Next Article in Journal
Developmental Delay and Male-Biased Sex Ratio in esr2b Knockout Zebrafish
Previous Article in Journal
Evaluation of Toll-like Receptor 4 (TLR4) Involvement in Human Atrial Fibrillation: A Computational Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Delving into the Metabolism of Sézary Cells: A Brief Review

1
BRIC, BoRdeaux Institute of onCology, UMR 1312, Inserm, Université de Bordeaux, 33000 Bordeaux, France
2
Center Jacques Loiselet for Medical Genetics and Genomics (CGGM), Faculty of Medicine, Saint Joseph University, Beirut P.O. Box 17-5208, Lebanon
3
Dermatology Department, Centre Hospitalier Universitaire de Bordeaux, 33075 Bordeaux, France
*
Author to whom correspondence should be addressed.
Genes 2024, 15(5), 635; https://doi.org/10.3390/genes15050635
Submission received: 29 March 2024 / Revised: 14 May 2024 / Accepted: 15 May 2024 / Published: 17 May 2024
(This article belongs to the Section Human Genomics and Genetic Diseases)

Abstract

:
Primary cutaneous lymphomas (PCLs) are a heterogeneous group of lymphoproliferative disorders caused by the accumulation of neoplastic T or B lymphocytes in the skin. Sézary syndrome (SS) is an aggressive and rare form of cutaneous T cell lymphoma (CTCL) characterized by an erythroderma and the presence of atypical cerebriform T cells named Sézary cells in skin and blood. Most of the available treatments for SS are not curative, which means there is an urgent need for the development of novel efficient therapies. Recently, targeting cancer metabolism has emerged as a promising strategy for cancer therapy. This is due to the accumulating evidence that metabolic reprogramming highly contributes to tumor progression. Genes play a pivotal role in regulating metabolic processes, and alterations in these genes can disrupt the delicate balance of metabolic pathways, potentially contributing to cancer development. In this review, we discuss the importance of targeting energy metabolism in tumors and the currently available data on the metabolism of Sézary cells, paving the way for potential new therapeutic approaches aiming to improve clinical outcomes for patients suffering from SS.

1. Introduction: Primary Cutaneous Lymphomas

Primary cutaneous lymphomas (PCLs) represent a heterogeneous group of lymphoproliferative disorders, characterized by the presence of neoplastic T or B lymphocytes in the skin, with no evidence of extracutaneous disease at the time of the diagnosis. In western countries, approximately 75% to 80% of PCLs originate from T cells (cutaneous T cell lymphomas, CTCLs), while cutaneous B cell lymphomas (CBCLs) account only for 20% to 25% [1]. Mycosis fungoides (MF) and Sézary syndrome (SS) represent the most common forms of CTCL, with MF being the most prevalent type and comprising almost 50% of all PCLs, while SS is less common and constitutes around 2% of CTCLs [1,2,3]. In the following sections, we will mainly focus on SS, the aggressive and leukemic subtype of CTCL. While genetic alterations in SS can affect normal cellular functions, their impact on metabolism, including possible disruptions in metabolic pathways that control cellular energy and nutrient utilization within Sézary cells, remains an area of exploration. Understanding the underlying genetic alterations in SS and their impact on cellular metabolism holds promise for the development of new targeted therapies aimed at correcting metabolic abnormalities and halting disease progression.

1.1. Sézary Syndrome

SS is a rare and aggressive subtype of CTCL. It was first described in 1938 by Albert Sézary and Yves Bouvrain as erythroderma associated with superficial lymphadenopathy, along with the presence of “monstrous cells in the dermis and the blood” [4].
SS predominantly affects men, often over the age of 60, and progresses rapidly. The prognosis remains poor, with a 5-year survival rate of 30% [3,5]. Clinical signs of this disease include pruriginous erythroderma, alopecia, palmoplantar keratoderma and onychodystrophy [6]. SS is defined by the triad of erythroderma, generalized lymphadenopathy and the presence of a circulating atypical CD4+ T cell population with cerebriform nuclei (Sézary cells) in the skin, lymph nodes and peripheral blood [1]. Additionally, at least one of the following criteria is also required: an absolute number of Sézary cells ≥ 1000/μL or an expansion of the CD4+ T cell population (CD4/CD8 ratio ≥ 10 or CD4+/CD7− ≥ 30% or CD4+/CD26− ≥ 40%) [1,7,8].
Several biomarkers are identified in Sézary cells, including cell surface markers and genetic alterations. Cell surface markers include TCR-Vβ and other molecules such as the natural cytotoxicity receptor (NCR) NKp46/NCR1, CD85j/Ig-like transcript 2 (ILT2) receptor, CD158k/KIR3DL2 receptor and PD-1, in addition to cutaneous lymphocyte antigen (CLA) and C-C chemokine receptor type 4 (CCR4) [6,9,10,11]. There is evidence suggesting that Sézary cells typically display a central memory T cell phenotype (TCM), marked by the presence of CCR7, L-selectin, CD27, CCR4 and CLA [12]. However, other research indicated a broader range of phenotypes beyond the TCM profile, as well as a phenotypic plasticity of these neoplastic cells. This is based on the expression of cell surface markers associated with stem cell memory (TSCM), effector memory (TEM) and naive or transitional memory T cells (TTM) [13].
Genomic alterations reported in SS patients include most frequently TP53 deletion/mutation and other mutations as well as diverse gene expression profiles and miRNA expression [6,14]. Also, it was reported that Sézary cells activate telomerase to maintain their telomeres through a specific methylation pattern on the hTERT promoter [15,16]. In addition, studies uncovered a high degree of molecular heterogeneity between patients with SS but also within the malignant T cell population in the same patient [17,18,19]. On one hand, the study of the molecular pathogenesis of the disease contributes to the understanding of Sézary cells’ behavior and response to available treatments but, on the other hand, it highlights the important level of molecular inter- and intra-heterogeneity, thus revealing the complexity in understanding SS and its treatment strategies.

1.2. Therapy for Sézary Syndrome

Despite the progress made in implementing therapies for SS, such as targeted therapies, including monoclonal antibodies with long-term responses (i.e., mogamulizumab, directed against CCR4, which has demonstrated a substantial extension in progression-free survival) [20,21,22], none of the existing treatments are curative. The exception is hematopoietic stem cell transplantation (HSCT), which has been associated with significantly longer progression-free survival in patients with advanced-stage CTCLs [23]. However, this curative approach can only be proposed for young and relatively healthy patients with advanced disease with a compatible donor and is associated with a non-negligible morbidity and mortality [6]. Generally, patients are treated using a stage-adapted approach that prioritizes sustaining their quality of life [24]. Moreover, it is recommended that each case of PCL should be brought up at a local or regional multidisciplinary meeting (including members specialized in the field of PCLs) to confirm the diagnosis and therapeutic management of the patient. Handling CTCLs involves three main steps: (1) establishing a definitive diagnosis and classifying the lymphoma according to the EORTC/WHO criteria, (2) determining the stage of the disease and (3) agreeing on a therapeutic approach and reassessing efficacy and side effects in a regular way [25]. Treatment of Sézary syndrome is better managed with systemic therapies (Table 1).
While existing treatments for this lymphoma focus on obtaining a regression of the lesions or slow down its progression, most patients still die from their disease. Consequently, there is an urgent necessity to find alternative effective treatments. Nowadays, metabolic reprogramming is one of the pivotal hallmarks of cancer [26]. Several studies showed that targeting metabolism in cancer has emerged as a promising strategy for novel therapeutic approaches [27]. Hence, exploring the metabolic pathway of Sézary cells could lead to the development of novel therapies with improved outcomes.
Table 1. Systemic therapies for SS treatment.
Table 1. Systemic therapies for SS treatment.
Comments
Systemic Therapies
  • Retinoids
(e.g., bexarotene)
Retinoids derive from vitamin A whose function is to interact with nuclear receptors (retinoic acid (RAR) and retinoic X receptor (RXR)).
Bexarotene activates RXRs and induces apoptosis and prevents malignant T cell homing to the skin by downregulating CCR4 and E-selectin [3,28].
  • Interferon α
IFN-α activates CD8+ T cells and NK cells and suppresses cytokine production from malignant lymphoma cells [29].
  • Extracorporeal photopheresis (ECP)
Patients’ white blood cells are exposed ex vivo to a photosensitizing agent (8-methoxypsolaren) and then to UVA light. Cells are then reinfused in the patient. The purpose is to induce an immune response against malignant T cells [28].
  • Monoclonal antibodies
Mogamulizumab selectively binds to CCR4—which is highly expressed in malignant T cells—and induces antibody-dependent cellular toxicity, thus destroying tumor cells [20,30].
Lacutamab (IPH4102) binds to CD158k, a cell surface marker aberrantly expressed in patients with SS [31]. IPH4102 is designed to deplete CD158k-expressing cells via antibody-dependent cell cytotoxicity and phagocytosis [22,32].
  • Antibody–drug conjugate
Brentuximab vedotin (BV) is an anti-CD30 antibody attached to monomethyl auristatin E (MMAE), an antitubulin agent. The binding of BV to CD30 and its internalization will allow MMAE to exert its action and inhibit the assemblage of the microtubules, induce cell cycle arrest and cause cell death due to apoptosis of tumor cells [21,33].
  • Histone deacetylase inhibitors (HDACis)
(e.g., romidepsin)
Histone deacetylases (HDACs) are epigenetic regulators of gene expression. Their inhibitors (HDACis) are reported to induce upregulation of proapoptotic genes, DNA damage and alterations in the assembly of kinetochores [34,35].
  • Chemotherapy
Single or combined agents can be administered and act via various mechanisms including topoisomerase inhibition, blocking DNA synthesis and interference with essential cellular processes [36].

2. Energy Metabolism in Cancer Cells

2.1. The Warburg Effect

In the 1920s, Otto Warburg et al. introduced the idea of metabolic reprogramming in cancer research [37]. They showed that cancer cells increased their glucose uptake and metabolized most of it into lactate, even in the presence of oxygen. This phenomenon, also referred to as the “Warburg effect”, leads to ATP synthesis via aerobic glycolysis. Knowing that the energetic yield of glycolysis is very low compared to oxidative phosphorylation (OXPHOS), Warburg hypothesized that cancer cells exhibit an irreversible defect in mitochondrial activity, leading to a metabolic switch towards aerobic glycolysis in order to sustain the cell’s energy demands [37,38].
Since Warburg’s seminal studies, a large number of complex metabolic profiles have been discovered in tumor cells, ranging from highly glycolytic phenotypes [39,40] to completely opposite profiles characterized by increased reliance on OXPHOS [41,42]. Growing evidence indicates that numerous biochemical metabolic pathways undergo reorganization during malignant transformation to fulfill the energy demands of cancer cells and support their biosynthetic fluxes, which are necessary for providing cellular building blocks such as nucleic acids, proteins and membranes, as well as for redox resetting to establish a new redox balance within tumors [39,43,44]. Furthermore, alterations in energy transduction modalities have direct implications for controlling cancer gene expression via transcriptional and epigenetic mechanisms driven by oncometabolites [45]. The pivotal role of mitochondria in tumorigenesis through gene regulation has been evidenced in numerous studies investigating various oncometabolites generated by the tricarboxylic acid (TCA) cycle [39,44] and glutaminolysis [46].

2.2. Role of Glycolysis in Cancer

When oxygen is available, normal cells produce ATP via OXPHOS in the mitochondria. However, cancer cells can prioritize energy production through aerobic glycolysis (Warburg effect). Albeit less efficient than OXPHOS, this metabolic pathway is characterized by high-speed energy production that supports enhanced cell proliferation [47]. In addition to fast ATP production, glycolysis generates many intermediates that can be used as substrates in other anabolic pathways to synthetize cellular components such as lipids, amino acids and nucleotides [40]. Thus, glycolysis fuels anabolic pathways to produce macromolecules that are required for cancer cells’ growth and proliferation. Moreover, glycolysis also contributes to migration of tumor cells. A study conducted by Shiraishi et al. demonstrated that higher glycolysis was associated with increased cytoskeletal remodeling, inducing cellular migration in prostate and breast cancer cells [48]. Furthermore, in the same article, the authors reported that blocking glycolysis impaired cancer cells’ migration, and mitochondrially derived ATP was insufficient to compensate glycolysis [48]. Glycolytic tumor cells convert glucose to lactate (Figure 1) which, once synthesized, is excreted outside of the cell, acidifying the pH in the tumor microenvironment (TME) [49]. Released lactate is reported to contribute significantly to tumor progression via different mechanisms such as angiogenesis, immune escape, cell migration and metastasis [50,51,52].

2.3. Role of Mitochondrial Metabolism in Cancer

Numerous findings demonstrated that mitochondrial metabolism is mainly predominant in some types of cancer cells and that this metabolic pathway contributes to cancer progression. Fogal et al. showed that OXPHOS supports tumor maintenance and malignancy. Knockdown of p32 protein, that is frequently overexpressed in some tumors, induced a significant metabolic switch from OXPHOS towards glycolysis in human cancer cells, resulting in impaired cell proliferation in vitro and tumor growth in vivo. Moreover, high levels of glycolysis in the absence of OXPHOS did not support tumor growth, suggesting that reliance on glycolysis alone may not be very beneficial in cancer cells [53]. Although aerobic glycolysis is known to promote metastasis, studies showed that mitochondrial metabolism also appears to contribute to the migration and invasion process of cancer cells. For instance, silencing of peroxisome proliferator-activated receptor-γ coactivator (PGC1-α), an inducer of mitochondrial biogenesis, impaired mitochondrial generation and OXPHOS, consequently leading to a decrease in metastatic activity in breast cancer cells [54]. Indeed, mitochondria play a significant role in tumor progression, yet they are also involved in resistance to treatment in cancer cells. For instance, docetaxel-resistant prostate cancer cells were found to have a more efficient respiratory phenotype compared to sensitive cells [55].

2.4. Targeting Cancer Metabolism

2.4.1. Targeting Glycolysis

It has been shown that aerobic glycolysis and mitochondrial metabolism can significantly contribute to the malignant phenotype of tumors. Thus, targeting energy metabolism has emerged as a promising strategy for cancer therapy and numerous metabolism-based drugs have been developed or are being tested in clinical trials [27]. Glycolytic phenotype is associated with an upregulation of glycolysis-related enzymes and transporters, making them attractive targets for cancer therapy. Glucose transporters (GLUTs) facilitate glucose uptake and their expression is often increased in different types of tumors. It was reported that treating breast cancer cells and non-small cell lung cancer (NSCLC) cells with antibodies targeting Glut-1 reduced proliferation in human cancer cell lines in vitro and enhanced apoptosis induced by chemotherapeutic agents [56]. Glut-1 knockdown has also been shown to suppress glycolysis and proliferation, leading to cell cycle arrest in prostate cancer cell line [57]. Additionally, recent investigations highlighted the significance of palmitoylation for maintaining Glut-1’s localization in the plasma membrane. DHHC9 is the enzyme responsible for this post-translational modification and its knockdown hampers glycolysis, cell proliferation and glioblastoma tumorigenesis in vivo [58]. Various studies also focused on targeting enzymes involved in the glycolytic pathway. Hexokinase 2 (HK2) is the first rate-limiting enzyme in glycolysis that catalyzes the conversion of glucose to glucose-6-phosphate (G6P) (Figure 1). This enzyme has been reported to be highly expressed in malignant tumors [59,60]. HK2’s expression was linked to resistance to gemcitabine (GEM) therapy in pancreatic tumor. Knockdown of this enzyme decreased cancer cell proliferation, migration and viability and promoted cell apoptosis in vitro. Moreover, reducing HK2 levels resulted in enhanced sensitivity to GEM treatment both in vitro and in vivo [61]. Indeed, oral administration of benitrobenrazide (BNBZ), an inhibitor that directly binds to HK2 (Figure 1), inhibited tumor growth in mouse xenograft models. In vitro, BNBZ is reported to impair glycolysis in cancer cells overexpressing HK2, as well as their proliferation, and to induce their apoptosis [62].
Another rate-limiting enzyme in the glycolytic pathway is phosphosfructokinase-1 (PFK-1). This enzyme catalyzes the phosphorylation of fructose-6-phosphate (F-6-P) to fructose 1,6-bisphosphate (F-1,6bisP). PFK-1 activity is regulated by PFKFB3 which converts F-6-P to fructose-2,6-bisphosphate (F-2,6-BP). PFK-1 can be activated by F-2,6-BP which is itself under the control of PFKFB3 [63] (Figure 1). Increased PFKFB3 activity has been reported in many cancer types, leading to the development of several inhibitors targeting this enzyme with promising results such as impaired migration and invasion capacity of cancer cells [64,65]. Moreover, pyruvate kinase M2 (PKM2) is the final rate-limiting enzyme in glycolysis. It converts phosphoenolpyruvate (PEP) to pyruvate (Figure 1). PKM2 is highly expressed in many cancers and is involved in promoting tumorigenesis, making it an attractive candidate for cancer therapy [66]. Reports showed that the inhibition of PKM2 can suppress proliferation, induce apoptosis and restore drug sensitivity in cancer cells [67,68].

2.4.2. Targeting Mitochondrial Metabolism

Several agents have also been developed to target mitochondrial metabolism in cancer cells. They belong to the family of mitocans whose mechanism of action relies on disrupting key actors of mitochondrial metabolism such as the enzymes of the Krebs cycle, the electron transport chain (ETC), proteins belonging to the Bcl-2 family and mitochondrial DNA. Some inhibitors also target anabolic pathways such as glutamine metabolism and fatty acid β-oxidation [38].
It is known that pyruvate can be converted into acetyl-CoA via decarboxylation that occurs in the mitochondria. This step is a critical linkage between glycolysis and the Krebs cycle. The enzyme complex responsible for this conversion is the pyruvate dehydrogenase complex (PDC) (Figure 2). Pyruvate dehydrogenase kinases (PDKs) act as inhibitors of the PDC and are present in four isoforms (PDK1–4). Their aberrant expression was reported in many types of cancer, making this kinase an attractive target in cancer therapy [63]. Evidence suggests that the inhibition of PDKs can lead to an increase in OXPHOS, therefore limiting the Warburg effect which promotes tumorigenesis [69]. Dichloroacetate (DCA) is an inhibitor of PDKs, and its role has been reported in glioblastoma. Indeed, DCA treatment in glioblastoma cells targeted PDK2 and induced depolarization of mitochondria, apoptosis, inhibition of hypoxia-inducible factor 1-alpha (HIF1-α), activation of p53 and suppression of angiogenesis both in vitro and in vivo [70].
The Krebs cycle, also known as the TCA cycle, represents a series of biochemical reactions taking place in the mitochondrial matrix. It plays an important role in the cell by providing redox balance, energy and intermediates for macromolecule synthesis [71]. Evidence showed that mutations in genes encoding enzymes of the TCA cycle have been associated with cancer progression [72]. For instance, mutations of isocitrate dehydrogenases 1 and 2 (IDH1 and IDH2) are commonly found in hematologic malignancies and solid tumors. These enzymes catalyze the conversion of isocitrate to α-ketoglutarate (αKG) (Figure 2). Mutations in IDH lead to the loss of its typical catalytic activity while acquiring a novel function, the conversion of αKG to the oncometabolite 2-hydroxyglutarate (2-HG) which contributes to tumorigenesis. AG-221 (enasidenib) is an oral inhibitor of mutant IDH2. This inhibitor is currently FDA-approved for the treatment of acute myeloid leukemia (AML) and other solid tumors and has shown promising results [63,73]. I-8 is another inhibitor targeting IDH1 (Figure 2). This compound was found to inhibit the production of 2-HG and suppress histone methylation. In addition, I-8 induced cellular differentiation and decreased tumor stem cell characteristics in vitro and in vivo [74]. The glutamine pathway has also attracted a lot of attention in cancer therapy. Glutamine serves as an energy source in many cancer types, providing nutrients and precursor molecules for their growth [75]. Glutamine enters the cell via transporters and can be converted into glutamate by glutaminases (GLSs). Then, glutamate is converted to αKG, the intermediate of the TCA cycle, via glutamate dehydrogenase (GDH) (Figure 2). Many studies are currently focusing on inhibiting GLS, which has been proven to have a critical role in various types of cancer [63]. CB-839 is an orally bioavailable inhibitor of GLS in phase I/II clinical trials and has demonstrated antitumor effects in triple-negative breast cancer (TNBC) [76]. Moreover, the ETC in the inner mitochondrial membrane has also been exploited for cancer therapy. It consists of a series of fixed protein complexes (I–IV) and mobile electron carriers (coenzyme Q and cytochrome c) where electrons are transferred, generating a proton gradient used to produce ATP (Figure 2). Inhibitors that are being developed aim to target these different complexes of the inner mitochondrial membrane [77,78].
A growing body of evidence has suggested that metabolism plays a crucial role in the development of resistance and metastasis [79]. Therefore, combining targeted therapies with drugs that exploit vulnerabilities in energy metabolism could lead to significant outcomes in overcoming drug resistance, a prevalent challenge in cancer treatment. For example, melanoma cells that acquired resistance to anti-BRAF treatments largely rely on complex-I-dependent OXPHOS [80,81]. Interestingly, OXPHOS inhibitors or mitochondrial uncouplers have been shown to be sensitive to vemurafenib in BRAF-mutant melanoma cell lines [81]. Similarly, BRAFV600E inhibition was significantly boosted both in vitro and in murine models when mitochondrial ETC I was inhibited by antidiabetic biguanides such as metformin [82,83]. However, metformin showed no efficacy in clinical trials, and the supra-pharmacological doses used in pre-clinical stages do not allow for definitive conclusions regarding its potential anticancer mechanism of action [84]. Another mechanism by which BRAF-mutant cells develop resistance to BRAF inhibitors is through upregulation of glutamine uptake and activation of GLS. Consequently, treatment with a GLS inhibitor, such as BPTES, can re-sensitize resistant cells to BRAF inhibition [85]. However, GLS inhibitors have not progressed to the clinical stage due to lack of efficacy, highlighting the need for innovative approaches to advance the field of metabolic inhibition in cancer therapy.

3. Metabolism of Sézary Cells: State of the Art

In healthy quiescent CD4+ T cells, metabolism primarily relies on OXPHOS to generate ATP. Upon antigen recognition, metabolism shifts towards an anabolic profile, increasing glucose and amino acid uptake, which better supports clonal expansion and effector phenotype [86]. However, our understanding of the energy metabolism in tumoral T cells, particularly in conditions like SS, remains incomplete.
In a recent study, single-cell RNA sequencing was used to assess the transcriptional profiles of the malignant T cell population in SS samples [87]. This analysis was conducted on four SS blood samples, including two SS patients where both blood and skin samples were analyzed. In the peripheral blood of advanced-stage SS, a heterogenous expansion of T cell clonotypes was identified and each sample exhibited the expansion of a dominant clone as well as of other less abundant ones. Gene expression signatures for each clonotype were established. The results confirm the genetic heterogeneity of Sézary cells, both between patients and within the same patient. Strikingly, the genetic signatures reveal, for the first time, that Sézary cells consistently enhance the expression of OXPHOS genes among a significant number of patients. Additionally, a comparison of T cell clonotype expansion between the blood and the skin of SS patients with advanced disease revealed that one dominant clonotype was expanded in both compartments in each patient. Across all studied patients, this major clonotype revealed common upregulated pathways including OXPHOS.
This finding shows that despite high heterogeneity in malignant SS clones, they were found to upregulate OXPHOS, suggesting a potential homogeneity in this metabolic pathway across SS cases. However, a study based on a larger sample size of patients could be considered to validate this observation. If the data are confirmed, this could be intriguing for therapeutic targeting as they indicate potential vulnerability of Sézary cells that could be exploited for treatment strategies which will focus on disrupting OXPHOS.
Additionally, this major clonotype showed other common upregulated pathways such as HER-2 and sirtuin. Sirtuins control the expression of genes involved in the metabolism via histone deacetylation. They also exert post-translational regulation on various substrates by removing chemical modifications, thereby altering their activity [88]. The sirtuin pathway is poorly characterized in SS, but increasing evidence has confirmed the involvement of sirtuins in cancers with their particular capacity to modulate cellular metabolism [89]. In human breast cancer cell lines, sirtuin 3 (SIRT3), a member of the sirtuin family, acts by indirectly destabilizing HIF1-α, thus inhibiting glycolysis. This protein appears to be downregulated in human breast cancer, and this is associated with an increased expression of HIF1-α target genes [90]. Moreover, knockdown of SIRT3 in a metastatic colon cancer cell line leads to decreased mitochondrial biogenesis and mitochondrial dysfunction, consequently affecting cellular viability [91]. In light of this, elucidating the potential involvement of sirtuins in SS could aid in the development of targeted therapeutic interventions.
ATP metabolism was described to have an influence on the immune response of T cells derived from patients with SS [92]. It is known that CD39 and CD73 are ectonucleotidases catalyzing the conversion of ATP or ADP to AMP, and AMP to adenosine (ADO), respectively [93]. Increased concentrations of ADO metabolite in the TME are reported to induce immunosuppressive mechanisms [94]. Indeed, it has been reported that aberrant expression of CD39 and CD73 in circulating or skin-homing CD4+ T cells of SS patients can affect the metabolism of ATP as well as the production of ADO, thus impacting immune responses [92]. In this study, patients with higher levels of CD39+ cells (CD39high) demonstrated an increased activity in regard to hydrolyzing ATP into AMP. Conversely, patients with higher levels of CD73+ cells (CD73high) exhibited strong AMP metabolism, generating high concentrations of ADO. Moreover, coculture experiments revealed that CD39high cells produced large amounts of ADO from ATP. This overproduction of ADO was not detected in CD73high cells. Altogether, these results suggest that CD39high cells, alongside surrounding cell types that express CD73, can create an environment rich in ADO to induce immunosuppression [92].
A recent study published by Wartewig et al. showed that the inactivation of Pdcd1, which encodes the inhibitory receptor programmed death-1 (PD-1) known for inhibiting the antitumor immune response, enforces T cells with oncogenic signaling to adjust their metabolism towards aerobic glycolysis [95,96]. This was confirmed in both genetic mouse models and blood samples of patients with CTCL, diagnosed with either SS or leukemic MF. RNA-seq analysis showed enhanced activity of the PI3K-AKT-mTOR pathway, enforced HIF1-α activity and increased activity of glycolysis genes in Pdcd1-mutant lymphomas, compared to wild type Pdcd1. These results uncover a vulnerability in CTCL-deficient in Pdcd1, that can further be exploited in the implementation of novel therapies that involve targeting energy metabolism.
Another study also described the involvement of the PI3K/AKT/mTOR signaling pathway in Sézary cells [97]. They identified that Sézary cells residing in the skin display a higher proliferation rate compared to those circulating in the blood. Interestingly, skin-resident Sézary cells exhibit a higher activation of mTORC1/mTORC2 compared to blood-circulating cells. This activation is influenced by SDF-1 and CCL21 chemokines, triggering mTORC1 signaling, which is known to be a central regulator of cell metabolism. Additionally, genetic alterations within the PI3K/AKT/mTOR cascade in Sézary cells strongly impact their cellular energy. For instance, the authors reported that loss of PTEN and LKB1, which attenuate the activation of mTORC1 under normal conditions, can lead to constitutive activation of mTORC1 in Sézary cells [98,99]. This induces a metabolic shift towards aerobic glycolysis. These results clearly show that metabolism might play a role in SS. Hence, conducting a more exhaustive study would be necessary to validate these observations.
All together, these findings emphasize the importance of considering metabolic reprogramming in the setting of SS.

4. Conclusions and Perspectives

In conclusion, available data regarding Sézary cells’ metabolism remain very scarce. A better understanding of the energy metabolism of these cells is primordial and could lead to a better understanding of the pathogenesis of SS and ideally a better therapeutic management of SS patients in the era of precision medicine and targeted therapies. Hence, further research is needed to decipher metabolic pathways behind Sézary cells’ survival. This knowledge will allow us to disrupt Sézary cells’ energy balance and to propose effective and well-tolerated therapeutic strategies, in combination with current treatments or in addition to the treatments offered today.

Funding

This work was supported by research grants from La ligue contre le Cancer Comité Gironde grant R24030GG, La société française de dermatologie grant R24062GG, l’université de Bordeaux, l’INSERM and a gift from Kyowa Kirin Pharma SAS grant D23003GG.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

The authors were grateful to La Region Nouvelle Aquitaine, France for the cofunding of our research project with a PhD studentship to C.C.

Conflicts of Interest

The authors declare no conflicts of interests.

References

  1. Willemze, R.; Cerroni, L.; Kempf, W.; Berti, E.; Facchetti, F.; Swerdlow, S.H.; Jaffe, E.S. The 2018 update of the WHO-EORTC classification for primary cutaneous lymphomas. Blood 2019, 133, 1703–1714. [Google Scholar] [CrossRef] [PubMed]
  2. Hristov, A.C.; Tejasvi, T.; Wilcox, R.A. Cutaneous T-cell lymphomas: 2021 update on diagnosis, risk-stratification, and management. Am. J. Hematol. 2021, 96, 1313–1328. [Google Scholar] [CrossRef] [PubMed]
  3. García-Díaz, N.; Piris, M.Á.; Ortiz-Romero, P.L.; Vaqué, J.P. Mycosis Fungoides and Sézary Syndrome: An Integrative Review of the Pathophysiology, Molecular Drivers, and Targeted Therapy. Cancers 2021, 13, 1931. [Google Scholar] [CrossRef] [PubMed]
  4. Shakeri, A. Albert Sézary-The Man, the Cell, and the Syndrome. JAMA Dermatol. 2018, 154, 496–497. [Google Scholar] [CrossRef] [PubMed]
  5. Najidh, S.; Tensen, C.P.; van der Sluijs-Gelling, A.J.; Teodosio, C.; Cats, D.; Mei, H.; Kuipers, T.B.; Out-Luijting, J.J.; Zoutman, W.H.; van Hall, T.; et al. Improved Sézary cell detection and novel insights into immunophenotypic and molecular heterogeneity in Sézary syndrome. Blood 2021, 138, 2539–2554. [Google Scholar] [CrossRef] [PubMed]
  6. Cristofoletti, C.; Narducci, M.G.; Russo, G. Sézary Syndrome, recent biomarkers and new drugs. Chin. Clin. Oncol. 2019, 8, 2. [Google Scholar] [CrossRef] [PubMed]
  7. Scarisbrick, J.J.; Hodak, E.; Bagot, M.; Stranzenbach, R.; Stadler, R.; Ortiz-Romero, P.L.; Papadavid, E.; Evison, F.; Knobler, R.; Quaglino, P.; et al. Blood classification and blood response criteria in mycosis fungoides and Sézary syndrome using flow cytometry: Recommendations from the EORTC cutaneous lymphoma task force. Eur. J. Cancer 2018, 93, 47–56. [Google Scholar] [CrossRef] [PubMed]
  8. Hristov, A.C.; Tejasvi, T.; Wilcox, R.A. Cutaneous T-cell lymphomas: 2023 update on diagnosis, risk-stratification, and management. Am. J. Hematol. 2023, 98, 193–209. [Google Scholar] [CrossRef] [PubMed]
  9. Dobos, G.; De Cevins, C.; Ly Ka So, S.; Jean-Louis, F.; Mathieu, S.; Ram-Wolff, C.; Resche-Rigon, M.; Bensussan, A.; Bagot, M.; Michel, L. The value of five blood markers in differentiating mycosis fungoides and Sézary syndrome: A validation cohort. Br. J. Dermatol. 2021, 185, 405–411. [Google Scholar] [CrossRef] [PubMed]
  10. Michel, L.; Jean-Louis, F.; Begue, E.; Bensussan, A.; Bagot, M. Use of PLS3, Twist, CD158k/KIR3DL2, and NKp46 gene expression combination for reliable Sézary syndrome diagnosis. Blood 2013, 121, 1477–1478. [Google Scholar] [CrossRef] [PubMed]
  11. Moins-Teisserenc, H.; Daubord, M.; Clave, E.; Douay, C.; Félix, J.; Marie-Cardine, A.; Ram-Wolff, C.; Maki, G.; Beldjord, K.; Homyrda, L.; et al. CD158k is a reliable marker for diagnosis of Sézary syndrome and reveals an unprecedented heterogeneity of circulating malignant cells. J. Investig. Dermatol. 2015, 135, 247–257. [Google Scholar] [CrossRef] [PubMed]
  12. Campbell, J.J.; Clark, R.A.; Watanabe, R.; Kupper, T.S. Sezary syndrome and mycosis fungoides arise from distinct T-cell subsets: A biologic rationale for their distinct clinical behaviors. Blood 2010, 116, 767–771. [Google Scholar] [CrossRef] [PubMed]
  13. Roelens, M.; Delord, M.; Ram-Wolff, C.; Marie-Cardine, A.; Alberdi, A.; Maki, G.; Homyrda, L.; Bensussan, A.; Bagot, M.; Toubert, A.; et al. Circulating and skin-derived Sézary cells: Clonal but with phenotypic plasticity. Blood 2017, 130, 1468–1471. [Google Scholar] [CrossRef] [PubMed]
  14. Chevret, E.; Merlio, J.-P. Sézary Syndrome: Translating Genetic Diversity into Personalized Medicine. J. Investig. Dermatol. 2016, 136, 1319–1324. [Google Scholar] [CrossRef] [PubMed]
  15. Chevret, E.; Andrique, L.; Prochazkova-Carlotti, M.; Ferrer, J.; Cappellen, D.; Laharanne, E.; Idrissi, Y.; Boettiger, A.; Sahraoui, W.; Ruiz, F.; et al. Telomerase functions beyond telomere maintenance in primary cutaneous T-cell lymphoma. Blood 2014, 123, 1850–1859. [Google Scholar] [CrossRef] [PubMed]
  16. Chebly, A.; Ropio, J.; Peloponese, J.-M.; Poglio, S.; Prochazkova-Carlotti, M.; Cherrier, F.; Ferrer, J.; Idrissi, Y.; Segal-Bendirdjian, E.; Chouery, E.; et al. Exploring hTERT promoter methylation in cutaneous T-cell lymphomas. Mol. Oncol. 2022, 16, 1931–1946. [Google Scholar] [CrossRef] [PubMed]
  17. Herrera, A.; Cheng, A.; Mimitou, E.P.; Seffens, A.; George, D.; Bar-Natan, M.; Heguy, A.; Ruggles, K.V.; Scher, J.U.; Hymes, K.; et al. Multimodal single-cell analysis of cutaneous T-cell lymphoma reveals distinct subclonal tissue-dependent signatures. Blood 2021, 138, 1456–1464. [Google Scholar] [CrossRef]
  18. Poglio, S.; Prochazkova-Carlotti, M.; Cherrier, F.; Gros, A.; Laharanne, E.; Pham-Ledard, A.; Beylot-Barry, M.; Merlio, J.-P. Xenograft and cell culture models of Sézary syndrome reveal cell of origin diversity and subclonal heterogeneity. Leukemia 2021, 35, 1696–1709. [Google Scholar] [CrossRef]
  19. Buus, T.B.; Willerslev-Olsen, A.; Fredholm, S.; Blümel, E.; Nastasi, C.; Gluud, M.; Hu, T.; Lindahl, L.M.; Iversen, L.; Fogh, H.; et al. Single-cell heterogeneity in Sézary syndrome. Blood Adv. 2018, 2, 2115–2126. [Google Scholar] [CrossRef] [PubMed]
  20. Beylot-Barry, M.; Quereux, G.; Nardin, C.; Duval-Modeste, A.-B.; Dereure, O.; Dalac-Rat, S.; Dobos, G.; Pham-Ledard, A.; Ram-Wolff, C.; D’Incan, M.; et al. Effectiveness of mogamulizumab in patients with Mycosis Fungoides or Sézary syndrome: A multicentre, retrospective, real-world French study. J. Eur. Acad. Dermatol. Venereol. JEADV 2023, 37, 1777–1784. [Google Scholar] [CrossRef] [PubMed]
  21. Lewis, D.J.; Haun, P.L.; Samimi, S.S.; Vittorio, C.C.; Villasenor-Park, J.; Barta, S.K.; Landsburg, D.J.; Svoboda, J.; Nasta, S.D.; Schuster, S.J.; et al. Brentuximab Vedotin for Relapsed or Refractory Sézary Syndrome. JAMA Dermatol. 2021, 157, 317–321. [Google Scholar] [CrossRef] [PubMed]
  22. Bagot, M.; Porcu, P.; Marie-Cardine, A.; Battistella, M.; William, B.M.; Vermeer, M.; Whittaker, S.; Rotolo, F.; Ram-Wolff, C.; Khodadoust, M.S.; et al. IPH4102, a first-in-class anti-KIR3DL2 monoclonal antibody, in patients with relapsed or refractory cutaneous T-cell lymphoma: An international, first-in-human, open-label, phase 1 trial. Lancet Oncol. 2019, 20, 1160–1170. [Google Scholar] [CrossRef] [PubMed]
  23. de Masson, A.; Beylot-Barry, M.; Ram-Wolff, C.; Mear, J.-B.; Dalle, S.; d’Incan, M.; Ingen-Housz-Oro, S.; Orvain, C.; Abraham, J.; Dereure, O.; et al. Allogeneic transplantation in advanced cutaneous T-cell lymphomas (CUTALLO): A propensity score matched controlled prospective study. Lancet 2023, 401, 1941–1950. [Google Scholar] [CrossRef] [PubMed]
  24. Latzka, J.; Assaf, C.; Bagot, M.; Cozzio, A.; Dummer, R.; Guenova, E.; Gniadecki, R.; Hodak, E.; Jonak, C.; Klemke, C.-D.; et al. EORTC consensus recommendations for the treatment of mycosis fungoides/Sézary syndrome—Update 2023. Eur. J. Cancer 2023, 195, 113343. [Google Scholar] [CrossRef] [PubMed]
  25. Beylot-Barry, M.; Dereure, O.; Vergier, B.; Barete, S.; Laroche, L.; Machet, L.; Delfau-Larue, M.-H.; D’Incan, M.; Grange, F.; Ortonne, N.; et al. Management of cutaneous T-cell lymphomas: Recommendations of the French Cutaneous Lymphoma Group. Ann. Dermatol. Venereol. 2010, 137, 611–621. [Google Scholar] [CrossRef] [PubMed]
  26. Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef] [PubMed]
  27. Stine, Z.E.; Schug, Z.T.; Salvino, J.M.; Dang, C.V. Targeting cancer metabolism in the era of precision oncology. Nat. Rev. Drug Discov. 2022, 21, 141–162. [Google Scholar] [CrossRef] [PubMed]
  28. Sethi, T.K.; Montanari, F.; Foss, F.; Reddy, N. How we treat advanced stage cutaneous T-cell lymphoma—Mycosis fungoides and Sézary syndrome. Br. J. Haematol. 2021, 195, 352–364. [Google Scholar] [CrossRef] [PubMed]
  29. Yoo, E.K.; Cassin, M.; Lessin, S.R.; Rook, A.H. Complete molecular remission during biologic response modifier therapy for Sézary syndrome is associated with enhanced helper T type 1 cytokine production and natural killer cell activity. J. Am. Acad. Dermatol. 2001, 45, 208–216. [Google Scholar] [CrossRef] [PubMed]
  30. Duvic, M.; Evans, M.; Wang, C. Mogamulizumab for the treatment of cutaneous T-cell lymphoma: Recent advances and clinical potential. Ther. Adv. Hematol. 2016, 7, 171–174. [Google Scholar] [CrossRef]
  31. Battistella, M.; Leboeuf, C.; Ram-Wolff, C.; Hurabielle, C.; Bonnafous, C.; Sicard, H.; Bensussan, A.; Bagot, M.; Janin, A. KIR3DL2 expression in cutaneous T-cell lymphomas: Expanding the spectrum for KIR3DL2 targeting. Blood 2017, 130, 2900–2902. [Google Scholar] [CrossRef]
  32. Marie-Cardine, A.; Viaud, N.; Thonnart, N.; Joly, R.; Chanteux, S.; Gauthier, L.; Bonnafous, C.; Rossi, B.; Bléry, M.; Paturel, C.; et al. IPH4102, a humanized KIR3DL2 antibody with potent activity against cutaneous T-cell lymphoma. Cancer Res. 2014, 74, 6060–6070. [Google Scholar] [CrossRef] [PubMed]
  33. Sanches, J.A.; Cury-Martins, J.; Abreu, R.M.; Miyashiro, D.; Pereira, J. Mycosis fungoides and Sézary syndrome: Focus on the current treatment scenario. An. Bras. Dermatol. 2021, 96, 458–471. [Google Scholar] [CrossRef] [PubMed]
  34. Lopez, A.T.; Bates, S.; Geskin, L. Current Status of HDAC Inhibitors in Cutaneous T-cell Lymphoma. Am. J. Clin. Dermatol. 2018, 19, 805–819. [Google Scholar] [CrossRef]
  35. Whittaker, S.J.; Demierre, M.-F.; Kim, E.J.; Rook, A.H.; Lerner, A.; Duvic, M.; Scarisbrick, J.; Reddy, S.; Robak, T.; Becker, J.C.; et al. Final results from a multicenter, international, pivotal study of romidepsin in refractory cutaneous T-cell lymphoma. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2010, 28, 4485–4491. [Google Scholar] [CrossRef] [PubMed]
  36. Alpdogan, O.; Kartan, S.; Johnson, W.; Sokol, K.; Porcu, P. Systemic therapy of cutaneous T-cell lymphoma (CTCL). Chin. Clin. Oncol. 2019, 8, 10. [Google Scholar] [CrossRef] [PubMed]
  37. Warburg, O. On the origin of cancer cells. Science 1956, 123, 309–314. [Google Scholar] [CrossRef] [PubMed]
  38. Martinez-Outschoorn, U.E.; Peiris-Pagés, M.; Pestell, R.G.; Sotgia, F.; Lisanti, M.P. Cancer metabolism: A therapeutic perspective. Nat. Rev. Clin. Oncol. 2017, 14, 11–31. [Google Scholar] [CrossRef] [PubMed]
  39. Ward, P.S.; Thompson, C.B. Metabolic reprogramming: A cancer hallmark even warburg did not anticipate. Cancer Cell 2012, 21, 297–308. [Google Scholar] [CrossRef] [PubMed]
  40. Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science 2009, 324, 1029–1033. [Google Scholar] [CrossRef] [PubMed]
  41. Ashton, T.M.; McKenna, W.G.; Kunz-Schughart, L.A.; Higgins, G.S. Oxidative Phosphorylation as an Emerging Target in Cancer Therapy. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2018, 24, 2482–2490. [Google Scholar] [CrossRef]
  42. Greene, J.; Segaran, A.; Lord, S. Targeting OXPHOS and the electron transport chain in cancer; Molecular and therapeutic implications. Semin. Cancer Biol. 2022, 86, 851–859. [Google Scholar] [CrossRef] [PubMed]
  43. Smolková, K.; Plecitá-Hlavatá, L.; Bellance, N.; Benard, G.; Rossignol, R.; Ježek, P. Waves of gene regulation suppress and then restore oxidative phosphorylation in cancer cells. Int. J. Biochem. Cell Biol. 2011, 43, 950–968. [Google Scholar] [CrossRef] [PubMed]
  44. Galluzzi, L.; Kepp, O.; Vander Heiden, M.G.; Kroemer, G. Metabolic targets for cancer therapy. Nat. Rev. Drug Discov. 2013, 12, 829–846. [Google Scholar] [CrossRef]
  45. Liu, Y.; Yang, C. Oncometabolites in Cancer: Current Understanding and Challenges. Cancer Res. 2021, 81, 2820–2823. [Google Scholar] [CrossRef]
  46. Villar, V.H.; Merhi, F.; Djavaheri-Mergny, M.; Durán, R.V. Glutaminolysis and autophagy in cancer. Autophagy 2015, 11, 1198–1208. [Google Scholar] [CrossRef]
  47. Lunt, S.Y.; Vander Heiden, M.G. Aerobic glycolysis: Meeting the metabolic requirements of cell proliferation. Annu. Rev. Cell Dev. Biol. 2011, 27, 441–464. [Google Scholar] [CrossRef] [PubMed]
  48. Shiraishi, T.; Verdone, J.E.; Huang, J.; Kahlert, U.D.; Hernandez, J.R.; Torga, G.; Zarif, J.C.; Epstein, T.; Gatenby, R.; McCartney, A.; et al. Glycolysis is the primary bioenergetic pathway for cell motility and cytoskeletal remodeling in human prostate and breast cancer cells. Oncotarget 2015, 6, 130–143. [Google Scholar] [CrossRef]
  49. Romero-Garcia, S.; Moreno-Altamirano, M.M.B.; Prado-Garcia, H.; Sánchez-García, F.J. Lactate Contribution to the Tumor Microenvironment: Mechanisms, Effects on Immune Cells and Therapeutic Relevance. Front. Immunol. 2016, 7, 52. [Google Scholar] [CrossRef]
  50. Hunt, T.K.; Aslam, R.; Hussain, Z.; Beckert, S. Lactate, with oxygen, incites angiogenesis. Adv. Exp. Med. Biol. 2008, 614, 73–80. [Google Scholar] [CrossRef] [PubMed]
  51. Fischer, K.; Hoffmann, P.; Voelkl, S.; Meidenbauer, N.; Ammer, J.; Edinger, M.; Gottfried, E.; Schwarz, S.; Rothe, G.; Hoves, S.; et al. Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood 2007, 109, 3812–3819. [Google Scholar] [CrossRef]
  52. Baumann, F.; Leukel, P.; Doerfelt, A.; Beier, C.P.; Dettmer, K.; Oefner, P.J.; Kastenberger, M.; Kreutz, M.; Nickl-Jockschat, T.; Bogdahn, U.; et al. Lactate promotes glioma migration by TGF-beta2-dependent regulation of matrix metalloproteinase-2. Neuro-oncology 2009, 11, 368–380. [Google Scholar] [CrossRef]
  53. Fogal, V.; Richardson, A.D.; Karmali, P.P.; Scheffler, I.E.; Smith, J.W.; Ruoslahti, E. Mitochondrial p32 protein is a critical regulator of tumor metabolism via maintenance of oxidative phosphorylation. Mol. Cell. Biol. 2010, 30, 1303–1318. [Google Scholar] [CrossRef]
  54. LeBleu, V.S.; O’Connell, J.T.; Gonzalez Herrera, K.N.; Wikman, H.; Pantel, K.; Haigis, M.C.; de Carvalho, F.M.; Damascena, A.; Domingos Chinen, L.T.; Rocha, R.M.; et al. PGC-1α mediates mitochondrial biogenesis and oxidative phosphorylation in cancer cells to promote metastasis. Nat. Cell Biol. 2014, 16, 992–1003, 1–15. [Google Scholar] [CrossRef] [PubMed]
  55. Ippolito, L.; Marini, A.; Cavallini, L.; Morandi, A.; Pietrovito, L.; Pintus, G.; Giannoni, E.; Schrader, T.; Puhr, M.; Chiarugi, P.; et al. Metabolic shift toward oxidative phosphorylation in docetaxel resistant prostate cancer cells. Oncotarget 2016, 7, 61890–61904. [Google Scholar] [CrossRef] [PubMed]
  56. Rastogi, S.; Banerjee, S.; Chellappan, S.; Simon, G.R. Glut-1 antibodies induce growth arrest and apoptosis in human cancer cell lines. Cancer Lett. 2007, 257, 244–251. [Google Scholar] [CrossRef] [PubMed]
  57. Xiao, H.; Wang, J.; Yan, W.; Cui, Y.; Chen, Z.; Gao, X.; Wen, X.; Chen, J. GLUT1 regulates cell glycolysis and proliferation in prostate cancer. Prostate 2018, 78, 86–94. [Google Scholar] [CrossRef] [PubMed]
  58. Zhang, Z.; Li, X.; Yang, F.; Chen, C.; Liu, P.; Ren, Y.; Sun, P.; Wang, Z.; You, Y.; Zeng, Y.-X.; et al. DHHC9-mediated GLUT1 S-palmitoylation promotes glioblastoma glycolysis and tumorigenesis. Nat. Commun. 2021, 12, 5872. [Google Scholar] [CrossRef] [PubMed]
  59. Chen, J.; Zhang, S.; Li, Y.; Tang, Z.; Kong, W. Hexokinase 2 overexpression promotes the proliferation and survival of laryngeal squamous cell carcinoma. Tumour Biol. J. Int. Soc. Oncodev. Biol. Med. 2014, 35, 3743–3753. [Google Scholar] [CrossRef] [PubMed]
  60. Patra, K.C.; Wang, Q.; Bhaskar, P.T.; Miller, L.; Wang, Z.; Wheaton, W.; Chandel, N.; Laakso, M.; Muller, W.J.; Allen, E.L.; et al. Hexokinase 2 is required for tumor initiation and maintenance and its systemic deletion is therapeutic in mouse models of cancer. Cancer Cell 2013, 24, 213–228. [Google Scholar] [CrossRef]
  61. Fan, K.; Fan, Z.; Cheng, H.; Huang, Q.; Yang, C.; Jin, K.; Luo, G.; Yu, X.; Liu, C. Hexokinase 2 dimerization and interaction with voltage-dependent anion channel promoted resistance to cell apoptosis induced by gemcitabine in pancreatic cancer. Cancer Med. 2019, 8, 5903–5915. [Google Scholar] [CrossRef] [PubMed]
  62. Zheng, M.; Wu, C.; Yang, K.; Yang, Y.; Liu, Y.; Gao, S.; Wang, Q.; Li, C.; Chen, L.; Li, H. Novel selective hexokinase 2 inhibitor Benitrobenrazide blocks cancer cells growth by targeting glycolysis. Pharmacol. Res. 2021, 164, 105367. [Google Scholar] [CrossRef] [PubMed]
  63. Farhadi, P.; Yarani, R.; Dokaneheifard, S.; Mansouri, K. The emerging role of targeting cancer metabolism for cancer therapy. Tumour Biol. J. Int. Soc. Oncodev. Biol. Med. 2020, 42, 1010428320965284. [Google Scholar] [CrossRef] [PubMed]
  64. De Oliveira, T.; Goldhardt, T.; Edelmann, M.; Rogge, T.; Rauch, K.; Kyuchukov, N.D.; Menck, K.; Bleckmann, A.; Kalucka, J.; Khan, S.; et al. Effects of the Novel PFKFB3 Inhibitor KAN0438757 on Colorectal Cancer Cells and Its Systemic Toxicity Evaluation In Vivo. Cancers 2021, 13, 1011. [Google Scholar] [CrossRef]
  65. Deng, J.; Cheng, Y.; Li, H.; He, X.; Yu, S.; Ma, J.; Li, X.; Chen, J.; Xiao, H.; Guan, H.; et al. PFKFB3 facilitates cell proliferation and migration in anaplastic thyroid carcinoma via the WNT/β-catenin signaling pathway. Endocrine 2024. [Google Scholar] [CrossRef] [PubMed]
  66. Zahra, K.; Dey, T.; Ashish; Mishra, S.P.; Pandey, U. Pyruvate Kinase M2 and Cancer: The Role of PKM2 in Promoting Tumorigenesis. Front. Oncol. 2020, 10, 159. [Google Scholar] [CrossRef] [PubMed]
  67. Goldberg, M.S.; Sharp, P.A. Pyruvate kinase M2-specific siRNA induces apoptosis and tumor regression. J. Exp. Med. 2012, 209, 217–224. [Google Scholar] [CrossRef] [PubMed]
  68. Yuan, S.; Qiao, T.; Zhuang, X.; Chen, W.; Xing, N.; Zhang, Q. Knockdown of the M2 Isoform of Pyruvate Kinase (PKM2) with shRNA Enhances the Effect of Docetaxel in Human NSCLC Cell Lines In Vitro. Yonsei Med. J. 2016, 57, 1312–1323. [Google Scholar] [CrossRef] [PubMed]
  69. Zhang, W.; Zhang, S.-L.; Hu, X.; Tam, K.Y. Targeting Tumor Metabolism for Cancer Treatment: Is Pyruvate Dehydrogenase Kinases (PDKs) a Viable Anticancer Target? Int. J. Biol. Sci. 2015, 11, 1390–1400. [Google Scholar] [CrossRef] [PubMed]
  70. Michelakis, E.D.; Sutendra, G.; Dromparis, P.; Webster, L.; Haromy, A.; Niven, E.; Maguire, C.; Gammer, T.-L.; Mackey, J.R.; Fulton, D.; et al. Metabolic modulation of glioblastoma with dichloroacetate. Sci. Transl. Med. 2010, 2, 31ra34. [Google Scholar] [CrossRef] [PubMed]
  71. Anderson, N.M.; Mucka, P.; Kern, J.G.; Feng, H. The emerging role and targetability of the TCA cycle in cancer metabolism. Protein Cell 2018, 9, 216–237. [Google Scholar] [CrossRef] [PubMed]
  72. Sainero-Alcolado, L.; Liaño-Pons, J.; Ruiz-Pérez, M.V.; Arsenian-Henriksson, M. Targeting mitochondrial metabolism for precision medicine in cancer. Cell Death Differ. 2022, 29, 1304–1317. [Google Scholar] [CrossRef] [PubMed]
  73. Kim, E.S. Enasidenib: First Global Approval. Drugs 2017, 77, 1705–1711. [Google Scholar] [CrossRef] [PubMed]
  74. Jia, P.; Wu, Y.; Du, H.; Yang, L.; Zhang, Z.; Ma, T.; Li, S.; Yuan, S.; Lu, L.; Zha, X. I-8, a novel inhibitor of mutant IDH1, inhibits cancer progression in vitro and in vivo. Eur. J. Pharm. Sci. Off. J. Eur. Fed. Pharm. Sci. 2019, 140, 105072. [Google Scholar] [CrossRef] [PubMed]
  75. Ghosh, P.; Vidal, C.; Dey, S.; Zhang, L. Mitochondria Targeting as an Effective Strategy for Cancer Therapy. Int. J. Mol. Sci. 2020, 21, 3363. [Google Scholar] [CrossRef] [PubMed]
  76. Gross, M.I.; Demo, S.D.; Dennison, J.B.; Chen, L.; Chernov-Rogan, T.; Goyal, B.; Janes, J.R.; Laidig, G.J.; Lewis, E.R.; Li, J.; et al. Antitumor activity of the glutaminase inhibitor CB-839 in triple-negative breast cancer. Mol. Cancer Ther. 2014, 13, 890–901. [Google Scholar] [CrossRef] [PubMed]
  77. Rohlena, J.; Dong, L.-F.; Ralph, S.J.; Neuzil, J. Anticancer drugs targeting the mitochondrial electron transport chain. Antioxid. Redox Signal. 2011, 15, 2951–2974. [Google Scholar] [CrossRef] [PubMed]
  78. Molina, J.R.; Sun, Y.; Protopopova, M.; Gera, S.; Bandi, M.; Bristow, C.; McAfoos, T.; Morlacchi, P.; Ackroyd, J.; Agip, A.-N.A.; et al. An inhibitor of oxidative phosphorylation exploits cancer vulnerability. Nat. Med. 2018, 24, 1036–1046. [Google Scholar] [CrossRef] [PubMed]
  79. Bergers, G.; Fendt, S.-M. The metabolism of cancer cells during metastasis. Nat. Rev. Cancer 2021, 21, 162–180. [Google Scholar] [CrossRef] [PubMed]
  80. Corazao-Rozas, P.; Guerreschi, P.; Jendoubi, M.; André, F.; Jonneaux, A.; Scalbert, C.; Garçon, G.; Malet-Martino, M.; Balayssac, S.; Rocchi, S.; et al. Mitochondrial oxidative stress is the Achille’s heel of melanoma cells resistant to Braf-mutant inhibitor. Oncotarget 2013, 4, 1986–1998. [Google Scholar] [CrossRef]
  81. Haq, R.; Shoag, J.; Andreu-Perez, P.; Yokoyama, S.; Edelman, H.; Rowe, G.C.; Frederick, D.T.; Hurley, A.D.; Nellore, A.; Kung, A.L.; et al. Oncogenic BRAF regulates oxidative metabolism via PGC1α and MITF. Cancer Cell 2013, 23, 302–315. [Google Scholar] [CrossRef] [PubMed]
  82. Roesch, A.; Vultur, A.; Bogeski, I.; Wang, H.; Zimmermann, K.M.; Speicher, D.; Körbel, C.; Laschke, M.W.; Gimotty, P.A.; Philipp, S.E.; et al. Overcoming intrinsic multidrug resistance in melanoma by blocking the mitochondrial respiratory chain of slow-cycling JARID1B(high) cells. Cancer Cell 2013, 23, 811–825. [Google Scholar] [CrossRef]
  83. Yuan, P.; Ito, K.; Perez-Lorenzo, R.; Del Guzzo, C.; Lee, J.H.; Shen, C.-H.; Bosenberg, M.W.; McMahon, M.; Cantley, L.C.; Zheng, B. Phenformin enhances the therapeutic benefit of BRAF(V600E) inhibition in melanoma. Proc. Natl. Acad. Sci. USA 2013, 110, 18226–18231. [Google Scholar] [CrossRef] [PubMed]
  84. Jaune, E.; Rocchi, S. Metformin: Focus on Melanoma. Front. Endocrinol. 2018, 9, 472. [Google Scholar] [CrossRef] [PubMed]
  85. Baenke, F.; Chaneton, B.; Smith, M.; Van Den Broek, N.; Hogan, K.; Tang, H.; Viros, A.; Martin, M.; Galbraith, L.; Girotti, M.R.; et al. Resistance to BRAF inhibitors induces glutamine dependency in melanoma cells. Mol. Oncol. 2016, 10, 73–84. [Google Scholar] [CrossRef] [PubMed]
  86. Bishop, E.L.; Gudgeon, N.; Dimeloe, S. Control of T Cell Metabolism by Cytokines and Hormones. Front. Immunol. 2021, 12, 653605. [Google Scholar] [CrossRef]
  87. Gaydosik, A.M.; Stonesifer, C.J.; Khaleel, A.E.; Geskin, L.J.; Fuschiotti, P. Single-Cell RNA Sequencing Unveils the Clonal and Transcriptional Landscape of Cutaneous T-Cell Lymphomas. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2022, 28, 2610–2622. [Google Scholar] [CrossRef] [PubMed]
  88. Dong, Z.; Cui, H. Sirtuins and cellular metabolism in cancers. In Sirtuin Biology in Cancer and Metabolic Disease; Elsevier: Amsterdam, The Netherlands, 2021; pp. 195–217. ISBN 978-0-12-822467-0. [Google Scholar]
  89. Mei, Z.; Zhang, X.; Yi, J.; Huang, J.; He, J.; Tao, Y. Sirtuins in metabolism, DNA repair and cancer. J. Exp. Clin. Cancer Res. CR 2016, 35, 182. [Google Scholar] [CrossRef] [PubMed]
  90. Finley, L.W.S.; Carracedo, A.; Lee, J.; Souza, A.; Egia, A.; Zhang, J.; Teruya-Feldstein, J.; Moreira, P.I.; Cardoso, S.M.; Clish, C.B.; et al. SIRT3 opposes reprogramming of cancer cell metabolism through HIF1α destabilization. Cancer Cell 2011, 19, 416–428. [Google Scholar] [CrossRef] [PubMed]
  91. Torrens-Mas, M.; Hernández-López, R.; Pons, D.-G.; Roca, P.; Oliver, J.; Sastre-Serra, J. Sirtuin 3 silencing impairs mitochondrial biogenesis and metabolism in colon cancer cells. Am. J. Physiol. Cell Physiol. 2019, 317, C398–C404. [Google Scholar] [CrossRef]
  92. Yakymiv, Y.; Marchisio, S.; Ortolan, E.; Bracci, C.; Senetta, R.; Rumore, M.R.; Tampieri, C.; Fia, M.; Ribero, S.; Funaro, A.; et al. CD39/CD73 dysregulation and adenosine metabolism contribute to T-cell immunosuppression in patients with Sézary syndrome. Blood 2023, 141, 111–116. [Google Scholar] [CrossRef] [PubMed]
  93. Longhi, M.S.; Robson, S.C.; Bernstein, S.H.; Serra, S.; Deaglio, S. Biological functions of ecto-enzymes in regulating extracellular adenosine levels in neoplastic and inflammatory disease states. J. Mol. Med. 2013, 91, 165–172. [Google Scholar] [CrossRef]
  94. Leone, R.D.; Emens, L.A. Targeting adenosine for cancer immunotherapy. J. Immunother. Cancer 2018, 6, 57. [Google Scholar] [CrossRef] [PubMed]
  95. Wartewig, T.; Daniels, J.; Schulz, M.; Hameister, E.; Joshi, A.; Park, J.; Morrish, E.; Venkatasubramani, A.V.; Cernilogar, F.M.; Van Heijster, F.H.A.; et al. PD-1 instructs a tumor-suppressive metabolic program that restricts glycolysis and restrains AP-1 activity in T cell lymphoma. Nat. Cancer 2023, 4, 1508–1525. [Google Scholar] [CrossRef] [PubMed]
  96. Chen, R.-Y.; Zhu, Y.; Shen, Y.-Y.; Xu, Q.-Y.; Tang, H.-Y.; Cui, N.-X.; Jiang, L.; Dai, X.-M.; Chen, W.-Q.; Lin, Q.; et al. The role of PD-1 signaling in health and immune-related diseases. Front. Immunol. 2023, 14, 1163633. [Google Scholar] [CrossRef]
  97. Cristofoletti, C.; Bresin, A.; Picozza, M.; Picchio, M.C.; Monzo, F.; Helmer Citterich, M.; Passarelli, F.; Frezzolini, A.; Scala, E.; Monopoli, A.; et al. Blood and skin-derived Sezary cells: Differences in proliferation-index, activation of PI3K/AKT/mTORC1 pathway and its prognostic relevance. Leukemia 2019, 33, 1231–1242. [Google Scholar] [CrossRef] [PubMed]
  98. Salmena, L.; Carracedo, A.; Pandolfi, P.P. Tenets of PTEN tumor suppression. Cell 2008, 133, 403–414. [Google Scholar] [CrossRef]
  99. Lizcano, J.M.; Göransson, O.; Toth, R.; Deak, M.; Morrice, N.A.; Boudeau, J.; Hawley, S.A.; Udd, L.; Mäkelä, T.P.; Hardie, D.G.; et al. LKB1 is a master kinase that activates 13 kinases of the AMPK subfamily, including MARK/PAR-1. EMBO J. 2004, 23, 833–843. [Google Scholar] [CrossRef]
Figure 1. Glycolysis and its inhibitors. Glucose transporter (GLUT); Hexokinase (HK); Phosphoglucose isomerase (PGI); 6-Phosphofructo-2-Kinase/Fructose-2,6-Biphosphatase3 (PFKFB3); Phosphofructokinase 1 (PFK1); Tripsephosphate isomerase (TP1); Glyceraldehyde phosphate dehydrogenase (GAPDH); Phosphoglycerate Kinase (PGK); Phosphoglycerate mutase (PGAM); Pyruvate Kinase M2 (PKM2); Lactate dehydrogenase A (LDHA); Benitrobenrazide (BNBZ).
Figure 1. Glycolysis and its inhibitors. Glucose transporter (GLUT); Hexokinase (HK); Phosphoglucose isomerase (PGI); 6-Phosphofructo-2-Kinase/Fructose-2,6-Biphosphatase3 (PFKFB3); Phosphofructokinase 1 (PFK1); Tripsephosphate isomerase (TP1); Glyceraldehyde phosphate dehydrogenase (GAPDH); Phosphoglycerate Kinase (PGK); Phosphoglycerate mutase (PGAM); Pyruvate Kinase M2 (PKM2); Lactate dehydrogenase A (LDHA); Benitrobenrazide (BNBZ).
Genes 15 00635 g001
Figure 2. Mitochondrial targets for cancer therapy. Pyruvate dehydrogenase complex (PDC); Pyruvate dehydrogenase kinases (PDKs); Dichloroacetate (DCA); isocitrate dehydrogenase (IDH); Glutaminase (GLS); Glutamate dehydrogenase (GDH); Complexes I, II, III, IV; Coenzyme Q (CoQ); Cytochrome c (Cyt c).
Figure 2. Mitochondrial targets for cancer therapy. Pyruvate dehydrogenase complex (PDC); Pyruvate dehydrogenase kinases (PDKs); Dichloroacetate (DCA); isocitrate dehydrogenase (IDH); Glutaminase (GLS); Glutamate dehydrogenase (GDH); Complexes I, II, III, IV; Coenzyme Q (CoQ); Cytochrome c (Cyt c).
Genes 15 00635 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cherfan, C.; Chebly, A.; Rezvani, H.R.; Beylot-Barry, M.; Chevret, E. Delving into the Metabolism of Sézary Cells: A Brief Review. Genes 2024, 15, 635. https://doi.org/10.3390/genes15050635

AMA Style

Cherfan C, Chebly A, Rezvani HR, Beylot-Barry M, Chevret E. Delving into the Metabolism of Sézary Cells: A Brief Review. Genes. 2024; 15(5):635. https://doi.org/10.3390/genes15050635

Chicago/Turabian Style

Cherfan, Carel, Alain Chebly, Hamid Reza Rezvani, Marie Beylot-Barry, and Edith Chevret. 2024. "Delving into the Metabolism of Sézary Cells: A Brief Review" Genes 15, no. 5: 635. https://doi.org/10.3390/genes15050635

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

Article Metrics

Back to TopTop