace Lost Cells through Mitosis. Cell Stem Cell 2019, 25, 174–183. [Google Scholar] [CrossRef]
  213. Lazzari, E.; Butler, J.M. The Instructive Role of the Bone Marrow Niche in Aging and Leukemia. Curr. Stem Cell Rep. 2018, 4, 291–298. [Google Scholar] [CrossRef] [PubMed]
  214. Che, M.; Kweon, S.M.; Teo, J.L.; Yuan, Y.C.; Melstrom, L.G.; Waldron, R.T.; Lugea, A.; Urrutia, R.A.; Pandol, S.J.; Lai, K.K.Y. Targeting the CBP/beta-Catenin Interaction to Suppress Activation of Cancer-Promoting Pancreatic Stellate Cells. Cancers 2020, 12, 1476. [Google Scholar] [CrossRef]
  215. Giordano, M.; Cavallaro, U. Different Shades of L1CAM in the Pathophysiology of Cancer Stem Cells. J. Clin. Med. 2020, 9, 1502. [Google Scholar] [CrossRef] [PubMed]
  216. Ferrer, A.I.; Trinidad, J.R.; Sandiford, O.; Etchegaray, J.P.; Rameshwar, P. Epigenetic dynamics in cancer stem cell dormancy. Cancer Metastasis Rev. 2020, 33, 413–427. [Google Scholar] [CrossRef]
  217. Rahmani, W.; Sinha, S.; Biernaskie, J. Immune modulation of hair follicle regeneration. NPJ Regen. Med. 2020, 5, 9. [Google Scholar] [CrossRef]
  218. Siveen, K.S.; Prabhu, K.; Krishnankutty, R.; Kuttikrishnan, S.; Tsakou, M.; Alali, F.Q.; Dermime, S.; Mohammad, R.M.; Uddin, S. Vascular Endothelial Growth Factor (VEGF) Signaling in Tumour Vascularization: Potential and Challenges. Curr. Vasc. Pharmacol. 2017, 15, 339–351. [Google Scholar] [CrossRef]
  219. Isidori, A.M.; Venneri, M.A.; Fiore, D. Angiopoietin-1 and Angiopoietin-2 in metabolic disorders: Therapeutic strategies to restore the highs and lows of angiogenesis in diabetes. J. Endocrinol. Investig. 2016, 39, 1235–1246. [Google Scholar] [CrossRef]
  220. Sabir, S.; Saleem, A.; Akhtar, M.F.; Saleem, M.; Raza, M. Increasing beta cell mass to treat diabetes mellitus. Adv. Clin. Exp. Med. Off. Organ Wroc. Med. Univ. 2018, 27, 1309–1315. [Google Scholar] [CrossRef]
  221. Li, J.N.; Li, W.; Cao, L.Q.; Liu, N.; Zhang, K. Efficacy of mesenchymal stem cells in the treatment of gastrointestinal malignancies. World J. Gastrointest. Oncol. 2020, 12, 365–382. [Google Scholar] [CrossRef]
  222. Jiang, Z.; White, R.A.; Wang, T.C. Adult Pancreatic Acinar Progenitor-like Populations in Regeneration and Cancer. Trends Mol. Med. 2020, 26, 758–767. [Google Scholar] [CrossRef]
  223. Wang, W.; Bochtler, T.; Wuchter, P.; Manta, L.; He, H.; Eckstein, V.; Ho, A.D.; Lutz, C. Mesenchymal stromal cells contribute to quiescence of therapy-resistant leukemic cells in acute myeloid leukemia. Eur. J. Haematol. 2017, 99, 392–398. [Google Scholar] [CrossRef] [PubMed]
  224. Nam, H.S.; Capecchi, M.R. Lrig1 expression prospectively identifies stem cells in the ventricular-subventricular zone that are neurogenic throughout adult life. Neural Dev. 2020, 15, 3. [Google Scholar] [CrossRef] [PubMed]
  225. Kim, J.; Jo, Y.H.; Jang, M.; Nguyen, N.N.Y.; Yun, H.R.; Ko, S.H.; Shin, Y.; Lee, J.S.; Kang, I.; Ha, J.; et al. PAC-5 Gene Expression Signature for Predicting Prognosis of Patients with Pancreatic Adenocarcinoma. Cancers 2019, 11, 1749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Grant, F.M.; Yang, J.; Nasrallah, R.; Clarke, J.; Sadiyah, F.; Whiteside, S.K.; Imianowski, C.J.; Kuo, P.; Vardaka, P.; Todorov, T.; et al. BACH2 drives quiescence and maintenance of resting Treg cells to promote homeostasis and cancer immunosuppression. J. Exp. Med. 2020, 217. [Google Scholar] [CrossRef] [PubMed]
  227. Hossen, M.N.; Rao, G.; Dey, A.; Robertson, J.D.; Bhattacharya, R.; Mukherjee, P. Gold Nanoparticle Transforms Activated Cancer-Associated Fibroblasts to Quiescence. Acs Appl. Mater. Interfaces 2019, 11, 26060–26068. [Google Scholar] [CrossRef] [PubMed]
  228. Dauer, P.; Zhao, X.; Gupta, V.K.; Sharma, N.; Kesh, K.; Gnamlin, P.; Dudeja, V.; Vickers, S.M.; Banerjee, S.; Saluja, A. Inactivation of Cancer-Associated-Fibroblasts Disrupts Oncogenic Signaling in Pancreatic Cancer Cells and Promotes Its Regression. Cancer Res. 2018, 78, 1321–1333. [Google Scholar] [CrossRef] [Green Version]
  229. Norton, J.; Foster, D.; Chinta, M.; Titan, A.; Longaker, M. Pancreatic Cancer Associated Fibroblasts (CAF): Under-Explored Target for Pancreatic Cancer Treatment. Cancers 2020, 12, 1347. [Google Scholar] [CrossRef]
  230. Cai, Z.; Liang, Y.; Xing, C.; Wang, H.; Hu, P.; Li, J.; Huang, H.; Wang, W.; Jiang, C. Cancerassociated adipocytes exhibit distinct phenotypes and facilitate tumor progression in pancreatic cancer. Oncol. Rep. 2019, 42, 2537–2549. [Google Scholar] [CrossRef] [Green Version]
  231. Le, Y.; Gao, H.; Richards, W.G.; Zhao, L.; Bleday, R.; Clancy, T.; Zhu, Z. VentX expression in tumor associated macrophages promotes phagocytosis and immunity against pancreatic cancers. JCI Insight 2020, 5. [Google Scholar] [CrossRef]
  232. Sunami, Y.; Rebelo, A.; Kleeff, J. Lipid Metabolism and Lipid Droplets in Pancreatic Cancer and Stellate Cells. Cancers 2017, 10, 3. [Google Scholar] [CrossRef] [Green Version]
  233. Mu, W.; Wang, Z.; Zoller, M. Ping-Pong-Tumor and Host in Pancreatic Cancer Progression. Front. Oncol. 2019, 9, 1359. [Google Scholar] [CrossRef] [PubMed]
  234. Saison-Ridinger, M.; DelGiorno, K.E.; Zhang, T.; Kraus, A.; French, R.; Jaquish, D.; Tsui, C.; Erikson, G.; Spike, B.T.; Shokhirev, M.N.; et al. Reprogramming pancreatic stellate cells via p53 activation: A putative target for pancreatic cancer therapy. PLoS ONE 2017, 12, e0189051. [Google Scholar] [CrossRef] [PubMed]
  235. Ambrosini, G.; Dalla Pozza, E.; Fanelli, G.; Di Carlo, C.; Vettori, A.; Cannino, G.; Cavallini, C.; Carmona-Carmona, C.A.; Brandi, J.; Rinalducci, S.; et al. Progressively De-Differentiated Pancreatic Cancer Cells Shift from Glycolysis to Oxidative Metabolism and Gain a Quiescent Stem State. Cells 2020, 9, 1572. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Histological types of pancreatic cancer, based on References [15,16].
Figure 1. Histological types of pancreatic cancer, based on References [15,16].
Ijms 21 05656 g001
Figure 2. Overall signaling cascades, including the TGF-beta SMAD cascade (reproduced with permission of Reactome Pathway, Fabregat et al. [184]).
Figure 2. Overall signaling cascades, including the TGF-beta SMAD cascade (reproduced with permission of Reactome Pathway, Fabregat et al. [184]).
Ijms 21 05656 g002
Figure 3. Schematic of biogenesis and of the “off”—“on” Hedgehog signaling. Reproduced with permission from Reactome (https://reactome.org/PathwayBrowser/#/R-HSA-5358351).
Figure 3. Schematic of biogenesis and of the “off”—“on” Hedgehog signaling. Reproduced with permission from Reactome (https://reactome.org/PathwayBrowser/#/R-HSA-5358351).
Ijms 21 05656 g003
Figure 4. TSP-1-CD36 signaling is inducing apoptosis of tumor associated epithelial cells. As a result of TSP-1 binding to CD36 on microvascular endothelial cells, phosphorylation and, therefore, activation of P59fyn (cytoplasmic protein kinase) occur. This in turn stimulates caspase-like proteases, which induce the phosphorylation of MAPK. Nuclear translocation of MAPK generates increased expression of caspase-3 and of proapoptotic receptors, leading to apoptosis. Mitochondrial damage leads to the release of reactive oxygen species and of cytochrome C, which are also triggers of apoptosis. Moreover, the binding of THC-1 to CD36 induces the recruitment of SHP-1 to the VEGFR2 complex, followed by VEGFR2 dephosphorylation and inhibition of the VEGF pathway and leading to anti-angiogenesis. (Reproduced with permission from Reference [53].)
Figure 4. TSP-1-CD36 signaling is inducing apoptosis of tumor associated epithelial cells. As a result of TSP-1 binding to CD36 on microvascular endothelial cells, phosphorylation and, therefore, activation of P59fyn (cytoplasmic protein kinase) occur. This in turn stimulates caspase-like proteases, which induce the phosphorylation of MAPK. Nuclear translocation of MAPK generates increased expression of caspase-3 and of proapoptotic receptors, leading to apoptosis. Mitochondrial damage leads to the release of reactive oxygen species and of cytochrome C, which are also triggers of apoptosis. Moreover, the binding of THC-1 to CD36 induces the recruitment of SHP-1 to the VEGFR2 complex, followed by VEGFR2 dephosphorylation and inhibition of the VEGF pathway and leading to anti-angiogenesis. (Reproduced with permission from Reference [53].)
Ijms 21 05656 g004
Table 1. Current mechanisms that can convert normal fibroblasts into cancer-associated fibroblast.
Table 1. Current mechanisms that can convert normal fibroblasts into cancer-associated fibroblast.
Process/ChangesActivated MoleculesReferences
Activation Ligandstransforming growth factor-β (TGFβ),
platelet-derived growth factors (PDGFs), epidermal growth factors (EGFs),
fibroblast growth factors (FGFs),
bone morphogenic proteins (BMPs),
Sonic Hedgehog (Shh)
[103]
Contact Signalsnotch signaling[118]
Inflammatory ModulatorsIL-1 →NFkB and IL-6 →STAT transcription factors → JAK–STAT signaling → contractile cytoskeleton and histone acetylation[119,120]
Physical Changes in ECstiffness and composition[121,122,123]
Physiological Stressheat-shock factors[124]
Genomic Stress/DNA Damage—Chemo-/Radiotherapy ROS[106,125]
Table 2. Cancer-associated fibroblasts regulate cancer progression through a dynamic secretome.
Table 2. Cancer-associated fibroblasts regulate cancer progression through a dynamic secretome.
Molecules Released by CAFsModified Processes that Orchestrate Tumor Development and Immune EvasionReferences
VEGFs, PDGFs, HGF, IL-8, SDF-1enhanced angiogenesis[140,141]
IL-6, IL-1enhanced inflammation[142]
IL-6, TGFβ, SDF-1, HGF, lysophosphatidylcholines-LPCsenhanced cell proliferation[143,144,145]
TGFβ, COX-2/PGE2enhanced motility[146]
IL-6, CXCL 12macrophage switch and immune evasion[147]
HIF1a, lactate dehydrogenase Aaltered metabolism[148]
differentiation factors, activin A, FGF2altered cell fate[149]
TGFβ, HGF, FGFs, NGF, IGFenhanced secretion of cytokines and growth factors[150,151]
Fibronectin, collagen 1, tenascin C, osteopontin—MMPsdeposition and remodeling of ECM[141,152,153,154]
ECM = extracellular matrix.
Table 3. Mechanisms involved in quiescence of different pancreatic cancer cell types.
Table 3. Mechanisms involved in quiescence of different pancreatic cancer cell types.
Cell Type Positioning (Tumor or Associated)Facts (or Proposed Mechanisms)References
Stromal“Pancreatic cancer-associated fibroblasts”“Gold nanoparticle transforms activated cancer-associated fibroblasts to quiescence by enhancing lipid synthesis and lipid utilization”.[227]
“Anticancer compound Minnelide revealed deregulation of the TGFb signalling pathway in CAF,[228]
resulting in an apparent reversal of their activated state to a quiescent, nonproliferative state”.[229]
“This heterogeneity explains why one type of CAF is found to support cancer invasiveness and metastases while another type does not”.
“Pancreatic cancer-associated adipocyte”“Cancer-associated adipocytes exhibit distinct phenotypes and facilitate tumor progression in pancreatic cancer”, but quiescence was not examined.[230]
Pancreatic tumor-associated macrophages“The expression of homeobox protein VentX, a master regulator of macrophage plasticity, is significantly decreased in the PDA-TAMs”.[231]
Stellate cells“Up-regulation of Ppar-γ which is associated with quiescence”.[214]
“In the healthy pancreas, PSCs are in the quiescent state and retain vitamin A-containing lipid droplets”.[232,233]
“PSC, quiescent in the healthy pancreas. During pancreatic injury, PSC develop a myofibroblast phenotype expressing αSMA1, actively proliferate and migrate. Activation of PSC is promoted by TGFβ, HGF, FGF, EGF, and sHH”.[234]
“p53 activation by Nutlin-3a induces profound transcriptional changes, which reprogram activated PSCs to quiescence”.
Cancer stem cell Quiescent stem cells are characterized by high chemo-resistance, clonogenic ability, and metastatic potential.[235]
Pancreatic progenitor-like cell “Dclk1+ and Stmn+ cells are long-lived, largely quiescent, and lack proliferation under resting conditions”.[222]

Share and Cite

MDPI and ACS Style

Tanase, C.; Gheorghisan-Galateanu, A.-A.; Popescu, I.D.; Mihai, S.; Codrici, E.; Albulescu, R.; Hinescu, M.E. CD36 and CD97 in Pancreatic Cancer versus Other Malignancies. Int. J. Mol. Sci. 2020, 21, 5656. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21165656

AMA Style

Tanase C, Gheorghisan-Galateanu A-A, Popescu ID, Mihai S, Codrici E, Albulescu R, Hinescu ME. CD36 and CD97 in Pancreatic Cancer versus Other Malignancies. International Journal of Molecular Sciences. 2020; 21(16):5656. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21165656

Chicago/Turabian Style

Tanase, Cristiana, Ancuta-Augustina Gheorghisan-Galateanu, Ionela Daniela Popescu, Simona Mihai, Elena Codrici, Radu Albulescu, and Mihail Eugen Hinescu. 2020. "CD36 and CD97 in Pancreatic Cancer versus Other Malignancies" International Journal of Molecular Sciences 21, no. 16: 5656. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21165656

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop