Hostname: page-component-76fb5796d-25wd4 Total loading time: 0 Render date: 2024-04-26T20:16:56.870Z Has data issue: false hasContentIssue false

Targeting N-cadherin (CDH2) and the malignant bone marrow microenvironment in acute leukaemia

Published online by Cambridge University Press:  03 May 2023

Jessica Parker
Affiliation:
Department of Applied Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK
Sean Hockney
Affiliation:
Department of Applied Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK
Orest W. Blaschuk
Affiliation:
Zonula Incorporated, Kirkland, QC H9J 2X2, Canada
Deepali Pal*
Affiliation:
Department of Applied Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK Wolfson Childhood Cancer Research Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Herschel Building Level 6, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
*
Corresponding author: Deepali Pal; Email: deepali.pal@northumbria.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

This review discusses current research on acute paediatric leukaemia, the leukaemic bone marrow (BM) microenvironment and recently discovered therapeutic opportunities to target leukaemia–niche interactions. The tumour microenvironment plays an integral role in conferring treatment resistance to leukaemia cells, this poses as a key clinical challenge that hinders management of this disease. Here we focus on the role of the cell adhesion molecule N-cadherin (CDH2) within the malignant BM microenvironment and associated signalling pathways that may bear promise as therapeutic targets. Additionally, we discuss microenvironment-driven treatment resistance and relapse, and elaborate the role of CDH2-mediated cancer cell protection from chemotherapy. Finally, we review emerging therapeutic approaches that directly target CDH2-mediated adhesive interactions between the BM cells and leukaemia cells.

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
Copyright © The Author(s), 2023. Published by Cambridge University Press

Background

Leukaemia accounts for 31% of cancer diagnoses in children up to 14 years of age in the UK, of which 401 children are diagnosed with acute lymphoblastic leukaemia (ALL) and 79 with acute myeloid leukaemia (AML) per year (Ref. 1); overall, the 5-year survival of ALL and AML is over 90 and 67%, respectively (Refs 1, Reference Bartram, Veys and Vora2). N-cadherin (CDH2) is a cell adhesion molecule that mediates adhesive interactions between leukaemia cells and the cells of the bone marrow (BM) (Refs Reference Borbaran-Bravo3, Reference Pal4). These interactions facilitate leukaemia cell survival, evasion from apoptosis and cell dormancy ultimately resulting in treatment resistance (Refs Reference Borbaran-Bravo3, Reference Pal4). Indeed, the niche-protected, dormant, non-apoptotic leukaemia cells may re-emerge in relapsed cases to develop resistance to therapy (Refs Reference Mrozik5, Reference Zhi6).

Dysregulation of normal blood homoeostasis is the main underlying developmental anomaly that leads to ALL and AML. Leukaemogenesis usually comprises a series of steps with an accumulation of genetic and epigenetic changes, inducing extensive alterations impacting cell growth, metabolism, cell cycle progression, cell death and differentiation, leading to preleukaemic haematopoietic stem cells (HSCs) and subsequently the development of ALL and AML (Refs Reference Yamashita7, Reference Belver and Ferrando8, Reference Iacobucci and Mullighan9). Because of the complexity of epigenetic and genetic mutations, it is not fully understood what cascade of events occur to give rise to the leukaemia phenotype and which perturbations are responsible for driving leukaemogenesis. Through advancement of technology, single-cell RNA sequencing has become more accurate, and been applied to examine leukaemia cells at the transcriptional level. A study by Watcham et al. (Ref. Reference Watcham, Kucinski and Gottgens10) presented data that suggest many leukaemia perturbations can gain advantage over wild-type cells, and drive cells into a more active state (Ref. Reference Watcham, Kucinski and Gottgens10). Indeed, many studies show that in utero mutations are becoming more recognised as commonplace in acute leukaemia and could be responsible for fusion genes in paediatric patients with ALL and AML; these mutations are known as an initiating event (Refs Reference Chen11, Reference Shin12, Reference Winer13). Fusion genes are chromosomal aberrations that have a role in leukaemogenesis (Ref. Reference Wang14), and can involve genes associated with protein kinase pathways, transcription factor and epigenetic modifications (Ref. Reference Chen11).

Across mammals the number of HSCs per individual is thought to be conserved, with approximately 300 HSCs at birth, compared with between 11 000 and 22 000 in adults. Development of childhood leukaemia depends on initial somatic mutations in HSCs, and because of the small HSC pool size these mutations are more likely to have a greater impact on the HSC population (Ref. Reference Rozhok, Salstrom and DeGregori15). The Knudson ‘two-hit’ hypothesis, established in 1971, suggested that dominantly inherited predisposition to cancer begins with a germline mutation, as can be seen with fusion genes; however a second, somatic mutation is needed for tumourigenesis (Ref. Reference Knudson16).

For example, only about 1% of children born with the ETV6-RUNX1 fusion gene develop the second-hit mutation that is needed to transform to ALL, indicating the fusion gene mutation is weakly penetrant (Ref. Reference Böiers17). Many somatic mutations such as TP53, RUNX1 and IKZF1 are found at the same sites of germline mutations in children who develop leukaemia (Refs Reference Demir18, Reference Maciel19, Reference Malla20). For example, a germline mutation at CEBPA leads to the development of AML with almost complete penetrance, this mutation is known to present favourable outcome (Ref. Reference Liao21).

Intensification of chemotherapeutic regimens is thought to be one of the main reasons for increased survival in childhood leukaemia; however, such treatment is associated with high morbidity and mortality rates. For example, in AML, high dose cytarabines used in young adults (15–24 years old) were reported to have a benefit to outcome; however, these results could not be translated to paediatric patients. The COG trial AAML1031 intensified induction chemotherapy with mitoxantrone and cytarabine and found that intensification did not achieve a survival benefit in paediatric patients, since remission rates were comparable with the AAML0531 trail which did not include intensifying induction chemotherapy. Moreover, additional haematological toxicity was found to be associated with treatment intensification, therefore showing an increased toxicity without any proportional benefit in treatment (Ref. Reference Elgarten22).

Studies have been conducted worldwide to analyse toxicity of paediatric acute leukaemia treatment (Refs Reference Hough23, Reference Zawitkowska24, Reference Franca25). Table 1 shows comparisons of different paediatric ALL protocols in the UK and two European countries and their associated toxicities. Results show that up to 49% of patients experienced an adverse event because of the chemotherapeutic agents used in their treatment, with toxicity-induced mortality rates up to 3.7%. The children studied by Zawitkowska et al. (Ref. Reference Zawitkowska24) were evaluated for the ‘grade’ of toxicity; it was found that children with grade 3 or higher were found to have a lower overall survival and event-free survival rate compared with children with a lower grade of treatment toxicity (Ref. Reference Zawitkowska24).

Table 1. Comparison of paediatric ALL protocols from the UK and two European countries, including study size and number of patients affected by adverse events

The table shows the top three most common toxicities and alongside mortality rates because of treatment (Refs Reference Hough23, Reference Zawitkowska24, Reference Franca25).

Chemoprotection induced by the leukaemia microenvironment is important in conferring treatment protection to cancer cells via mechanisms that include leukaemia cell–BM niche interactions and malignant dormancy (Refs Reference Passaro26, Reference Barbier27). ALL chemotherapies include DNA damaging and spindle poisons, which target the S and M phases of the cell cycle. These therapies rely on targeting actively cycling leukaemia cells, and therefore are ineffective against dormant cells which consequently lead to treatment resistance and relapse (Refs Reference Ebinger28, Reference Pal, Heidenreich and Vormoor29). To improve efficacy of treatment and limit treatment failure and relapse, approaches including targeted therapy, immunotherapy and gene therapy are being explored.

Targeted therapy includes risk stratification, an approach where patients are grouped based on disease risk or therapy response from diagnostic tests. In a clinical trial for paediatric ALL (JPLSG MLL-10 trial), patients were stratified into three risk groups according to their KMT2A gene rearrangement status (KMT2A-r), age and presence of central nervous system (CNS) leukaemia (Ref. Reference Tomizawa30). High-dose cytarabine was given to KMT2A-r patients with haematopoietic stem cell transplant (HSCT) option being reserved for high-risk patients. Consequently, this removed the requirement for HSCT in patients with KMT2A-r (Ref. Reference Tomizawa30). Although patient stratification has contributed to the improved survival rates for paediatric ALL, intensifying chemotherapy attains a plateau where there is no additional benefit to patients but only an increased toxicity exposure. To overcome the limitations of targeted therapy, novel approaches need to be incorporated into the treatment protocol.

Immunotherapies have been explored to overcome the challenges presented by conventional targeted therapies. For example, blinatumomab presented promising results in a phase I/II trial with paediatric patients with relapsed/refractory ALL (Ref. Reference Pal, Heidenreich and Vormoor29). In a phase III trial in paediatric patients with B-ALL at high risk of relapse, blinatumomab was superior to conventional consolidation therapy (Ref. Reference von Stackelberg31). However, blinatumomab presents unique and significant toxicities of neurological events and cytokine release syndrome (CRS), which includes pyrexia, headache, nausea, fatigue and hypotension, although these findings were presented from adults with relapsed B-ALL (Ref. Reference Topp32). CRS has been seen to be infrequent in low minimal residual disease (MRD) settings and most neurological events could be reversed through interrupting infusions (Ref. Reference Gökbuget33), suggesting that blinatumomab could be effective with minimal toxicity in patients with low MRD, although alternatives would be needed in other patients.

Gene therapy is another emerging route to overcome the challenges of conventional therapies. T-cell therapy involves genetically engineering chimeric antigen receptor (CAR) T cells, coupling an anti-CD19 domain to intracellular T-cell signalling domains of the T-cell receptor, which redirects cytotoxic T lymphocytes to cells expressing the CD19 antigen, in B cell leukaemia (Ref. Reference Kalos34). Anti-CD19 CAR T-cell therapy, tisagenlecleucel, has been FDA-approved after high remission rates were found in patients with ALL and while severe toxicities were observed these effects were reversible (Refs Reference Maude35, Reference Maude36).

The roles of cadherins in the leukaemia microenvironment

Classical cadherins are a calcium-dependent adhesion molecule family, grouped into type-I and type-II subgroups based on the molecular features of their interactions via the cadherin motifs (Ref. Reference Nollet, Kools and Van Roy37). Neural (N)-cadherin (CDH2) and epithelial (E)-cadherin (CDH1) are type-I cadherins which are characterised by the cell adhesion recognition motif His-Ala-Val (HAV) in their first extracellular domain (Refs Reference Blaschuk38, Reference Kashef39). CDH1 is a tumour suppressor protein which plays an important role in regulating tissue homoeostasis by modulating permeability barriers (i.e. tight junctions) between compartments, and the functional state of CDH1 determines metastatic potential (Ref. Reference Na40). Functional activity of CDH1 can be modified in response to environmental factors and CDH1 can be activated by monoclonal antibodies to inhibit metastasis at multiple stages of the metastatic cascade (Ref. Reference Na40). CDH2 is typically known for its role in morphogenetic processes in health such as during the formation of cardiac and neural tissue, and in diseases such as solid tumours. Moreover, recent research indicates overexpression of CDH2 in HSCs show increased HSC attachment to BM endosteal surfaces (Ref. Reference Mrozik5). In disease, loss of CDH1 and upregulation of CDH2 in cancer cells leads to metastatic dissemination and activation of several epithelial–mesenchymal transition (EMT) transcription factors (Ref. Reference Onder41). EMT is a cellular morphogenetic transition from a non-motile, epithelial phenotype into a migratory, mesenchymal-like phenotype and is thought to be a driving force in tumourigenesis and metastasis (Refs Reference Christofori42, Reference Gupta and Massagué43, Reference Thiery and Sleeman44, Reference Thiery45, Reference Craene and Berx46). CDH2 has been identified as an important molecule of interest in leukaemia. A recent study demonstrated that this adhesion molecule was upregulated in leukaemia cells primed by BM niche cells (Ref. Reference Pal4). Furthermore, MILE study and Bloodspot database showed that multiple haematological malignancies exhibited CDH2 upregulation compared with healthy BMs (Refs Reference Bagger, Kinalis and Rapin47, Reference Haferlach48).

On a related note, osteoblast (OB)–cadherin (CDH11) a type-II cadherin (Ref. Reference Maude35) important in the formation of the neural crest (NC) cells, has been further shown in disease models to cause tumour growth, cell survival and EMT (Refs Reference Piao49, Reference Row50, Reference Yoshioka51). It has been further suggested that intracellular downstream signalling of CDH11 is essential for maintenance and survival of premigratory NC cells. In addition, cells require CDH11 for physiological cell–cell, adhesion-related EMT in the preparatory steps prior to migration (Ref. Reference Manohar52). However, the biological role of CDH11 in leukaemia has not yet been explored.

The role of CDH2 in the BM niches and in chemoprotection

Biological systems are complex where their complexity is characterised by multicellularity, degeneracy and redundancy of the component cell types. The BM is a viscous tissue within the bone comprised of two well-defined niches – endosteal and perivascular, where HSCs are found in close proximity to OB and endothelial cells (ECs) (Ref. Reference Bello, Park and Lee53). All blood lineages and immune cells are derived from the common precursor, HSCs (Ref. Reference Jiang54), which retains the ability for both multipotency and self-renewal (Ref. Reference Zhang55). The two niches are intertwined to create a functional microenvironment, that facilitates cell communication during HSC development consequently helping to maintain the full blood cell forming potential of HSCs (Ref. Reference Zhang55).

It has been well-established that the endosteal niche is filled with mesenchymal stromal cells (MSCs), osteoprogenitor cells, pre-OBs, mature OBs, osteocytes and osteoclasts (Ref. Reference Le, Andreeff and Battula56). OBs play an important role in maintaining a functional microenvironment and are involved in stem cell quiescence and proliferation (Ref. Reference Kajiume57). For example, SDF-1α in OBs is associated with HSC mobility (Ref. Reference Mangialardi, Cordaro and Madeddu58). In disorders that affect HSCs, such as myelodysplastic syndrome, it has been shown that suppressing osteogenic differentiation from MSCs leads to their impairment in supporting HSCs (Ref. Reference Hayashi59). Non-collagenous bone matrix proteins, such as osteopontin and osteocalcin, regulate cell migration and bone mineralisation and are believed to be linked to cell proliferation, osteogenic differentiation and angiogenesis; however, these processes are yet to be defined in leukaemia cell biology (Ref. Reference Carvalho60).

Because of inaccessibility of reliable animal models, the niche microenvironment of ALL has not been well-established. However, the remodelling of the BM vasculature following AML leukaemogenesis has been studied and it was found that AML cells aid the niche transformation into a preferential leukaemia microenvironment. These changes are anatomically diverse; although vasculature in the endosteum was lost through disease progression, central vessels survived with compromised function. This process was thought to be because of the production of pro-inflammatory and anti-angiogenic cytokines from AML cells in the endosteal lining which degrade the surrounding endothelium, as well as stromal osteoblastic cells, together leading to the reduced capacity to support HSCs. Vasculature was maintained in T-ALL murine models suggesting this vascular remodelling is specific to AML (Refs Reference Passaro26, Reference Duarte61). The inflammatory cytokine, TNF-α, secreted by AML cells, directly induces E-selectin which plays a role in promoting malignant cell survival, proliferation and chemoresistance (Ref. Reference Barbier27). AML engraftment also induces exogenous nitric oxide overproduction, which affects HSC motility and increases HSC activation leading to reduction in their repopulating activity (Ref. Reference Passaro26). Increased vascular leakiness was observed in AML xenografts after induction therapy, leading to poor drug delivery and the formation of areas with low perfusion rates, where leukaemia cell migration resulted in microenvironment-induced treatment resistance (Ref. Reference Passaro26).

Peri-arteriolar stromal cells which are innervated by the sympathetic nervous system and express neural markers NG2 and nestin (NG2+/nestin+ MSCs), have previously been found to control HSC quiescence and haematopoiesis (Ref. Reference Kunisaki62). The BM is known to be the site of dormant-disseminated tumour cells (DTCs) and Nobre et al. (Ref. Reference Nobre63) found that NG2+/nestin+ MSCs drive DTC dormancy which indicate that the perivascular niche is important for both HSC and DTC dormancy. NG2+/nestin+ MSCs produce TGF-β2 and BMP7, which signal a quiescent pathway through TGFBRIII and BMPRII, thereby activating SMAD, p38 and p27 pathways leading to dormancy (Ref. Reference Nobre63). Treatment induced damage of endosteal and perivascular niches have also been reported (Ref. Reference De Rooij, Zwaan and van den Heuvel-Eibrink64). Further research determined that leukaemia cells have an important function in the development of a new therapy-induced niche formation. Following treatment, secretion of cytokines and growth factors were found to increase in the microenvironment likely because of secretion by the leukaemia cells (Ref. Reference De Rooij, Zwaan and van den Heuvel-Eibrink64). Indeed the leukaemia niche has been reported to be transient, beginning initially as nestin+ cells maturing into α-SMA+ cells before terminating with fibre residues (Ref. Reference De Rooij, Zwaan and van den Heuvel-Eibrink64).

In keeping with these studies, recent research shows upregulation of CDH2, a known marker of EMT, in niche-primed leukaemia cells. This study demonstrated that knockdown of CDH2 in leukaemia cells reduce their proliferation while increasing sensitivity to dexamethasone treatment (Refs Reference Borbaran-Bravo3, Reference Pal4). Under physiological conditions, CDH2 plays a role in osteogenesis in the endosteal niche, specifically in maintaining the precursor OB pool (Ref. Reference Alimperti and Andreadis65). CDH2-mediated interactions with OBs are thought to play a role in supporting HSC function, with HSC–OB cell interactions enabling adhesion of HSCs to cells present in the endosteal niche (Refs Reference Mrozik5, Reference Zhao66). CDH2 is also expressed by various cell types associated with the HSC niche (Fig. 1), including stromal cells in the endosteal niche, and ECs and their associated pericytes in the microvascular of the perivascular niche (Ref. Reference Mrozik5).

Figure 1. Schematic diagram of the BM microenvironment under normal conditions and following leukaemogenesis and treatment in AML (top right) and ALL (top left). After leukaemogenesis and treatment, the microenvironment is remodelled, pro-inflammatory and anti-angiogenic cytokines are produced resulting in the loss of vasculature in the endosteal and osteoblastic cells. Adapted from Refs Reference Passaro26, Reference Barbier27, Reference Bello, Park and Lee53, Reference Jiang54, Reference Zhang55, Reference Le, Andreeff and Battula56, Reference Kajiume57, Reference Mangialardi, Cordaro and Madeddu58, Reference Hayashi59, Reference Carvalho60, Reference Duarte61, Reference Kunisaki62, Reference Nobre63.

CDH2 upregulation has been reported in human leukaemic BMs (Ref. Reference Pal4). A recent study has shown that CDH2 upregulation by niche-primed leukaemia is associated with increased cancer proliferation and acquisition of treatment resistance and importantly this interaction is druggable using the CDH2 antagonist ADH-1 (Refs Reference Borbaran-Bravo3, Reference Pal4). In adult AML CDH2 supports tumour growth and aids in maintaining self-renewal characteristics of leukaemia stem cells (LSCs), as CDH2+ cells have been found to engraft on NOD/SCID mice at a higher proportion than CDH2 cells (Ref. Reference Zhi6). CDH2 is also thought to support microenvironment-induced treatment protection in AML (Ref. Reference Tabe and Konopleva67). Indeed, adhesion interactions between LSCs and the BM microenvironment activate signalling cascades, which regulate functions including cell survival, evasion of apoptosis and cell dormancy. LSC interactions with the BM microenvironment enable them to evade the cytotoxic effects of chemotherapeutic agents, suggesting there is a reliance on adhesive interactions between AML LSCs and the BM for chemoprotection (Refs Reference Mrozik5, Reference Barwe, Quagliano and Gopalakrishnapillai68). CDH2 overexpression in HSCs decreases in vitro cell division rate, this is likely because of the sequestration of the CDH2 binding, intracellular β-catenin to the plasma membrane, thus suppressing its activity as a transcription factor in the nucleus (Refs Reference Zhi6, Reference Blaschuk38). In support of this, adult AML BM contains CDH2+ LSCs which are found in a quiescent state in G0/G1 cell cycle arrest, which renders them less sensitive to chemotherapy (Refs Reference Zhi6, Reference Arai69). Lastly, in adult AML, CDH2 is also thought to play a role in drug resistance, CDH2+ LSCs were found to have a higher IC50 of VP-16, an anti-leukaemia therapeutic drug, than the CDH2 population (Refs Reference Zhi6, Reference Zhi70).

Pathways associated with CDH2

There are many pathways that are associated with CDH2 in various malignancies. Two pathways of relevance to this review are the Wnt/β-catenin pathway and the phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway, as detailed in Figure 2. It is of note, that there is limited research into these signalling pathways in acute leukaemia, clearly indicating an area warranting further exploration.

Figure 2. Schematic diagram of the pathways and transcription factors associated with CDH2, including the PI3K/Akt/mTOR pathway and the Wnt/β-catenin pathway. Arrows represent activation; bars represent inhibition, double-ended arrows in the pathway indicate upregulation of a molecule results in downregulation of the other and vice versa. Adapted from Refs Reference Chiarini71, Reference Dandekar72, Reference Gang73, Reference Grüninger74, Reference Zou85.

Hyperactivation of the PI3K/Akt/mTOR signalling pathway has been reported in 88% of ALL patients and is associated with poor prognosis and chemotherapeutic resistance (Ref. Reference Grüninger74). The PI3K–Akt–mTOR pathway is important for haematopoietic cells, regulating functions such as HSC proliferation, differentiation and survival, and is furthermore constitutively activated in AML cells (Refs Reference Nepstad78, Reference Nepstad79). The presence of PI3K/Akt/mTOR pathway has been well-established in solid tumours and dimerisation and phosphorylation of PI3K leads to the downstream activation of Akt. Akt stimulates cell survival by upregulating mouse double minute 2 homologue (MDM2), which inhibits p53, and upregulates BCL2, both leading to inhibition of apoptosis. Akt activation subsequently triggers the phosphorylation of mTOR (Ref. Reference Pothongsrisit and Pongrakhananon81). mTOR is a conserved serine/threonine kinase that belongs to the PI3K-related kinase family and has also been well-established in solid tumours. It is a constituent of two signalling complexes, mTORC1 involved in mRNA translation and protein synthesis and mTORC2 which controls cell survival and migration (Refs Reference Rabanal-Ruiz, Otten and Korolchuk82, Reference Zhang and Manning84, Reference Zou85). There is evidence to link p70S6K to the Akt/mTOR pathway in AML (Ref. Reference Murray77). In solid tumours p70S6K activates the transcription factors, slug and snail, which downregulates CDH1 and upregulates CDH2, leading to EMT (Ref. Reference Liang75).

Dysregulation in the Wnt/β-catenin pathway can lead to initiation and progression of cancer, including haematological malignancies, and β-catenin activation has been found to contribute to ALL and AML drug resistance (Refs Reference Chiarini71, Reference Dandekar72, Reference Gang73, Reference Wang83). The inactivation of GSK3β from Akt-dependent phosphorylation prevents β-catenin phosphorylation, leading to the activation of β-catenin-independent genes and uncontrolled cell proliferation (Ref. Reference Chiarini71). CDH2 regulates Wnt/β-catenin signalling, a conserved pathway that plays a role in physiological processes, including differentiation, proliferation and cell fate determination. CDH1 is known to inhibit the activation of the Wnt pathway, and in ALL CDH1 has been shown to be decreased, indicating Wnt pathway activation (Ref. Reference Ma76). Additionally in ALL, Akt has been shown to inhibit GSK3β leading to the activation of β-catenin (Ref. Reference Perry80).

Therapeutic approaches targeting CDH2

Exherin (ADH-1)

The CDH2 antagonist ADH-1 is a cyclic pentapeptide which competitively inhibits CDH2 (Fig. 3), as it contains the cadherin cell adhesion recognition sequence HAV (Refs Reference Blaschuk38, Reference Yarom86). The proposed mechanism of action of ADH-1 in cancer is that it results in apoptosis in vitro, and causes inhibition of tumour cell migration in addition to altering the tumour vasculature in vivo (Refs Reference Shintani87, Reference Li, Price and Figg88, Reference Lammens89). Pal et al. (Ref. Reference Pal4) found that ADH-1 showed high efficacy in vitro and in vivo against patient-derived ALL cells, where ADH-1 reduced proliferation of ALL cells in vitro, as indicated by a reduced number of blasts in the S phase of the cell cycle (Ref. Reference Pal4). This research further assessed ADH-1 activity on CDH2 knockdown ALL cells, where ADH-1 treatment sensitivity was confirmed only in the wild-type ALL cells that did not harbour the CDH2 knockdown, thereby corroborating specificity of ADH-1 against CDH2 and suggesting against the likelihood of off-target effects (Ref. Reference Pal4). This study further validated ADH-1 to show efficacy both as a single agent and in combination with dexamethasone, in a patient-derived xenograft (PDX) mouse model, where addition of ADH-1 to dexamethasone did not result in any additional toxicity (Ref. Reference Pal4). Of note, ADH-1 is an FDA-approved compound with ‘orphan drug’ status for use in melanomas (Ref. Reference Perotti90), and ADH-1 treatment in patients with solid tumours was well tolerated resulting only in a few adverse events, most of which were grade 1 or 2, thereby showing a better tolerance than most current treatments (Ref. Reference Guo91). These findings indicate ADH-1 to be a potentially promising therapeutic agent that could be repurposed from solid cancers to leukaemia treatment.

Figure 3. ADH-1 competitively binds to CDH2 on BM cells, preventing leukaemia–niche cell binding of leukaemia cells within the BM microenvironment. Adapted from Refs Reference Blaschuk38, Reference Yarom86.

In addition, ADH-1-modified liposomes (A-LP) have been successfully constructed with the aim of enhancing chemotherapy efficacy and preventing metastasis and was tested using a PTX-resistant breast cancer cell line, MCF7 PTX-R, which was established into a tumour model using subcutaneous inoculation into the right flanks of female BALB/c nude mice. Results found that cellular uptake was increased because of the CDH2 expressed after EMT in the MCF7 PTX-R cells (Ref. Reference Guo91). Treatment with the A-LP showed cancer cells to have an increased chemo-sensitivity, with EMT to be somewhat suppressed.

ADH-1 has been further shown to improve immunotherapy by tumour-infiltrating lymphocyte (TIL)-related treatment. The immune dysfunction mechanism including programmed death ligand-1 (PDL-1) and indole amine 2,3-dioxygenase (IDO-1) induces apoptosis, both PDL-1 and IDO-1 are increased after EMT and immunosuppression is enforced. Therefore targeting CDH2 improved the efficacy of TIL-related treatment by decreasing PDL-1 and IDO-1, and indeed ADH-1 with TIL-treatment reduced tumour size and increased survival in the mouse models (Ref. Reference Sun92). Although ADH-1 has been documented in cancer pre-clinical studies and solid tumour clinical trials, in-depth mechanism of action of this drug remains unexplored. Further research needs to be conducted to develop an in-depth understanding of ADH-1, including scrutiny of any possible mechanisms of resistance that could arise following ADH-1 treatment. In addition, several next-generation antagonists, including small-molecule inhibitors of CDH2 are being developed (Ref. Reference Blaschuk38) and their role as potential anti-leukaemia treatment needs to be investigated.

CDH2 small-molecule antagonists

Much less is known concerning the biological effects of other types of CDH2 antagonists, as they have not been extensively developed for use as cancer therapeutics (Ref. Reference Blaschuk38). A large number of non-peptidyl peptidomimetics of ADH-1 have been recently identified (Refs Reference Mrozik93, Reference Gour94), for example, the small-molecule LCRF-0006 is an ADH-1 peptidomimetic that inhibits CDH2 function, induces apoptosis in multiple myeloma (MM) and synergises with bortezomib to enhance MM cell death in vitro (Ref. Reference Mrozik93).

Non-peptide peptidomimetics of the CDH2 Trp-containing amino-terminus have also been discovered and are being developed as cancer therapeutics (Refs Reference Blaschuk38, Reference Vaisburg and Blaschuk95). In particular, the peptidomimetic-designated Compound 15, a piperidin-4-amine which acts as a CDH2 antagonist, has been shown to induce apoptosis of MM, glioblastoma and pancreatic cancer cells, as well as fibroblast and cancer-associated death in vitro (Refs Reference Blaschuk38, Reference Smits, Blaschuk and Willerth96). However, the ability of this small molecule to affect leukaemia blast viability as well as its mechanism of action remains unexplored.

Targeting other pathways in combination with CDH2

Although targeted therapy underpinning oncogene addiction has shown great promise in cancer treatment, it is associated with emergence of treatment-resistant clones. Combinatorial therapies target multiple cancer pathways, and thereby aim to mitigate occurrence of treatment resistance. Furthermore, up to 40% of ALL patients present with CNS involvement, because of the ability of leukaemia cells to penetrate the blood–brain–barrier (BBB) (Ref. Reference Mitchell97). Although it is now well-established that achieving CNS clearance in ALL is essential for long-term disease cure, CNS-directed therapy is associated with significant toxicity (Ref. Reference Halsey and Escherich98). This highlights need for new and improved combinatorial treatments in ALL to prevent treatment resistance and mitigate treatment toxicity. Indeed combination therapies containing dexamethasone, a glucocorticoid routinely used to treat ALL, with venetoclax or ADH-1 have been shown to increase leukaemia-free long-term survival in pre-clinical mouse models and patients (Refs Reference Pal4, Reference Peirs99, Reference Scherr100). Furthermore, ADH-1 and dexamethasone have been found to show high efficacy when tested in combination on PDX mouse models transplanted with high risk ALL. The ADH-1/dexamethasone combination was found to significantly reduce the proportion of leukaemia blasts in vivo compared with the dexamethasone-only arm, and moreover addition of ADH-1 to dexamethasone did not result in any additional toxicity (Ref. Reference Pal4).

Adults with BCR-ABL+ ALL have poor prognosis; therefore, dexamethasone was tested in a triple combination with venetoclax and tyrosine kinase inhibitors (TKIs), imatinib or dasatinib. Both combinations were shown to be superior to single agents and double combinations in terms of tumour size and survival, although the combination with dasatinib was shown to be more effective (Ref. Reference Scherr100). Researching the value of adding ADH-1 to a dexamethasone/venetoclax/TKI is warranted especially in high risk and/or refractory disease to assess if this combination would improve efficacy and minimise emergence of treatment resistant clones (Ref. Reference Scherr100). Other drugs and pathways where adding ADH-1 as a combinatorial treatment might be valuable is as discussed below.

Dysregulation in the PI3K/Akt/mTOR pathway has been well-established as a component of AML pathogenesis. Many pharmacological inhibitors within this pathway have been evaluated in preclinical settings; however, there is yet to be meaningful clinical effectiveness of inhibition of this pathway for AML. Buparlisib is an oral pan-class I PI3K inhibitor, and has completed a phase I trial of patients with acute leukaemia and at doses of 80 mg/day was found to be tolerable with a modest single-agent efficacy. Buparlisib has also been seen to cross the BBB which is of importance in ALL with CNS infiltration (Refs Reference Ragon101, Reference de Gooijer102).

Idelalisib is a PI3K-δ inhibitor, more specifically p110δ a primary PI3K isoform in B cells and has shown activity in lymphoid malignancies and been FDA-approved for relapsed chronic lymphocytic leukaemia (CLL), follicular lymphoma and small lymphocytic lymphoma (Ref. Reference Brown103). Haematological malignancies such as relapsed CLL, follicular lymphoma and small lymphocytic lymphoma have been observed to depend on pre-B cell receptor signalling, which can also be seen in the majority of TCF3-PBX1 BCP-ALLs. The specificity of idelalisib to p110δ, results in a low toxicity profile, making it a promising therapeutic for TCF3-PBX1 BCP-ALL patients (Refs Reference Miller104, Reference Eldfors105). Interestingly, significant CDH2 upregulation in TCF3-PBX1 leukaemic BM combined with high ADH-1 efficacy seen in TCF3-HLF PDX samples would suggest that combining a CDH2 antagonist with idelalisib might be potentially beneficial.

mTOR inhibitors have shown promise in preclinical models of ALL through direct inhibition of tumour cell growth and reversal of glucocorticoid resistance and have demonstrated in vitro synergy with dexamethasone (Ref. Reference Silic-Benussi106). Everolimus presents these preclinical characteristics as a single agent, making it a good candidate for combination treatment. Moreover, there is a phase II study of everolimus in combination with vincristine, prednisone, pegaspargase and doxorubicin in relapsed ALL (Ref. Reference Place107). Everolimus was also tested in chronic myeloid leukaemia patients and found that in combination with imatinib, treatment was effective in both sensitive and resistant cases (Ref. Reference Alves108).

BEZ-235 is a dual pan-class I PI3K and mTOR inhibitor that has been tested in adult patients with relapsed/refractory acute leukaemia. Clinical development of BEZ-235 has been terminated because of suboptimal pharmacokinetic properties. Although this study found that efficacy observed in ALL patients warrant further clinical exploration into dual PI3K/mTOR inhibitors, in particular patients with Ph + BCP-ALL or T-ALL may benefit from these treatments (Ref. Reference Lang109). Given the link between CDH2, mTOR and EMT all of which play an important role in cancer biology (the role of EMT in non-epithelial cancers such as leukaemia is an emerging concept (Ref. Reference Chen110)), including niche-driven leukaemia cell behaviour, combining a CDH2 antagonist with mTOR inhibitors may have a potential therapeutic benefit.

Despite the role Wnt plays within acute leukaemia and its connection with CDH2, there has not been any clinical or preclinical testing with Wnt inhibitors, suggesting a potential area of further research, some pre-clinical antagonists are highlighted in Table 2 (Refs Reference Amado111, Reference Chen112, Reference Liu113, Reference Pan114, Reference Sohn115). Table 2 highlights inhibitors that have been tested against other cell lines and malignancies.

Table 2. List of the therapeutics, their targets and their progressions through clinical trials

Conclusion

In conclusion, CDH2 is an important molecule in both the healthy and malignant BM microenvironment, supporting both non-malignant haematopoietic cells and leukaemia cells. CDH2 supports tumour growth and promotes microenvironment-mediated treatment protection, decrease cell division rate and potentially plays a role in cancer dormancy. ADH-1, a first generation CDH2 inhibitor used in solid tumour clinical trials, demonstrated a well-tolerated toxicity profile and therefore may be an ideal candidate for combinatorial treatment in acute leukaemia. It is important to note that only CDH2 antagonists target the extracellular domain of cell surface receptors making them a unique class of therapeutic drugs. Furthermore, targeting other pathways that are associated with CDH2 may overcome environment-mediated drug resistance and may help reduce the rate of relapse in paediatric acute leukaemia. Next-generation CDH2 antagonists such as small-molecule inhibitors with improved potency and formulation are emerging as a unique class of anti-cancer therapeutics. These are potentially capable of targeting microenvironment-mediated malignant dormancy and treatment resistance in leukaemia and following in-depth preclinical and clinical validation may provide improved and low toxicity treatment options in paediatric leukaemia.

Conflict of interest

OWB holds shares in Zonula Incorporated. The company is developing N-cadherin antagonists (such as Compound 15) for the treatment of fibroblast-associated diseases. DP and her team which includes JP and SH, are collaborating in studies investigating the ability of Compound 15 to act as a therapeutic for the treatment of ALL.

References

England PH (2021) Children, teenagers and young adults UK cancer statistics report 2021. Internet.Google Scholar
Bartram, J, Veys, P and Vora, A (2020) Improvements in outcome of childhood acute lymphoblastic leukaemia (ALL) in the UK – a success story of modern medicine through successive UKALL trials and international collaboration. British Journal of Haematology 191, 562567.Google Scholar
Borbaran-Bravo, N et al. (2022) Regenerative medicine meets translational oncology: modeling leukemic bone marrow niche. Cell Reports Medicine 3, 100724.CrossRefGoogle ScholarPubMed
Pal, D et al. (2022) hiPSC-derived bone marrow milieu identifies a clinically actionable driver of niche-mediated treatment resistance in leukemia. Cell Reports Medicine 3, 100717.CrossRefGoogle ScholarPubMed
Mrozik, KM et al. (2018) N-cadherin in cancer metastasis, its emerging role in haematological malignancies and potential as a therapeutic target in cancer. BMC Cancer 18, 116.CrossRefGoogle ScholarPubMed
Zhi, L et al. (2016) N-cadherin aided in maintaining the characteristics of leukemic stem cells. The Anatomical Record 299, 990998.CrossRefGoogle ScholarPubMed
Yamashita, M et al. (2020) Dysregulated haematopoietic stem cell behaviour in myeloid leukaemogenesis. Nature Reviews Cancer 20, 365382.CrossRefGoogle ScholarPubMed
Belver, L and Ferrando, A (2016) The genetics and mechanisms of T cell acute lymphoblastic leukaemia. Nature Reviews Cancer 16, 494507.CrossRefGoogle ScholarPubMed
Iacobucci, I and Mullighan, CG (2017) Genetic basis of acute lymphoblastic leukemia. Journal of Clinical Oncology 35, 975983.CrossRefGoogle ScholarPubMed
Watcham, S, Kucinski, I and Gottgens, B (2019) New insights into hematopoietic differentiation landscapes from single-cell RNA sequencing. Blood, The Journal of the American Society of Hematology 133, 14151426.Google ScholarPubMed
Chen, X et al. (2021) Fusion gene map of acute leukemia revealed by transcriptome sequencing of a consecutive cohort of 1000 cases in a single center. Blood Cancer Journal 11, 110.CrossRefGoogle ScholarPubMed
Shin, S-Y et al. (2021) Recurrent somatic mutations and low germline predisposition mutations in Korean ALL patients. Scientific Reports 11, 8893.CrossRefGoogle ScholarPubMed
Winer, P et al. (2020) Germline variants in predisposition genes in children with down syndrome and acute lymphoblastic leukemia. Blood Advances 4, 672675.CrossRefGoogle ScholarPubMed
Wang, Y et al. (2017) Recurrent fusion genes in leukemia: an attractive target for diagnosis and treatment. Current Genomics 18, 378384.CrossRefGoogle ScholarPubMed
Rozhok, AI, Salstrom, JL and DeGregori, J (2016) Stochastic modeling reveals an evolutionary mechanism underlying elevated rates of childhood leukemia. Proceedings of the National Academy of Sciences of the USA 113, 10501055.CrossRefGoogle ScholarPubMed
Knudson, AG Jr (1971) Mutation and cancer: statistical study of retinoblastoma. Proceedings of the National Academy of Sciences of the USA 68, 820823.CrossRefGoogle ScholarPubMed
Böiers, C et al. (2018) A human IPS model implicates embryonic B-myeloid fate restriction as developmental susceptibility to B acute lymphoblastic leukemia-associated ETV6-RUNX1. Developmental Cell 44, 362377. e7.CrossRefGoogle Scholar
Demir, S et al. (2020) Therapeutic targeting of mutant p53 in pediatric acute lymphoblastic leukemia. Haematologica 105, 170.CrossRefGoogle ScholarPubMed
Maciel, ALT et al. (2022) IKZF1 deletions associate with CRLF2 overexpression leading to a poor prognosis in B-cell precursor acute lymphoblastic leukaemia. Translational Oncology 15, 101291.CrossRefGoogle ScholarPubMed
Malla, TM et al. (2023) Fishing for ETV6/RUNX1 fusion and MLL gene rearrangements and their additional abnormalities in childhood acute lymphoblastic leukemia patients of Kashmir. Gene 856, 147128.CrossRefGoogle Scholar
Liao, XY et al. (2022) CEBPA are independent good prognostic factors in pediatric acute myeloid leukemia. Hematological Oncology 40, 258268.CrossRefGoogle ScholarPubMed
Elgarten, CW et al. (2021) Outcomes of intensification of induction chemotherapy for children with high-risk acute myeloid leukemia: a report from the children's oncology group. Pediatric Blood & Cancer 68, e29281.CrossRefGoogle ScholarPubMed
Hough, R et al. (2016) Efficacy and toxicity of a paediatric protocol in teenagers and young adults with Philadelphia chromosome negative acute lymphoblastic leukaemia: results from UKALL 2003. British Journal of Haematology 172, 439451.CrossRefGoogle Scholar
Zawitkowska, J et al. (2019) Grade 3 and 4 toxicity profiles during therapy of childhood acute lymphoblastic leukemia. In vivo 33, 13331339.CrossRefGoogle ScholarPubMed
Franca, R et al. (2017) Pharmacogenetics and induction/consolidation therapy toxicities in acute lymphoblastic leukemia patients treated with AIEOP-BFM ALL 2000 protocol. The Pharmacogenomics Journal 17, 410.CrossRefGoogle ScholarPubMed
Passaro, D et al. (2017) Increased vascular permeability in the bone marrow microenvironment contributes to disease progression and drug response in acute myeloid leukemia. Cancer Cell 32, 324341. e6.CrossRefGoogle ScholarPubMed
Barbier, V et al. (2020) Endothelial E-selectin inhibition improves acute myeloid leukaemia therapy by disrupting vascular niche-mediated chemoresistance. Nature Communications 11, 2042.CrossRefGoogle ScholarPubMed
Ebinger, S et al. (2016) Characterization of rare, dormant, and therapy-resistant cells in acute lymphoblastic leukemia. Cancer Cell 30, 849862.CrossRefGoogle ScholarPubMed
Pal, D, Heidenreich, O and Vormoor, J (2016) Dormancy stems the tide of chemotherapy. Cancer Cell 30, 825826.CrossRefGoogle ScholarPubMed
Tomizawa, D et al. (2020) A risk-stratified therapy for infants with acute lymphoblastic leukemia: a report from the JPLSG MLL-10 trial. Blood 136, 18131823.CrossRefGoogle ScholarPubMed
von Stackelberg, A et al. (2016) Phase I/phase II study of blinatumomab in pediatric patients with relapsed/refractory acute lymphoblastic leukemia. Journal of Clinical Oncology 34, 43814389.CrossRefGoogle ScholarPubMed
Topp, MS et al. (2015) Safety and activity of blinatumomab for adult patients with relapsed or refractory B-precursor acute lymphoblastic leukaemia: a multicentre, single-arm, phase 2 study. The Lancet. Oncology 16, 5766.CrossRefGoogle ScholarPubMed
Gökbuget, N et al. (2018) Blinatumomab for minimal residual disease in adults with B-cell precursor acute lymphoblastic leukemia. Blood 131, 15221531.CrossRefGoogle ScholarPubMed
Kalos, M et al. (2011) T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Science Translational Medicine 3, 95ra7395ra73.CrossRefGoogle ScholarPubMed
Maude, SL et al. (2018) Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. New England Journal of Medicine 378, 439448.CrossRefGoogle Scholar
Maude, SL et al. (2014) Chimeric antigen receptor T cells for sustained remissions in leukemia. New England Journal of Medicine 371, 15071517.CrossRefGoogle ScholarPubMed
Nollet, F, Kools, P and Van Roy, F (2000) Phylogenetic analysis of the cadherin superfamily allows identification of six major subfamilies besides several solitary members. Journal of Molecular Biology 299, 551572.CrossRefGoogle ScholarPubMed
Blaschuk, OW (2022) Potential therapeutic applications of N-cadherin antagonists and agonists. Frontiers in Cell and Developmental Biology 10, 866200.CrossRefGoogle ScholarPubMed
Kashef, J et al. (2009) Cadherin-11 regulates protrusive activity in xenopus cranial neural crest cells upstream of trio and the small GTPases. Genes & Development 23, 13931398.CrossRefGoogle ScholarPubMed
Na, T-Y et al. (2020) The functional activity of E-cadherin controls tumor cell metastasis at multiple steps. Proceedings of the National Academy of Sciences of the USA 117, 59315937.CrossRefGoogle ScholarPubMed
Onder, TT et al. (2008) Loss of E-cadherin promotes metastasis via multiple downstream transcriptional pathways. Cancer Research 68, 36453654.CrossRefGoogle ScholarPubMed
Christofori, G (2003) Changing neighbours, changing behaviour: cell adhesion molecule-mediated signalling during tumour progression. The EMBO Journal 22, 23182323.CrossRefGoogle ScholarPubMed
Gupta, GP and Massagué, J (2006) Cancer metastasis: building a framework. Cell 127, 679695.CrossRefGoogle ScholarPubMed
Thiery, JP and Sleeman, JP (2006) Complex networks orchestrate epithelial–mesenchymal transitions. Nature Reviews Molecular Cell Biology 7, 131142.CrossRefGoogle ScholarPubMed
Thiery, JP et al. (2009) Epithelial-mesenchymal transitions in development and disease. Cell 139, 871890.CrossRefGoogle ScholarPubMed
Craene, BD and Berx, G (2013) Regulatory networks defining EMT during cancer initiation and progression. Nature Reviews Cancer 13, 97110.CrossRefGoogle ScholarPubMed
Bagger, FO, Kinalis, S and Rapin, N (2019) BloodSpot: a database of healthy and malignant haematopoiesis updated with purified and single cell mRNA sequencing profiles. Nucleic Acids Research 47, D881D8D5.CrossRefGoogle ScholarPubMed
Haferlach, T et al. (2010) Clinical utility of microarray-based gene expression profiling in the diagnosis and subclassification of leukemia: report from the international microarray innovations in leukemia study group. Journal of Clinical Oncology 28, 25292537.CrossRefGoogle ScholarPubMed
Piao, J et al. (2017) Substrate stiffness affects epithelial–mesenchymal transition of cervical cancer cells through miR-106b and its target protein DAB2. International Journal of Oncology 50, 20332042.CrossRefGoogle ScholarPubMed
Row, S et al. (2016) Cadherin-11 is a novel regulator of extracellular matrix synthesis and tissue mechanics. Journal of Cell Science 129, 29502961.Google ScholarPubMed
Yoshioka, R et al. (2015) Quantitative analysis of cadherin-11 and β-catenin signalling during proliferation of rheumatoid arthritis-derived synovial fibroblast cells. Journal of Pharmacy and Pharmacology 67, 10751082.CrossRefGoogle ScholarPubMed
Manohar, S et al. (2020) Cadherin-11 is required for neural crest specification and survival. Frontiers in Physiology 11, 563372.CrossRefGoogle ScholarPubMed
Bello, AB, Park, H and Lee, S-H (2018) Current approaches in biomaterial-based hematopoietic stem cell niches. Acta Biomaterialia 72, 115.CrossRefGoogle ScholarPubMed
Jiang, N et al. (2016) Hematopoietic stem cells in neural-crest derived bone marrow. Scientific Reports 6, 111.CrossRefGoogle ScholarPubMed
Zhang, P et al. (2019) The physical microenvironment of hematopoietic stem cells and its emerging roles in engineering applications. Stem Cell Research & Therapy 10, 113.CrossRefGoogle ScholarPubMed
Le, PM, Andreeff, M and Battula, VL (2018) Osteogenic niche in the regulation of normal hematopoiesis and leukemogenesis. Haematologica 103, 1945.CrossRefGoogle ScholarPubMed
Kajiume, T et al. (2021) Osteoblastic adherence regulates hematopoietic stem cell self-renewal and differentiation: a conceptional in vitro and in vivo study. Stem Cell Investigation 8, 21.CrossRefGoogle ScholarPubMed
Mangialardi, G, Cordaro, A and Madeddu, P (2016) The bone marrow pericyte: an orchestrator of vascular niche. Regenerative Medicine 11, 883895.CrossRefGoogle ScholarPubMed
Hayashi, Y et al. (2022) MDS cells impair osteolineage differentiation of MSCs via extracellular vesicles to suppress normal hematopoiesis. Cell Reports 39, 110805.CrossRefGoogle ScholarPubMed
Carvalho, MS et al. (2018) Biomimetic matrices for rapidly forming mineralized bone tissue based on stem cell-mediated osteogenesis. Scientific Reports 8, 116.CrossRefGoogle ScholarPubMed
Duarte, D et al. (2018) Inhibition of endosteal vascular niche remodeling rescues hematopoietic stem cell loss in AML. Cell Stem Cell 22, 6477. e6.CrossRefGoogle ScholarPubMed
Kunisaki, Y et al. (2013) Arteriolar niches maintain haematopoietic stem cell quiescence. Nature 502, 637643.CrossRefGoogle ScholarPubMed
Nobre, AR et al. (2021) Bone marrow NG2+/nestin+ mesenchymal stem cells drive DTC dormancy via TGF-β2. Nature Cancer 2, 327339.CrossRefGoogle Scholar
De Rooij, JD, Zwaan, CM and van den Heuvel-Eibrink, M (2015) Pediatric AML: from biology to clinical management. Journal of Clinical Medicine 4, 127149.CrossRefGoogle ScholarPubMed
Alimperti, S and Andreadis, ST (2015) CDH2 and CDH11 act as regulators of stem cell fate decisions. Stem Cell Research 14, 270282.CrossRefGoogle ScholarPubMed
Zhao, M et al. (2019) N-cadherin-expressing bone and marrow stromal progenitor cells maintain reserve hematopoietic stem cells. Cell Reports 26, 652669. e6.CrossRefGoogle ScholarPubMed
Tabe, Y and Konopleva, M (2015) Role of microenvironment in resistance to therapy in AML. Current Hematologic Malignancy Reports 10, 96103.CrossRefGoogle ScholarPubMed
Barwe, SP, Quagliano, A and Gopalakrishnapillai, A (2017) Eviction from the sanctuary: development of targeted therapy against cell adhesion molecules in acute lymphoblastic leukemia. Seminars in Oncology 44, 101112.CrossRefGoogle ScholarPubMed
Arai, F et al. (2012) Role of N-cadherin in the regulation of hematopoietic stem cells in the bone marrow niche. Annals of the New York Academy of Sciences 1266, 7277.CrossRefGoogle ScholarPubMed
Zhi, L et al. (2010) Enrichment of N-Cadherin and Tie2-bearing CD34+/CD38−/CD123+ leukemic stem cells by chemotherapy-resistance. Cancer Letters 296, 6573.CrossRefGoogle ScholarPubMed
Chiarini, F et al. (2020) The role played by Wnt/β-catenin signaling pathway in acute lymphoblastic leukemia. International Journal of Molecular Sciences 21, 1098.CrossRefGoogle ScholarPubMed
Dandekar, S et al. (2014) Wnt inhibition leads to improved chemosensitivity in paediatric acute lymphoblastic leukaemia. British Journal of Haematology 167, 8799.CrossRefGoogle ScholarPubMed
Gang, EJ et al. (2014) Small-molecule inhibition of CBP/catenin interactions eliminates drug-resistant clones in acute lymphoblastic leukemia. Oncogene 33, 21692178.CrossRefGoogle ScholarPubMed
Grüninger, PK et al. (2022) Functional characterization of the PI3K/AKT/mTOR signaling pathway for targeted therapy in B-precursor acute lymphoblastic leukemia. Cancer Gene Therapy 29, 17511760.CrossRefGoogle ScholarPubMed
Liang, F et al. (2019) The crosstalk between STAT3 and p53/RAS signaling controls cancer cell metastasis and cisplatin resistance via the Slug/MAPK/PI3K/AKT-mediated regulation of EMT and autophagy. Oncogenesis 8, 59.CrossRefGoogle ScholarPubMed
Ma, Y et al. (2022) Triptolide inhibits T-cell acute lymphoblastic leukaemia by affecting aberrant epigenetic events in the Wnt signalling pathway. Journal of Chemotherapy 11, 110.CrossRefGoogle ScholarPubMed
Murray, HC et al. (2023) Synergistic targeting of DNA-PK and KIT signaling pathways in KIT mutant acute myeloid leukemia. Molecular & Cellular Proteomics 22, 100503.CrossRefGoogle ScholarPubMed
Nepstad, I et al. (2019) Effects of insulin and pathway inhibitors on the PI3K-Akt-mTOR phosphorylation profile in acute myeloid leukemia cells. Signal Transduction and Targeted Therapy 4, 20.CrossRefGoogle ScholarPubMed
Nepstad, I et al. (2018) Clonal heterogeneity reflected by PI3K-AKT-mTOR signaling in human acute myeloid leukemia cells and its association with adverse prognosis. Cancers 10, 332.CrossRefGoogle ScholarPubMed
Perry, JM et al. (2020) Overcoming Wnt–β-catenin dependent anticancer therapy resistance in leukaemia stem cells. Nature Cell Biology 22, 689700.CrossRefGoogle ScholarPubMed
Pothongsrisit, S and Pongrakhananon, V (2021) Targeting the PI3K/AKT/mTOR signaling pathway in lung cancer: an update regarding potential drugs and natural products. Molecules 26, 4100.Google Scholar
Rabanal-Ruiz, Y, Otten, EG and Korolchuk, VI (2017) mTORC1 as the main gateway to autophagy. Essays in Biochemistry 61, 565584.Google ScholarPubMed
Wang, Y et al. (2010) The Wnt/β-catenin pathway is required for the development of leukemia stem cells in AML. Science 327, 16501653.CrossRefGoogle ScholarPubMed
Zhang, Y and Manning, BD (2015) mTORC1 signaling activates NRF1 to increase cellular proteasome levels. Cell Cycle 14, 20112017.CrossRefGoogle ScholarPubMed
Zou, Z et al. (2016) Targeted inhibition of rictor/mTORC2 in cancer treatment: a new era after rapamycin. Current Cancer Drug Targets 16, 288304.CrossRefGoogle ScholarPubMed
Yarom, N et al. (2013) Phase I clinical trial of Exherin (ADH-1) in patients with advanced solid tumors. Current Clinical Pharmacology 8, 8188.Google ScholarPubMed
Shintani, Y et al. (2008) ADH-1 suppresses N-cadherin-dependent pancreatic cancer progression. International Journal of Cancer 122, 7177.CrossRefGoogle ScholarPubMed
Li, H, Price, DK and Figg, WD (2007) ADH1, an N-cadherin inhibitor, evaluated in preclinical models of angiogenesis and androgen-independent prostate cancer. Anti-Cancer Drugs 18, 563568.CrossRefGoogle ScholarPubMed
Lammens, T et al. (2012) N-cadherin in neuroblastoma disease: expression and clinical significance. PLoS ONE 7, e31206.CrossRefGoogle ScholarPubMed
Perotti, A et al. (2009) Clinical and pharmacological phase I evaluation of Exherin™(ADH-1), a selective anti-N-cadherin peptide in patients with N-cadherin-expressing solid tumours. Annals of Oncology 20, 741745.CrossRefGoogle ScholarPubMed
Guo, Z et al. (2018) Improvement of chemosensitivity and inhibition of migration via targeting tumor epithelial-to-mesenchymal transition cells by ADH-1-modified liposomes. Drug Delivery 25, 112121.CrossRefGoogle ScholarPubMed
Sun, Y et al. (2021) N-cadherin inhibitor creates a microenvironment that protect TILs from immune checkpoints and Treg cells. Journal for Immunotherapy of Cancer 9, e002138.CrossRefGoogle ScholarPubMed
Mrozik, KM et al. (2020) LCRF-0006, a small molecule mimetic of the N-cadherin antagonist peptide ADH-1, synergistically increases multiple myeloma response to bortezomib. FASEB BioAdvances 2, 339.CrossRefGoogle ScholarPubMed
Gour, BJ et al. (2000) Inventors Peptidomimetic modulators of cell adhesion. United States.Google Scholar
Vaisburg, A and Blaschuk, OW (2018) Inventors modulators of cell adhesion, methods and compositions therefor. United States.Google Scholar
Smits, IP, Blaschuk, OW and Willerth, SM (2020) Novel N-cadherin antagonist causes glioblastoma cell death in a 3D bioprinted co-culture model. Biochemical and Biophysical Research Communications 529, 162168.CrossRefGoogle Scholar
Mitchell, CD et al. (2005) Benefit of dexamethasone compared with prednisolone for childhood acute lymphoblastic leukaemia: results of the UK medical research council ALL97 randomized trial. British Journal of Haematology 129, 734745.CrossRefGoogle ScholarPubMed
Halsey, C and Escherich, G (2021) A ‘Goldilocks’ approach to CNS leukemia is needed. Blood 138, 288289.CrossRefGoogle ScholarPubMed
Peirs, S et al. (2014) ABT-199 mediated inhibition of BCL-2 as a novel therapeutic strategy in T-cell acute lymphoblastic leukemia. Blood. The Journal of the American Society of Hematology 124, 37383747.Google ScholarPubMed
Scherr, M et al. (2019) Optimized induction of mitochondrial apoptosis for chemotherapy-free treatment of BCR-ABL+ acute lymphoblastic leukemia. Leukemia 33, 13131323.CrossRefGoogle ScholarPubMed
Ragon, BK et al. (2017) Buparlisib, a PI3K inhibitor, demonstrates acceptable tolerability and preliminary activity in a phase I trial of patients with advanced leukemias. American Journal of Hematology 92, 711.CrossRefGoogle Scholar
de Gooijer, MC et al. (2018) Buparlisib is a brain penetrable pan-PI3K inhibitor. Scientific Reports 8, 10784.CrossRefGoogle ScholarPubMed
Brown, JR et al. (2014) Idelalisib, an inhibitor of phosphatidylinositol 3-kinase p110δ, for relapsed/refractory chronic lymphocytic leukemia. Blood. The Journal of the American Society of Hematology 123, 33903397.Google ScholarPubMed
Miller, BW et al. (2015) FDA approval: idelalisib monotherapy for the treatment of patients with follicular lymphoma and small lymphocytic lymphoma. Clinical Cancer Research 21, 15251529.CrossRefGoogle ScholarPubMed
Eldfors, S et al. (2017) Idelalisib sensitivity and mechanisms of disease progression in relapsed TCF3-PBX1 acute lymphoblastic leukemia. Leukemia 31, 5157.CrossRefGoogle ScholarPubMed
Silic-Benussi, M et al. (2022) mTOR inhibition downregulates glucose-6-phosphate dehydrogenase and induces ROS-dependent death in T-cell acute lymphoblastic leukemia cells. Redox Biology 51, 102268.CrossRefGoogle ScholarPubMed
Place, AE et al. (2018) Phase I trial of the mTOR inhibitor everolimus in combination with multi-agent chemotherapy in relapsed childhood acute lymphoblastic leukemia. Pediatric Blood & Cancer 65, e27062.CrossRefGoogle ScholarPubMed
Alves, R et al. (2019) Everolimus in combination with imatinib overcomes resistance in chronic myeloid leukaemia. Medical Oncology 36, 110.CrossRefGoogle ScholarPubMed
Lang, F et al. (2020) A phase I study of a dual PI3-kinase/mTOR inhibitor BEZ235 in adult patients with relapsed or refractory acute leukemia. BMC Pharmacology and Toxicology 21, 114.CrossRefGoogle ScholarPubMed
Chen, X et al. (2018) Retrospective analysis of 36 fusion genes in 2479 Chinese patients of de novo acute lymphoblastic leukemia. Leukemia Research 72, 99104.CrossRefGoogle ScholarPubMed
Amado, NG et al. (2014) Isoquercitrin suppresses colon cancer cell growth in vitro by targeting the Wnt/β-catenin signaling pathway. Journal of Biological Chemistry 289, 3545635467.CrossRefGoogle ScholarPubMed
Chen, B et al. (2009) Small molecule-mediated disruption of Wnt-dependent signaling in tissue regeneration and cancer. Nature Chemical Biology 5, 100107.CrossRefGoogle ScholarPubMed
Liu, N et al. (2011) High levels of β-catenin signaling reduce osteogenic differentiation of stem cells in inflammatory microenvironments through inhibition of the noncanonical Wnt pathway. Journal of Bone and Mineral Research 26, 20822095.CrossRefGoogle ScholarPubMed
Pan, F et al. (2018) Inhibitory effects of XAV939 on the proliferation of small-cell lung cancer H446 cells and Wnt/β-catenin signaling pathway in vitro. Oncology Letters 16, 19531958.Google ScholarPubMed
Sohn, S-H et al. (2019) INC280 inhibits Wnt/β-catenin and EMT signaling pathways and its induce apoptosis in diffuse gastric cancer positive for c-MET amplification. BMC Research Notes 12, 17.CrossRefGoogle ScholarPubMed
Figure 0

Table 1. Comparison of paediatric ALL protocols from the UK and two European countries, including study size and number of patients affected by adverse events

Figure 1

Figure 1. Schematic diagram of the BM microenvironment under normal conditions and following leukaemogenesis and treatment in AML (top right) and ALL (top left). After leukaemogenesis and treatment, the microenvironment is remodelled, pro-inflammatory and anti-angiogenic cytokines are produced resulting in the loss of vasculature in the endosteal and osteoblastic cells. Adapted from Refs 26, 27, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63.

Figure 2

Figure 2. Schematic diagram of the pathways and transcription factors associated with CDH2, including the PI3K/Akt/mTOR pathway and the Wnt/β-catenin pathway. Arrows represent activation; bars represent inhibition, double-ended arrows in the pathway indicate upregulation of a molecule results in downregulation of the other and vice versa. Adapted from Refs 71, 72, 73, 74, 85.

Figure 3

Figure 3. ADH-1 competitively binds to CDH2 on BM cells, preventing leukaemia–niche cell binding of leukaemia cells within the BM microenvironment. Adapted from Refs 38, 86.

Figure 4

Table 2. List of the therapeutics, their targets and their progressions through clinical trials