Review
Cell-adhesion molecules and their soluble forms: Promising predictors of ‘‘tumor progression’’ and relapse in leukemia
Tumor Biology November 2018: 1–12 Ó The Author(s) 2018 Article reuse guidelines: sagepub.com/journals-permissions DOI: 10.1177/1010428318811525 journals.sagepub.com/home/tub
Ieda Cristina Cunha Ferreira e Fonseca1 , Felipe Andre´s Cordero da Luz1,2, Isadora Akemi Uehara1 and Marcelo Jose´ Barbosa Silva1
Abstract Some surface markers are used to discriminate certain leukemic subpopulations that retain a greater oncogenic potential than others, and, for this reason, they were termed as leukemic stem cells, similar to the concept of cancer stem cells in carcinoma. Among these surface markers are proteins involved in cell–cell adhesion or cell–matrix adhesion, and they may play a role in the relapse of leukemia, similar to metastasis in carcinomas. The most important are epithelial cadherin, neural cadherin, epithelial cell-adhesion molecule, and CD44, which can be cleaved and released, and their soluble forms were found increased in serum levels of cancer patients, being implicated, in some cases, with progression, metastases, and relapse. In this review, we highlighted the role of these four adhesion molecules in carcinomas and hematological malignancies, mainly leukemia, and discuss if the serum levels of soluble forms can be correlated with the surface protein status on the leukemic cells. Accession of the soluble forms looks attractive, but their use as markers in cancer must be studied in association with other parameters, as there are significant changes in levels in other pathological conditions besides cancer. Studies correlating the levels of the forms with the status of the membrane-bound proteins in leukemic (stem) cells and correlating those parameters with relapse in leukemia may afford important knowledge and applicability of those serum markers in clinical practice. For instance, the expression of the membrane-bound forms of these adhesion proteins may have promising clinical use in leukemia and other hematological malignancies. Keywords Cell-adhesion molecules, soluble forms, tumor progression, relapse, leukemia
Date received: 8 June 2018; accepted: 15 October 2018
Introduction Several cell-surface proteins are important in cancer, and hematological malignancies are not an exception. These cellular components are used for immunophenotyping, a procedure critical for diagnosis and treatment of leukemia and other hematological disorders. For example, the blast cell count (CD34+ cells) is critical in leukemia treatment, and monitoring that parameter serves as an indicator of active disease, minimal residual disease, and remission.1 Some patients, considered to be in morphological remission, had in excess of 5% leukemic blasts. Those patients considered resistant disease
have more than 15% blasts. Minimal residual disease (MDR) is considered if patients present less than 1% of 1
Laboratory of Tumor Biomarkers and Osteoimmunology, Institute of Biomedical Sciences, Federal University of Uberlaˆndia, Uberlaˆndia, Brazil 2 Nucleus of Cancer Prevention and Research, Cancer Hospital, Federal University of Uberlaˆndia, Uberlaˆndia, Brazil Corresponding author: Ieda Cristina Cunha Ferreira e Fonseca, Laboratory of Tumor Biomarkers and Osteoimmunology, Institute of Biomedical Sciences, Federal University of Uberlaˆndia, 2B 211, Umuarama, Uberlaˆndia 38405-320, Minas Gerais, Brazil. Email:
[email protected]
Creative Commons Non Commercial CC BY-NC: This article is distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 License (http://www.creativecommons.org/licenses/by-nc/4.0/) which permits non-commercial use, reproduction and distribution of the work without further permission provided the original work is attributed as specified on the SAGE and Open Access pages (https://us.sagepub.com/en-us/nam/open-access-at-sage).
2 blasts.2 However, other surface markers are important for clinical use, as some attribute different properties to leukemic blast cells, like CD123, and/or are lineage markers, like CD19.3 Several surface markers can be helpful for therapy strategy choice.4,5 In that trend of thought, some surface markers, such as CD123, lineage markers (CD2, CD5, CD7, CD11b, CD11c, CD19, CD22, CD56), C-type lectin-like molecule-1 (CLL-1), CD96, CD47, and T-cell immunoglobulin and mucin-domain containing-3 (TIM-3), are widely used to discriminate certain leukemic subpopulations that retain a greater oncogenic potential than others, and, for this reason, they were termed leukemic stem cells (LSCs), like the concept of cancer stem cells (CSCs) in carcinoma. In a general way, these LSCs retain several characteristics of normal hematopoietic stem cells (HSCs), such as the potential of generating ‘‘mature’’ leukemic cells. Notably, some studies found that mature leukemic cells can recapitulate stem properties, asserted by surface markers related with normal HSCs, like CD34 and eventually CD38. However, other surface markers besides those related to HSCs contribute to that oncogenic property.3 Some of these surface markers are proteins involved in cell–cell adhesion or cell–matrix adhesion. For example, CD34 participates in adhesion of HSCs with bone marrow matrix and stromal cells.6 HSCs interact with several stromal cells and extracellular matrix components in the bone marrow, and it is not different with leukemic cells, mainly those that retain stem cell–like properties.3,7 For example, CD44 is involved in bone marrow homing and also with stem cell properties in acute myeloid leukemia, and its depletion by antibodies resulted in the clearance of medullary and free leukemic cells in graft mice models.8 There are some organs and tissues, such as the central nervous system and bone marrow, that treatmentresistant leukemic cells are set and permanent in quiescence, rising after completion or in the interval of an aggressive treatment (relapse).1,9 In this matter, some of these adhesion molecules may play a role in relapse of leukemia,3,7 similar to metastasis in carcinomas. The most important and well-studied adhesion molecules that play a role in metastases are epithelial cadherin (E-cadherin), neural cadherin (N-cadherin), epithelial cell-adhesion molecule (EpCAM), and CD44. These membrane proteins exert critical roles in cell–cell or cell–matrix interaction as well as differentiation, motility, and metastasis.10–17 Those surface markers can be cleaved and released,13,18–20 and their soluble forms were found increased in serum levels of cancer patients,19,21–28 being implicated, in some cases, with progression and metastases.19 Moreover, the soluble forms of some of these surface markers increase in the serum of patients with hematological malignancies, such as leukemia.29–34
Tumor Biology As the source of the soluble forms are the membrane-bound proteins, one question can be raised: can the serum levels of soluble forms be correlated with the surface protein expression on the leukemic cells? In this review, we highlighted the role of these four adhesion molecules in carcinomas and hematological malignancies, mainly leukemia, and discuss evidence about this matter. In the first topic, we will briefly describe the biology of the four cell-adhesion molecules (CAMs) and biogenesis of the soluble forms. The second topic will be divided into two subtopics: one for carcinomas, briefly describing the role of membrane forms, then their soluble forms in tumor progression and metastasis and the second subtopic will focus on their role in leukocyte malignancies, mainly leukemia, and thereafter the findings about their soluble forms in hematological malignancies will be presented. The third topic will discourse about their non-specific role, like inflammation, and discriminate between general processes and cancer. Finally, in the fourth topic, we will resume the important data for this review and propose a hypothetical use of the soluble forms in leukemia.
Biology and biogenesis E-cadherin is a member of the cadherins (calciumdependent adhesion integrins) family, formerly found abundant in epithelial tissues, with an important role in cell–cell adhesion, like desmosomes and adherents junctions. It interacts with cadherin residues of other cells to establish homophilic interactions.35 E-cadherin also perform heterophilic interactions with immune cell through their E-cadherin ligands (CD103, killer cell lectin-like receptor subfamily G member 1 (KLRG1)), inhibiting immune cell activation and proliferation.35 Cadherins have important role in cell architecture by indirect interaction with cytoskeleton through their cytoplasmic (amino-terminal) domain. The cytoplasmic domain directly binds either b-catenin or g-catenin/plakoglobin that, in turn, mediate interaction with actin. In addition, p120 is another catenin member that interacts with the juxta membrane region of E-cadherin, strengthening its adhesive properties.36 In another perspective, the E-cadherin cytoplasmic domain acts as a docking area for b-catenin, thus repressing the proliferative function of b-catenin37 in an adhesion-independent manner.38 Loss of E-cadherin leads to b-catenin release, promoting cell proliferation, gain of motility, and other characteristics typical of a migrating cell.35 In addition to the wellknown role in inhibiting b-catenin transcriptional properties, E-cadherin also modulates phosphatidylinositol 3-Kinase (PI3K)/protein kinase B (Akt) signaling pathway, Ras homolog guanosine triphosphate
Fonseca et al. (Rho GTPase) and inhibits nuclear factor kB (NFkB) activity.35 N-cadherin is another member of the cadherins family. Similar to E-cadherin, N-cadherin plays a role in cell–cell adhesion, like desmosomes and adherents junctions of mesenchymal and neural cells and other nonepithelial cells.39 Its cytoplasmic domain, similar to Ecadherin, interact with catenin members, impacting cell architecture through indirect interaction with cytoskeleton.36 Similar to E-cadherin, N-cadherin represses the pro-proliferative properties of b-catenin in several cell types.39 However, in other cells, it was observed that N-cadherin interact with Wnt co-receptor LRP6, activating the Wnt/b-catenin pathway, and promote Akt activation, impeding b-catenin degradation,40 that, strikingly, occurs the opposite in other cell types.41 The soluble forms of these cadherins, that is, the extracellular domain of them is released by proteolytic cleavage activity of sheddases, like matrix metalloproteases (MMPs), membrane-type matrix metalloproteases (MT-MMPs), a disintegrin and metalloproteinases (ADAMs), kallikrein, plasmin, and other sheddases.18 By intuition, cleavage of E-cadherin and N-cadherin could release b-catenin, leading to increased cell proliferation and migration because they act as cytoplasmic docking of b-catenin.42 However, it was observed a counterintuitive role regarding this matter. It was observed that their intracellular domain inhibits b-catenin signaling.43 Also, it was observed that N-cadherin intracellular domain inhibits b-catenin but abolishes contact-dependent inhibition of cell proliferation and promotes anchorage-independent cell growth by modulating other pathways.44 It is important to state that these studies were performed by ectopically expressing a construct of the cytoplasmic domain only and can differ from the physiological cytoplasmic release (proteolytic cleavage). They can be intracellularly cleaved by caspase 3 or presenilin, releasing their cytoplasmic domain, and can be extracellularly by ADAM10, for example, releasing their extracellular domain. Interestingly, ADAM10, that is, extracellular cleavage of E-cadherin and N-cadherin leads to release and activation of b-catenin signaling.42 Yet, the proteolytic pathway of these cadherins, although could result in increased cell proliferation, has several differences regarding cell signaling. In accordance, presenilin could favor degradation of b-catenin. However, it was also observed that presenilin-mediated proteolysis of N-cadherin lead to nuclear translocation and transcriptional activity of b-catenin.42 Further discussion in this article will focus in extracellular proteolytic cleavage, as it releases the soluble form of the four cell-adhesion molecules presented. EpCAM is another abundant, calcium-independent, transmembrane glycoprotein that exerts homophilic cell–cell interaction in epithelial cells.45 Recent studies
3 observed that EpCAM is also involved in the b-catenin/Wnt (wingless-type MMTV integration site family) signaling pathway;46–48 cleavage of EpCAM by ADAM or presenilin leads to cell signaling by an EpCAM intracellular domain/FHL2 (four and a half LIM domains 2)/b-catenin complex,12 that result in cell proliferation,49,50 and release of the extracellular domain. Although EpCAM is an adhesion molecule, it could act as a negative dominant of E- and N-cadherin on cell– cell adhesion11,12 and has an important role in cell motility.10 For a review, refer the study performed by Schnell et al.51 CD44 is a transmembrane glycoprotein, and, different from previous, is involved in cell–extracellular matrix interaction. Its extracellular domain interacts can recruit matrix metalloproteinases to cell surface, that, in turn, cleaves cell surface and extracellular proteins, such as pro-form of transforming growth factor (TGF)-b, type IV collagen, and others. However, the most known interaction is with hyaluronic acid (HA). The intracellular domain is responsible of interactions with cell skeleton proteins, and, thus, can regulate cell shape and migration.52 By far, the most notorious roles of CD44 in cell migration is leukocyte recruitment and trans-endothelial migration. Lymphocytes use CD44-HA interactions to roll to inflamed sites. In contrast, neutrophils can firmly adhere by CD44-HA interaction. Several studies demonstrated that CD44 or HA impairment led to decrease leukocyte recruitment to disease.53 For more details about CD44 role in cellular biology, see Chen et al.54 Its cleavage by membrane type 1 matrix metalloproteases (MT1-MMP) and presenilin-1/y-secretase leads to extracellular domain release (soluble CD44) and translocation of the intracellular domain that acts as a transcription factor, increasing expression of CD44 itself, inflammation, survival, and stem markers, depending on context.13
Roles in cancer Solid cancer Membrane-bound proteins. Epithelial-to-mesenchymal transition (EMT) is the most important mechanism that allows epithelial malignant cells to migrate, leading to distant establishment of a secondary tumor (metastasis). During this cell reprogramming process, epithelial cells, which are naturally attached to their tissue, lose their epithelial markers, like E-cadherin, and gain mesenchymal characteristics, like N-cadherin expression, being able to detach from tissue and migrate to a distant site where they settle and return to an epithelial state by the reverse event, mesenchymal-to-epithelial transition (MET). Several transcription factors regulate EMT and MET.14,16 Although traditional EMT is recognized as loss of E-cadherin and gain of
4 N-cadherin, co-existence of E-cadherin and N-cadherin, by only N-cadherin upregulation, occurs in a process named partial EMT.15 EpCAM, first known as epithelial specific antigen (ESA), was first described upregulated in colorectal cancer with clinical relevance.55 EpCAM+ cells have a greater potential to initiate tumor engraftment. Al-Hajj et al.56 demonstrated that breast CSCs (CD24–/CD44+) EpCAM+ cells are capable of inducing tumors in mice, different than their EpCAM– counterparts. These findings apply to other cancer tumor-initiating cells such as colorectal, ovarian, and hepatocellular carcinoma CSCs.57–60 Also, CSCs EpCAM expression has been associated with therapy resistance and tumor bulk maintenance.56,58,59,61 Importantly, EpCAM is one of the most reliable markers of circulating tumor cells (CTCs) in various carcinomas, and the presence of EpCAM+ CTCs is related to poor outcome, like colorectal cancer, head and neck cancer, ovarian cancer, and breast cancer.62–67 CD44 is another CAM abundant in CSCs13,17,68,69 and is involved in cell motility, invasion, angiogenesis, and metastasis.13,17 Its intracellular domain that remains after extracellular domain cleavage plays an important role in cell signaling to promote metastasis.13 CD44 is widely expressed as the common (standard) isoform (CD44s), but it has several variants (CD44vs) that interact with different growth factors. For example, CD44v3 binds to fibroblast growth factor 2 (FGF2), and CD44v6 can bind to vascular endothelial growth factor (VEGF), osteopontin (OPN), and hepatocyte growth factor (HGF). These properties of CD44 variants have a great impact in pro-tumoral features.70 Interestingly, CD44s and CD44vs have particular roles in different cancers.13,69 Therefore, these CAMs have important roles in carcinoma progression. E-cadherin loss is the most common event in cancer progression, but increases in N-cadherin, EpCAM, and CD44, independent of increases in E-cadherin, are of greater importance, as these three molecules are involved with invasion, motility, and stemness. Soluble proteins. Soluble epithelial cadherin (sE-cadherin) plays an important role in cancer progression, since several signaling pathways associated with tumor progression are activated, like epidermal growth factor receptor (EGFR) family members (EGFR and human epidermal growth factor receptor-type 2 (HER2)) and insulin-like growth factor 1 (IGF-1R), thus activating mitogen-activated protein kinase (MAPK)/extracellular signal–regulated kinases (ERKs) and PI3K/Akt signaling. Also, the antitumor signaling pathway Hippo is inactivated. sE-cadherin changes cytotoxic cell activity by binding to KLRG1 of natural killer (NK) and
Tumor Biology TCD8+ cells, maintaining the cells in an inactive state19 by triggering the inhibitory signaling of KLRG1 similar to membrane-bound E-cadherin.35 Interestingly, sEcadherin levels have a positive correlation with cancer progression, with higher levels in advanced/metastatic states compared to initial stages and decrease in breast cancer patients after chemotherapy.19 Moreover, increases in sE-cadherin and their levels serve as predictive of relapse in prostate and bladder cancer.71,72 This corroborates with the hypothesis that tumors may be the main source of sE-cadherin. Importantly, sE-cadherin inhibits the adherent role of E-cadherin in cell– cell adhesion by binding with E-cadherin, dislocating membrane-bound E-cadherin homophilic interaction between two cells.18,19 So, even if primary tumors express high levels of E-cadherin that indicate less chance of metastasis, the association with higher levels of sE-cadherin can be associated with elevated chances of metastasis by all signaling that it triggers and by inhibiting E-cadherin homophilic interaction.19 Soluble neural cadherin (sN-cadherin) stimulates migration of endothelial cells, angiogenesis via fibroblast growth factor receptor (FGFR) activation, neurite outgrowth, and perineural invasion in prostate cancer.18 Increased levels of serum sN-cadherin in patients with bone sarcoma disease and soft tissue sarcoma, compared to healthy controls, were also observed and correlated with tumor stage and grade, respectively.24 Although scarce knowledge exists about sN-cadherin in tumor progression, the information suggests an important role for mesenchymal and endothelial cells in angiogenesis and invasion. Regarding soluble epithelial cell-adhesion molecule (sEpCAM), although the intracellular domain (EpICD) has the main role of EpCAM in cancer, the extracellular domain of EpCAM/soluble form (EpEX) enhances the cleavage of EpCAM, leading to intracellular signaling by EpICD.12 In accordance, elevated sEpCAM levels in ascites associated with ovarian tumor were correlated with positive cytology for malignancy.26 It was associated with shorter overall survival in patients with ovarian cancer treated with catumaxomab, a bispecific antibody that targets EpCAM.25 Elevated urinary sEpCAM concentrations were observed in bladder cancer and found as an independent indicator of poor prognosis,22 and elevated serum concentrations were also observed in epithelial ovarian cancer.27 Proteolytic cleavage of CD44 is important for cell signaling by its intracellular domain.13 Yet, the importance and biological functions of the soluble form (extracellular domain) of CD44 (sCD44) is not known. However, correlations between serum levels of sCD44 and tumor progression in colorectal cancer28 and overall survival in HER2-overexpressing breast cancer patients21 and in pre-malignant lesions of cervical cancer23 were observed.
Fonseca et al. Therefore, although not well elucidated, the soluble forms of E-cadherin, N-cadherin, EpCAM, and CD44 play important roles in cancer progression and metastasis, and some of them act as ligands of several receptors involved in this process. Moreover, several studies demonstrated that these soluble forms originate from the tumor tissue and are suggestive of tumor status and progression. Thus, it is reasonable to hypothesize that something similar occurs in hematological malignancies.
Hematological malignancies Membrane-bound proteins. Regarding hematological diseases, E-cadherin can be modulated by genetic and epigenetic events in leukemias. CDH1 (E-cadherin coding gene) is widely repressed by epigenetic alterations (methylation), leading to excess of leukemic blast cells and poor prognosis in acute lymphoblastic leukemia (ALL), chronic myeloblastic leukemia (CML), acute myeloblastic leukemia (AML), and acute promyelocytic leukemia (APL)73–76 and has a trend of correlation with N/K-RAS mutations in AML.75 Concordantly, restoration of E-cadherin expression (with inhibitors of histone deacetylases) in leukemic cells led to cell homophilic adhesion77 and downregulation of proliferation markers.78 Interestingly, different from previously mentioned leukemias that repress E-cadherin expression, it was observed that erythroid AML expresses surface Ecadherin.79 It was observed that N-cadherin is important for LSC homing to the marrow niche.80 In concordance, in AML, N-cadherin+ cells were enriched in the CD34+ cell compartment (LSCs), retained stem cell–like properties, and had greater oncogenic potential compared to N-cadherin– (CD34+ LSCs) cells.81 Also, N-cadherin is the most abundant cadherin expressed by T-cell lymphomas and other leukemic lineage cells, promoting homophilic N-cadherin interaction with mesenchymal cells.82 One study observed an increase of cytoplasmic stabilized b-catenin in B acute lymphoblastic leukemia (BALL) with (1;19) (q23;13) translocation. Interestingly, it was observed that b-catenin was docking in the Ncadherin intracellular domain, increasing N-cadherin levels and reinforcing the interaction between leukemic cell and bone marrow stroma.20 In another perspective, the expression of E-cadherin and N-cadherin by stromal cells have an important role in hematological malignancies. In mice, CDH1 and CDH2 (N-cadherin coding gene), as well as their coding proteins, are expressed by epithelial and mesenchymal thymus stromal cells, respectively. It was observed a biphasic expression pattern of expression of CDH1 and CDH2, as well as their proteins, in thymic stromal cell of mice models of thymic lymphoma: during acute
5 gamma-irradiation, an upregulation is observed before establishment of lymphoma, but, when efficiently established, a strong loss of their expression is observed.83 Because human thymic stromal cells exert heterophilic interaction with CD103+ thymocytes through E-cadherin, resulting in a positive regulation of cell proliferation,84 De Yzaguirre et al.83 suggest a pro-tumoral role of stromal cadherin upregulation at initial stages, with downregulation during lymphoma progression. These data altogether suggest an important role of cadherin expression not only by hematologic cells but also by stromal cell during progression of hematological malignancies. The scientific literature presents a few studies regarding EpCAM in hematological malignancies. One study demonstrated that EpCAM is frequently overexpressed in leukemic cells of AML patients, compared to healthy bone marrow cells, and these cells exhibit enhanced chemoresistance and oncogenesis. They found that anti-EpCAM antibody depleted subcutaneous, disseminated, and intramedullary engrafted mice with AML cells, suggesting that this adhesion molecule may be used as a therapeutic target. Interestingly, it was observed that EpCAM is enriched in AML LSCs (CD34+) and EpCAM+ cells co-express several LSC markers, like CD123 and CD44. Importantly, it was observed that EpCAMhi AML cells also express high levels of CD44.85 CD44v are widely expressed in stem cells, like HSCs, in stark contrast to differentiated cells. Leukemic cells, mainly LSCs, overexpress CD44 in resemblance to HSCs but mostly the variant isoforms.70 As mentioned previously, CD44, mainly CD44v, bind to several growth factors, and this characteristic supports leukemic cell apoptosis resistance, drug resistance, and proliferation.70 One study observed that CD44 plays an important role in the establishment of leukemic cells in AML. A specific antibody against CD44 was used to treat leukemia in mice models. They demonstrated that mice treated with the antibody did not develop the tumor compared to mice not treated with the antibody, and the medullary portion did not present with any leukemic cells. Also, they observed that CD44 is enriched with LSCs.8 Although being observed in gastrointestinal lymphoma, it is interesting that weak CD44 expression was associated with better prognosis than strong CD44 expression.86 It can be appreciated that these adhesion molecules in hematological malignancies have roles similar to carcinomas. For example, E-cadherin loss in AML led to increases in blasts, but its expression is of concern in histiocytosis, plasmacytomas, and malignancies of leukocytic origin that allocate in epithelial tissue, as might be expected by their adherent nature. Also, N-cadherin is involved with homing to bone marrow (mesenchymal tissue), like the metastasis process. And, finally, CD44
6 and EpCAM are linked with stemness, motility, and therapy resistance in leukemias. These data demonstrate a great resemblance with carcinomas (Figure 1).
Tumor Biology Soluble protein. sE-cadherin seems to participate in leukemia development. Patients with ALL, acute nonlymphocytic leukemia, CML, myelodysplastic
Figure 1. Role of E-cadherin, N-cadherin, EpCAM and CD44 in leukemic progression. (a) Interaction of adhesion molecules evokes several cell signaling related to aggressiveness, like invasiveness and migration in leukemic cells (black arrows). Proteolytic cleavage leads to increase of the soluble forms of adhesion molecules, participating in disease progression by acting as ligand and releasing the intracellular domain of these domains, evoking different cell signaling (gray arrows). (b) Evolution of leukemic disease and hypothetical role of adhesion molecules in progression. At primary disease, therapy eventually lead to selection and expansion of resistant clones (relapse), with de novo increase of serum forms of adhesion molecules, and/or pressure to quiescence. Adhesion molecules may play important role allowing homing and invasion to extramedullary tissue, cell–cell and cell–matrix adhesion, until quiescence breakdown and relapse.
Fonseca et al. syndrome (MDS), and non-Hodgkin lymphoma (NHL) presented with increased serum sE-cadherin than healthy controls and were positively correlated with stages and progression. An important finding is that serum sE-cadherin in blast crisis of CML is higher than in the chronic phase.34 Another study observed similar findings in a cohort of MDS patients: elevated serum sE-cadherin correlates with poor prognosis.30 Moreover, in newly diagnosed multiple myeloma (MM), high serum sE-cadherin levels (~six-fold) were observed compared to healthy controls and correlated with lactate dehydrogenase (LDH) levels, a non-specific tumor serum marker that was found as an independent prognosis factor of survival.31 These data suggest that sE-cadherin may be produced from malignant hematological cells and in accordance with the abundance of these cells. Also, it may be associated with proliferation and extramedullary infiltration of malignant cells.30,31,34 Increases in serum sCD44 were observed in pediatric ALL, independent of lineage, and decreased with treatment and remission to levels similar to that of healthy controls, without correlation between sCD44 and LDH levels, but had a weak correlation with peripheral leukocyte counts.33 In addition, increases in serum sCD44 levels were correlated with Hodgkin’s disease (HD), NHL, Burkitt’s lymphoma (BL), and ALL, but not AML, compared to healthy controls, with decreases to normal values in complete remission. There was positive correlation with stage and poor prognosis in HD and NHL, and positive correlation between serum sCD44 levels with tumor tissue CD44 levels in HD and BL.32 Another study observed similar levels in serum sCD44 in newly diagnosed children and adults with BALL compared to healthy controls. However, sCD44 was higher in patients with white blood cells (WBCs) .50 3 109/L than patients with WBC \50 3 109/L and in patients with a peripheral blast percentage .50 compared to patients with a peripheral blast percentage \50, presence of splenomegaly, extramedullary involvement, and in some cytogenetic conditions.29 Elevation in serum sCD44 levels was detected in patients with MDS compared to healthy controls and correlated with poor prognosis.30 These data suggest that sCD44 has its origins in tumors by proteolytic cleavage of the CD44, making it a potential reliable biomarker of clinical monitoring and prognosis marker in hematological diseases.29,32 As far as we know, no data are available about serum levels of sN-cadherin and sEpCAM in hematological malignancies. Despite scarce data, the soluble forms of E-cadherin and CD44 are indicative of active disease in hematological malignancies and play roles in progression of these cancers.
7
Soluble proteins: noise, non-specific, or something else? The soluble forms of these adhesion molecules are mostly studied in cancer by their potential role as biomarkers, because most of them are derived from tumor tissue surface proteins. However, an important question arises: can their serum levels vary in other diseases? As an example, bacterial proteases can cleave E-cadherin, producing sE-cadherin.87 Thus, its ‘‘unspecific’’ role must be elucidated. Elevated serum sE-cadherin levels was observed in bacterial and viral infections, organ failure, and benign tumors, and increased levels were observed in psoriasis, acute pancreatitis, diabetes, diabetic nephropathy, HIV infection, and sepsis.87 It is important to point out that echinococcosis alters hepatic parenchyma that could be misinterpreted as hepatic cancer. One study observed lower plasma sEcadherin concentrations in patients with echinococcosis compared to healthy controls, with a positive correlation between sE-cadherin levels and CDH1 expression by quantitative polymerase chain reaction (qPCR) from leukocytes. In that study, no difference was observed in sN-cadherin plasma concentrations of patients with echinococcosis. In this scenario, the authors suggest sEcadherin as a potential differential marker between echinococcosis and hepatic cancer, which has elevated sE-cadherin levels.88 These data are of importance because some cases of echinococcosis clinically mimic hepatic malignancy.89 The adhesion molecule sCD44 did not change in serum from patients with bacterial infection compared to healthy controls.33 However, sCD44 levels increased in the acute phase of Trypanosoma cruzi infection compared to healthy controls, but levels decreased in the chronic phase with advanced cardiopathy compared to healthy controls and patients with milder disease.90 Recently, it was observed that sCD44 increases in hepatic steatosis91 and chronic and aggressive periodontal disease,92 and its levels correlate with the evolution of endometriosis.93 One study analyzed sEpCAM in endometriosis, and did not find any correlation.94 Thus, the soluble forms of these adhesion molecules suffer significant changes in other pathological conditions besides cancer.
Leukemic relapses: ‘‘metastasis’’ of white blood cell cancer? As mentioned previously, these CAMs appear to have roles in hematological malignancies. Interestingly, some studies observed that the transcription factors that regulate EMT and MET in carcinomas have a role in hematological malignancies, including leukemia.
8 Importantly, it was observed that CML LSCs are marked by ‘‘EMT’’ transcription factors.95 Thus, it is not illogical that other similar gene control and cell reprogramming processes in carcinomas occur in leukemia. So, monitoring traditional surface markers of carcinomas may have applicability in leukemia. Importantly, as mentioned previously, EpCAM is one of the most reliable markers of CTCs in various carcinomas,62–66 and, intriguingly, it appears to have a similar role in AML.85 In carcinomas, the preconized cancer progression has metastasis as ‘‘the final step.’’ As reviewed in this article, some processes are critical for this downregulation of E-cadherin and/or upregulation of N-cadherin with concomitant upregulation of CD44 and, eventually, EpCAM. These processes promote cell motility, invasion, and migration as well as cell adhesion with endothelial cells and mesenchymal tissue, allowing cell attachment in bone, lung, cerebral, and other tissues as well as stemness, therapy resistance, and general cell robustness. Hiding of leukemic cells in ‘‘sanctuaries’’ can be related to the metastasis process of carcinoma, if looked at closely. However, quiescence of LSCs is a common mechanism involved in leukemic recurrence,96,97 that is rarely involved with metastasis in solid cancer, although quiescence is a property of CSC, cells that poses strong metastatic features.98 Interestingly, it was observed that N-cadherins are important for quiescence and self-renewal in several adults stem cells.99 E-cadherin, although a marker of epithelial (differentiated) cells, as well has a role in quiescence and self-renewal in adults stem cells, including neuronal stem cells.99 EpCAM is widely expressed in tumor-initiating cells (also known as CSCs) that are almost quiescent (EpCAM and its potential role in tumor-initiating cells). CD44 is important for quiescence and differentiation in neuronal100 and other stem cells101 and leukemia as well.70 Interestingly, it was observed that reduced b-catenin increases adhesiveness, quiescence, and cadherin expression, including CDH1 in HSCs.102 As aforementioned, Eand N-cadherins and EpCAM can stabilize and inhibit b-catenin signaling. So, these adhesion molecules could have a role in hiding and quiescence of leukemic cells, thus being factors associated with recurrence. De novo expression of CAMs or halt of proteolysis of CAM may be involved in this process. In accordance with this trend of thought, degradation of several surface molecules, such as integrins, by MMPs from granulocyte-colony stimulating factor (G-CSF)-induced neutrophils lead to HSCs quiescence break and mobilization to bloodstream, which is also observed in xenograft models of leukemia.96 In this context, the soluble forms of these adhesion molecules probably have predictive value in relapse and may have use for relapse monitoring in leukemia, as occured in several carcinomas.71,72
Tumor Biology In this trend of thought, one study observed an association between the percentage of cells expressing CD44 and the occurrence of disease relapse in children with B-cell progenitor ALL lacking adverse or favorable genetics.103 Importantly, CD44 was observed in extramedullary manifestations of myeloid leukemia.104 These data support the hypothesis that these classic adhesion molecules with great importance in carcinoma metastasis have a role in leukemic relapse and invasiveness.
Concluding remarks The adhesion molecules, mainly E-cadherin, N-cadherin, CD44, and EpCAM, may have similar roles in the infiltration and relapse of leukemia as their roles in the metastases of carcinomas. Accession of the soluble forms looks attractive, since they are produced from the membrane-bound forms of leukemic cells. However, there are significant changes in levels in other pathological conditions besides cancer. For this reason, their use as markers in leukemia must be studied in association with clinical data, leukemia type, and specific mutations and translocations. Yet, studies correlating the levels of the forms with the status of the membrane-bound proteins in LSCs and correlating those parameters with relapse in leukemia may afford important knowledge and applicability of those serum markers in clinical practice. Declaration of conflicting interests The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Funding The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: Pro-Rectory of Research and Post-Graduation of the Federal University of Uberlaˆndia funded the fee related with this publication.
ORCID iD Ieda Cristina Cunha Ferreira e Fonseca https://orcid.org/ 0000-0001-9694-7888 https://orcid.org/0000-0002Marcelo Jose´ Barbosa Silva 5807-4286
References 1. Health Quality Ontario. Minimal residual disease evaluation in childhood acute lymphoblastic leukemia: a clinical evidence review. Ont Health Technol Assessment Ser 2016; 16: 1–52.
Fonseca et al. 2. Inaba H, Coustan-Smith E, Cao X, et al. Comparative analysis of different approaches to measure treatment response in acute myeloid leukemia. J Clin Oncol 2012; 30: 3625–3632. 3. Lutz C, Hoang VT, Buss E, et al. Identifying leukemia stem cells—is it feasible and does it matter? Cancer Lett 2013; 338: 10–14. 4. Rodrigues CA, Goncalves MV, Ikoma MR, et al. Diagnosis and treatment of chronic lymphocytic leukemia: recommendations from the Brazilian Group of Chronic Lymphocytic Leukemia. Rev Bras Hematol Hemoter 2016; 38: 346–357. 5. Walter RB, Appelbaum FR, Estey EH, et al. Acute myeloid leukemia stem cells and CD33-targeted immunotherapy. Blood 2012; 119: 6198–6208. 6. Nielsen JS and McNagny KM. Novel functions of the CD34 family. J Cell Sci 2008; 121: 3683–3692. 7. Rashidi A and DiPersio JF. Targeting the leukemiastroma interaction in acute myeloid leukemia: rationale and latest evidence. Ther Adv Hematol 2016; 7: 40–51. 8. Jin L, Hope KJ, Zhai Q, et al. Targeting of CD44 eradicates human acute myeloid leukemic stem cells. Nat Med 2006; 12: 1167–1174. 9. Zhou J and Chng WJ. Identification and targeting leukemia stem cells: the path to the cure for acute myeloid leukemia. World J Stem Cells 2014; 6: 473–484. 10. Gaiser MR, Lammermann T, Feng X, et al. Cancer-associated epithelial cell adhesion molecule (EpCAM; CD326) enables epidermal Langerhans cell motility and migration in vivo. Proc Natl Acad Sci U S A 2012; 109: E889–E897. 11. Litvinov SV, Balzar M, Winter MJ, et al. Epithelial cell adhesion molecule (Ep-CAM) modulates cell-cell interactions mediated by classic cadherins. J Cell Biol 1997; 139: 1337–1348. 12. Maetzel D, Denzel S, Mack B, et al. Nuclear signalling by tumour-associated antigen EpCAM. Nat Cell Biol 2009; 11: 162–171. 13. Senbanjo LT and Chellaiah MA. CD44: a multifunctional cell surface adhesion receptor is a regulator of progression and metastasis of cancer cells. Front Cell Dev Biol 2017; 5: 18. 14. Cichon MA, Nelson CM and Radisky DC. Regulation of epithelial-mesenchymal transition in breast cancer cells by cell contact and adhesion. Cancer Inform 2015; 14: 1–13. 15. Grigore AD, Jolly MK, Jia D, et al. Tumor budding: the name is EMT. Partial EMT. J Clin Med 2016; 5: E51. 16. Chou YS and Yang MH. Epithelial-mesenchymal transition-related factors in solid tumor and hematological malignancy. J Chin Med Assoc 2015; 78: 438–445. 17. Thapa R and Wilson GD. The importance of CD44 as a stem cell biomarker and therapeutic target in cancer. Stem Cells Int 2016; 2016: 2087204. 18. De Wever O, Derycke L, Hendrix A, et al. Soluble cadherins as cancer biomarkers. Clin Exp Metastasis 2007; 24: 685–697.
9 19. Hu QP, Kuang JY, Yang QK, et al. Beyond a tumor suppressor: soluble E-cadherin promotes the progression of cancer. Int J Cancer 2016; 138: 2804–2812. 20. Nygren MK, Dosen-Dahl G, Stubberud H, et al. Betacatenin is involved in N-cadherin-dependent adhesion, but not in canonical Wnt signaling in E2A-PBX1-positive B acute lymphoblastic leukemia cells. Exp Hematol 2009; 37: 225–233. 21. Baek JM, Jin Q, Ensor J, et al. Serum CD44 levels and overall survival in patients with HER2-positive breast cancer. Breast Cancer Res Treat 2011; 130: 1029–1036. 22. Bryan RT, Shimwell NJ, Wei W, et al. Urinary EpCAM in urothelial bladder cancer patients: characterisation and evaluation of biomarker potential. Br J Cancer 2014; 110: 679–685. 23. Dasari S, Rajendra W and Valluru L. Evaluation of soluble CD44 protein marker to distinguish the premalignant and malignant carcinoma cases in cervical cancer patients. Med Oncol 2014; 31: 139. 24. Niimi R, Matsumine A, Iino T, et al. Soluble neuralcadherin as a novel biomarker for malignant bone and soft tissue tumors. BMC Cancer 2013; 13: 309. 25. Seeber A, Braicu I, Untergasser G, et al. Detection of soluble EpCAM (sEpCAM) in malignant ascites predicts poor overall survival in patients treated with catumaxomab. Oncotarget 2015; 6: 25017–25023. 26. Seeber A, Martowicz A, Spizzo G, et al. Soluble EpCAM levels in ascites correlate with positive cytology and neutralize catumaxomab activity in vitro. BMC Cancer 2015; 15: 372. 27. Tas F, Karabulut S, Serilmez M, et al. Clinical significance of serum epithelial cell adhesion molecule (EPCAM) and vascular cell adhesion molecule-1 (VCAM-1) levels in patients with epithelial ovarian cancer. Tumour Biol 2014; 35: 3095–3102. 28. Yamane N, Tsujitani S, Makino M, et al. Soluble CD44 variant 6 as a prognostic indicator in patients with colorectal cancer. Oncology 1999; 56: 232–238. 29. Amirghofran Z, Asiaee E and Kamazani FM. Soluble CD44 and CD44v6 and prognosis in children with B-cell acute lymphoblastic leukemia. Asia Pac J Clin Oncol 2016; 12: e375–e382. 30. Loeffler-Ragg J, Steurer M, Ulmer H, et al. Elevated levels of serum CD44 and E-cadherin predict an unfavourable outcome in myelodysplastic syndromes. Leukemia 2006; 20: 2064–2067. 31. Syrigos KN, Harrington KJ, Karayiannakis AJ, et al. Circulating soluble E-cadherin levels are of prognostic significance in patients with multiple myeloma. Anticancer Res 2004; 24: 2027–2031. 32. Tacyildiz N, Cavdar AO, Yavuz G, et al. Serum levels and differential expression of CD44 in childhood leukemia and malignant lymphoma: correlation with prognostic criteria and survival. Pediatr Int 2001; 43: 354–360. 33. Takeuchi M, Tanizawa A, Fukumoto Y, et al. Serum soluble CD44 in pediatric patients with acute leukemia. J Pediatr Hematol Oncol 1999; 21: 384–388.
10 34. Takubo T, Kanashima H, Terada Y, et al. Clinical significance of serum E-cadherin levels in patients with haematological malignancies. Haematologia 2002; 31: 319–325. 35. Van den Bossche J, Malissen B, Mantovani A, et al. Regulation and function of the E-cadherin/catenin complex in cells of the monocyte-macrophage lineage and DCs. Blood 2012; 119: 1623–1633. 36. Anastasiadis PZ and Reynolds AB. The p120 catenin family: complex roles in adhesion, signaling and cancer. J Cell Sci 2000; 113(Pt 8): 1319–1334. 37. Stockinger A, Eger A, Wolf J, et al. E-cadherin regulates cell growth by modulating proliferation-dependent betacatenin transcriptional activity. J Cell Biol 2001; 154: 1185–1196. 38. Gottardi CJ, Wong E and Gumbiner BM. E-cadherin suppresses cellular transformation by inhibiting betacatenin signaling in an adhesion-independent manner. J Cell Biol 2001; 153: 1049–1060. 39. Marie PJ and Hay E. Cadherins and Wnt signalling: a functional link controlling bone formation. Bonekey Rep 2013; 2: 330. 40. Zhang J, Shemezis JR, McQuinn ER, et al. AKT activation by N-cadherin regulates beta-catenin signaling and neuronal differentiation during cortical development. Neural Dev 2013; 8: 7. 41. Hay E, Nouraud A and Marie PJ. N-cadherin negatively regulates osteoblast proliferation and survival by antagonizing Wnt, ERK and PI3K/Akt signalling. PLoS ONE 2009; 4: e8284. 42. Heuberger J and Birchmeier W. Interplay of cadherinmediated cell adhesion and canonical Wnt signaling. Cold Spring Harbor Perspect Biol 2010; 2: a002915. 43. Sadot E, Simcha I, Shtutman M, et al. Inhibition of betacatenin-mediated transactivation by cadherin derivatives. Proc Natl Acad Sci U S A 1998; 95: 15339–15344. 44. Ozawa M. The N-cadherin cytoplasmic domain confers anchorage-independent growth and the loss of contact inhibition. Sci Rep 2015; 5: 15368. 45. Litvinov SV, Velders MP, Bakker HA, et al. Ep-CAM: a human epithelial antigen is a homophilic cell-cell adhesion molecule. J Cell Biol 1994; 125: 437–446. 46. Yamashita T, Budhu A, Forgues M, et al. Activation of hepatic stem cell marker EpCAM by Wnt-beta-catenin signaling in hepatocellular carcinoma. Cancer Res 2007; 67: 10831–10839. 47. Gostner JM, Fong D, Wrulich OA, et al. Effects of EpCAM overexpression on human breast cancer cell lines. BMC Cancer 2011; 11: 45. 48. Zhou FQ, Qi YM, Xu H, et al. Expression of EpCAM and Wnt/beta-catenin in human colon cancer. Genet Mol Res 2015; 14: 4485–4494. 49. Park SY, Bae JS, Cha EJ, et al. Nuclear EpICD expression and its role in hepatocellular carcinoma. Oncol Rep 2016; 36: 197–204. 50. Jachin S, Bae JS, Sung JJ, et al. The role of nuclear EpICD in extrahepatic cholangiocarcinoma: association with beta-catenin. Int J Oncol 2014; 45: 691–698. 51. Schnell U, Cirulli V and Giepmans BN. EpCAM: structure and function in health and disease. Biochim Biophys Acta 2013; 1828: 1989–2001.
Tumor Biology 52. Ponta H, Sherman L and Herrlich PA. CD44: from adhesion molecules to signalling regulators. Nat Rev Mol Cell Biol 2003; 4: 33–45. 53. McDonald B and Kubes P. Interactions between CD44 and hyaluronan in leukocyte trafficking. Front Immunol 2015; 6: 68. 54. Chen C, Zhao S, Karnad A, et al. The biology and role of CD44 in cancer progression: therapeutic implications. J Hematol Oncol 2018; 11: 64. 55. Han S, Zong S, Shi Q, et al. Is Ep-CAM expression a diagnostic and prognostic biomarker for colorectal cancer? A systematic meta-analysis. Ebiomedicine 2017; 20: 61–69. 56. Al-Hajj M, Wicha MS, Benito-Hernandez A, et al. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 2003; 100: 3983–3988. 57. Dalerba P, Dylla SJ, Park IK, et al. Phenotypic characterization of human colorectal cancer stem cells. Proc Natl Acad Sci U S A 2007; 104: 10158–10163. 58. Tayama S, Motohara T, Narantuya D, et al. The impact of EpCAM expression on response to chemotherapy and clinical outcomes in patients with epithelial ovarian cancer. Oncotarget 2017; 8: 44312–44325. 59. Dai XM, Huang T, Yang SL, et al. Peritumoral EpCAM is an independent prognostic marker after curative resection of HBV-related hepatocellular carcinoma. Dis Markers 2017; 2017: 8495326. 60. Endaya B, Guan SP, Newman JP, et al. Human mesenchymal stem cells preferentially migrate toward highly oncogenic human hepatocellular carcinoma cells with activated EpCAM signaling. Oncotarget 2017; 8: 54629–54639. 61. Rassouli FB, Matin MM and Saeinasab M. Cancer stem cells in human digestive tract malignancies. Tumour Biol 2016; 37: 7–21. 62. Nicolazzo C, Raimondi C, Francescangeli F, et al. EpCAM-expressing circulating tumor cells in colorectal cancer. Int J Biol Markers 2017; 32: e415–e420. 63. Tinhofer I, Hristozova T, Stromberger C, et al. Monitoring of circulating tumor cells and their expression of EGFR/phospho-EGFR during combined radiotherapy regimens in locally advanced squamous cell carcinoma of the head and neck. Int J Radiat Oncol Biol Phys 2012; 83: e685–e690. 64. Alix-Panabieres C and Pantel K. Challenges in circulating tumour cell research. Nat Rev Cancer 2014; 14: 623–631. 65. Lee M, Kim EJ, Cho Y, et al. Predictive value of circulating tumor cells (CTCs) captured by microfluidic device in patients with epithelial ovarian cancer. Gynecol Oncol 2017; 145: 361–365. 66. De Wit S, van Dalum G, Lenferink AT, et al. The detection of EpCAM(+) and EpCAM(–) circulating tumor cells. Sci Rep 2015; 5: 12270. 67. Kaigorodova EV, Tarabanovskaya NA, Staheeva MN, et al. Effect of small and radical surgical injury on the level of different populations of circulating tumor cells in the blood of breast cancer patients. Neoplasma 2017; 64: 437–443. 68. Baillie R, Tan ST and Itinteang T. Cancer stem cells in oral cavity squamous cell carcinoma: a review. Front Oncol 2017; 7: 112.
Fonseca et al. 69. Zavros Y. Initiation and maintenance of gastric cancer: a focus on CD44 variant isoforms and cancer stem cells. Cell Mol Gastroenterol Hepatol 2017; 4: 55–63. 70. Zoller M. CD44, hyaluronan, the hematopoietic stem cell, and leukemia-initiating cells. Front Immunol 2015; 6: 235. 71. Tsaur I, Thurn K, Juengel E, et al. sE-cadherin serves as a diagnostic and predictive parameter in prostate cancer patients. J Exp Clin Cancer Res 2015; 34: 43. 72. Griffiths TR, Brotherick I, Bishop RI, et al. Cell adhesion molecules in bladder cancer: soluble serum E-cadherin correlates with predictors of recurrence. Br J Cancer 1996; 74: 579–584. 73. Buhring HJ, Muller T, Herbst R, et al. The adhesion molecule E-cadherin and a surface antigen recognized by the antibody 9C4 are selectively expressed on erythroid cells of defined maturational stages. Leukemia 1996; 10: 106–116. 74. Melki JR, Vincent PC, Brown RD, et al. Hypermethylation of E-cadherin in leukemia. Blood 2000; 95: 3208–3213. 75. Zhang TJ, Zhou JD, Ma JC, et al. CDH1 (E-cadherin) expression independently affects clinical outcome in acute myeloid leukemia with normal cytogenetics. Clin Chem Lab Med 2017; 55: 123–131. 76. Ewerth D, Schmidts A, Hein M, et al. Suppression of APC/CCdh1 has subtype specific biological effects in acute myeloid leukemia. Oncotarget 2016; 7: 48220–48230. 77. Rao Q, Wang JY, Meng J, et al. Low-expression of Ecadherin in leukaemia cells causes loss of homophilic adhesion and promotes cell growth. Cell Biol Int 2011; 35: 945–951. 78. Jordaan G, Liao W and Sharma S. E-cadherin gene reexpression in chronic lymphocytic leukemia cells by HDAC inhibitors. BMC Cancer 2013; 13: 88. 79. Ohgami RS, Chisholm KM, Ma L, et al. E-cadherin is a specific marker for erythroid differentiation and has utility, in combination with CD117 and CD34, for enumerating myeloblasts in hematopoietic neoplasms. Am J Clin Pathol 2014; 141: 656–664. 80. Burk AS, Monzel C, Yoshikawa HY, et al. Quantifying adhesion mechanisms and dynamics of human hematopoietic stem and progenitor cells. Sci Rep 2015; 5: 9370. 81. Qiu S, Jia Y, Xing H, et al. N-cadherin and Tie2 positive CD34+CD38–CD123+ leukemic stem cell populations can develop acute myeloid leukemia more effectively in NOD/SCID mice. Leuk Res 2014; 38: 632–637. 82. Kawamura-Kodama K, Tsutsui J, Suzuki ST, et al. Ncadherin expressed on malignant T cell lymphoma cells is functional, and promotes heterotypic adhesion between the lymphoma cells and mesenchymal cells expressing Ncadherin. J Invest Dermatol 1999; 112: 62–66. 83. De Yzaguirre MM, Hernandez JS, Navarro PF, et al. Epigenetic silencing of E- and N-cadherins in the stroma of mouse thymic lymphomas. Carcinogenesis 2006; 27: 1081–1089. 84. Kutlesa S, Wessels JT, Speiser A, et al. E-cadherinmediated interactions of thymic epithelial cells with
11 CD103+ thymocytes lead to enhanced thymocyte cell proliferation. J Cell Sci 2002; 115: 4505–4515. 85. Zheng X, Fan X, Fu B, et al. EpCAM inhibition sensitizes chemoresistant leukemia to immune surveillance. Cancer Res 2017; 77: 482–493. 86. Joensuu H, Ristamaki R, Klemi PJ, et al. Lymphocyte homing receptor (CD44) expression is associated with poor prognosis in gastrointestinal lymphoma. Br J Cancer 1993; 68: 428–432. 87. Grabowska MM and Day ML. Soluble E-cadherin: more than a symptom of disease. Front Biosci 2012; 17: 1948–1964. 88. Giebultowicz J, Polanska-Plachta M, Wroczynski P, et al. How echinoccocosis affects potential cancer markers in plasma: galectin-3, sN-cadherin and sE-cadherin? A preliminary report. Diagn Pathol 2012; 7: 17. 89. Pohnan R, Ryska M, Hytych V, et al. Echinococcosis mimicking liver malignancy: a case report. Int J Surg Case Rep 2017; 36: 55–58. 90. Laucella S, De Titto EH, Segura EL, et al. Soluble cell adhesion molecules in human Chagas’ disease: association with disease severity and stage of infection. Am J Trop Med Hyg 1996; 55: 629–634. 91. Patouraux S, Rousseau D, Bonnafous S, et al. CD44 is a key player in non-alcoholic steatohepatitis. J Hepatol 2017; 67: 328–338. 92. Kaur S, Narayanswamy S and Ramesh AV. Comparative evaluation of salivary soluble CD44 levels in periodontal health and disease. J Indian Soc Periodontol 2014; 18: 734–738. 93. Mashayekhi F, Aryaee H, Mirzajani E, et al. Soluble CD44 concentration in the serum and peritoneal fluid samples of patients with different stages of endometriosis. Arch Gynecol Obstet 2015; 292: 641–645. 94. Kuessel L, Wenzl R, Proestling K, et al. Soluble VCAM1/soluble ICAM-1 ratio is a promising biomarker for diagnosing endometriosis. Hum Reprod 2017; 32: 770–779. 95. Kahlert UD, Joseph JV and Kruyt FAE. EMT- and MET-related processes in nonepithelial tumors: importance for disease progression, prognosis, and therapeutic opportunities. Mol Oncol 2017; 11: 860–877. 96. Essers MA and Trumpp A. Targeting leukemic stem cells by breaking their dormancy. Mol Oncol 2010; 4: 443–450. 97. Norkin M, Uberti JP and Schiffer CA. Very late recurrences of leukemia: why does leukemia awake after many years of dormancy? Leuk Res 2011; 35: 139–144. 98. Chen W, Dong J, Haiech J, et al. Cancer stem cell quiescence and plasticity as major challenges in cancer therapy. Stem Cells Int 2016; 2016: 1740936. 99. Chen S, Lewallen M and Xie T. Adhesion in the stem cell niche: biological roles and regulation. Development 2013; 140: 255–265. 100. Su W, Foster SC, Xing R, et al. CD44 transmembrane receptor and hyaluronan regulate adult hippocampal neural stem cell quiescence and differentiation. J Biol Chem 2017; 292: 4434–4445.
12 101. Cao H, Heazlewood SY, Williams B, et al. The role of CD44 in fetal and adult hematopoietic stem cell regulation. Haematologica 2016; 101: 26–37. 102. Lopez-Ruano G, Prieto-Bermejo R, Ramos TL, et al. PTPN13 and beta-catenin regulate the quiescence of hematopoietic stem cells and their interaction with the bone marrow niche. Stem Cell Reports 2015; 5: 516–531.
Tumor Biology 103. Khan NI, Cisterne A, Devidas M, et al. Expression of CD44, but not CD44v6, predicts relapse in children with B cell progenitor acute lymphoblastic leukemia lacking adverse or favorable genetics. Leuk Lymphoma 2008; 49: 710–718. 104. Takahashi H, Koh K, Kato M, et al. Acute myeloid leukemia with mediastinal myeloid sarcoma refractory to acute myeloid leukemia therapy but responsive to Lasparaginase. Int J Hematol 2012; 96: 136–140.