Next Article in Journal
Morphofunctional Changes in Brain and Peripheral Blood in Adult and Aged Wistar Rats with AlCl3-Induced Neurodegeneration
Previous Article in Journal
Metformin Protects Rat Skeletal Muscle from Physical Exercise-Induced Injury
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Value of Neutrophil Gelatinase-Associated Lipocalin Receptor as a Novel Partner of CD38 in Chronic Lymphocytic Leukemia: From an Adverse Prognostic Factor to a Potential Pharmacological Target?

1
Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Inserm UMRS1138, Drug Resistance in Hematological Malignancies Team, F-75006 Paris, France
2
Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Service d’Hématologie Biologique, F-75013 Paris, France
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(9), 2335; https://doi.org/10.3390/biomedicines11092335
Submission received: 18 July 2023 / Revised: 14 August 2023 / Accepted: 18 August 2023 / Published: 22 August 2023
(This article belongs to the Section Cancer Biology and Oncology)

Abstract

:
Chronic lymphocytic leukemia (CLL) is characterized by the accumulation of neoplastic B lymphocytes that escape death, and correlates with the expression of negative prognostic markers such as the CD38 antigen. Although certain new drugs approved by the US Food and Drug Administration improve the clinical outcome of CLL patients, drug resistance and disease relapse still occur. Like CD38, neutrophil gelatinase-associated lipocalin receptor (NGAL-R) is frequently overexpressed in CLL cells. Here, we evaluated the concomitant surface expression of NGAL-R and CD38 in leukemic blood cells from 52 CLL patients (37 untreated, 8 in clinical remission, and 7 relapsed). We provide evidence of a positive correlation between NGAL-R and CD38 levels both in the interpatient cohorts (p < 0.0001) and in individual patients, indicating a constitutive association of NGAL-R and CD38 at the cell level. Patients with progressing CLL showed a time-dependent increase in NGAL-R/CD38 levels. In treated CLL patients who achieved clinical remission, NGAL-R/CD38 levels were decreased, and were significantly lower than in the untreated and relapsed groups (p < 0.02). As NGAL-R and CD38 participate in CLL cell survival, envisioning their simultaneous inhibition with bispecific NGAL-R/CD38 antibodies capable of inducing leukemic cell death might provide therapeutic benefit for CLL patients.

1. Introduction

Chronic lymphocytic leukemia (CLL) is defined as a slow-developing disease caused by a peripheral accumulation of CD5+ B lymphocytes in the peripheral blood, bone marrow, and secondary lymphoid organs [1]. The leukemic cells (which are mostly quiescent) mainly accumulate because they are unable to develop a cell death program—even though proliferating pools are found in the bone marrow and lymph nodes [1]. Besides the conventional chemotherapeutic agents, the main drugs currently prescribed are signaling inhibitors targeting B cell receptor (BCR)-associated kinase (i.e., Bruton’ s tyrosine kinase (BTK) inhibitors ibrutinib and acalabrutinib), or the antagonist of the B-cell lymphoma-2 (Bcl-2) anti-apoptotic protein (venetoclax) [2]. In a few cases, the combination of classical agents with CD20 monoclonal antibodies (mAbs) (i.e., rituximab and obituzumab) is also proposed [2]. However, these therapies are often accompanied by adverse effects or favored mutations associated with drug resistance, as well as non-genomic resistance [3,4,5,6,7,8]. Thus, treatment of CLL remains a challenge in the clinic, and novel therapies are needed. In this way, the identification of new molecules that could predict the prognosis and achieve treatment in CLL is of general interest.
CLL cells are characterized by high expression of CD5, CD19 and CD23 antigens [9]. Other surface markers may have prognostic importance in CLL, such as CD38, which plays a dual role as a receptor and ectoenzyme (bearing both cyclase and hydrolase activities) [9]. CD38 interacts with the CD31 antigen constitutively expressed by endothelial cells, allowing leukocyte adhesion and migration [9]. High CD38 expression in CLL is associated with a hyperproliferative phenotype, and is an independent adverse prognostic factor [9,10]. CD38-mediated CLL cell survival occurs through the activation of an AKT/SYK/Mcl-1 signaling [11]. Preclinical studies (ex vivo experiments and CLL xenograft models) have demonstrated the antitumor efficacy of the CD38 mAb daratumumab in monotherapy or combination therapy with ibrutinib [12]. Further, CD38 mAbs have recently emerged as promising treatment options in CLL [2,13]. The combination of daratumumab with ibrutinib in CLL patients—untreated, refractory, or relapsed with a poor prognosis (e.g., 17p deletion and/or TP53 mutation)—is being studied in three active phase Ib/II clinical trials (NCT03447808, NCT03734198, NCT04230304). The initial results of the NCT03734198 study showed no significant objective responses compared with ibrutinib alone [14]. A recent strategy of immunotherapy uses bispecific Abs (Bi-Abs), which have more advantages than monospecific Abs. For instance, a CD38/CD3 Bi-Ab (ISB-1342) that targets tumor cells (via CD38) and immune effector T cells (via CD3) is in phase II for relapsed/refractory multiple myeloma (R/R MM) (NCT033309111); the putative efficacy of ISB-1342 in CLL remains to be tested. Given the widespread expression of CD38 in immune populations (e.g., in plasma cells, T and NK cells, myeloid precursor cells, and dendritic cells) [15], CD38/CD3 Bi-Abs may substantially increase the risk of worse syndromic events. To further enhance therapeutic effects, an alternative approach aims at optimizing Bi-Abs which recognize two tumor antigens on tumor cells co-expressing both antigens [16]. Thus, a CD19/CD47 Bi-Ab (TG-1801) has recently entered into clinical development for CLL treatment (NCT04806035).
Neutrophil gelatinase-associated lipocalin (NGAL) is a secreted protein which is capable of binding low-molecular-weight ligands and of capturing siderophores that bind iron with high affinity [17]. NGAL interacts as ligand with a cell surface receptor NGAL-R (also known as SLC22A17) that is able to bind NGAL alone or is bound to a siderophore and iron [17]. NGAL-R belongs to the solute carrier 22 (SLC22) family of organic ion transporters [17]. Recent work from our group showed that (i) serum levels of NGAL (derived from circulating CLL cells) are significantly elevated in untreated CLL patients [18], and (ii) NGAL-R (absent in normal B, T and myeloid compartments) is frequently overexpressed by CLL cells from untreated patients [18]. Moreover, we presented first evidence that the NGAL-R/NGAL complex enhances the survival of CLL cells by activating an SRC/STAT3/Mcl-1 signaling pathway [18].
So far, there are no data on the association of CD38 and NGAL-R during the progression of CLL and following therapeutic treatment. The goal of the present study was to determine whether blood CLL cells co-express CD38 and NGAL-R, and to this end we assessed the levels of CD38 and NGAL-R in CLL cells at the same time point for each patient, as a function of disease severity and treatment.

2. Materials and Methods

2.1. Patients

Peripheral blood was collected from 52 patients (during the period from 2016 to 2018) diagnosed with CLL. Patients were diagnosed according to the International Workshop on CLL (IWCLL) criteria, and staged according to the Binet system [19]. The main exclusion criteria included neoplastic disease other than CLL, diabetes, hepatic fibrosis, and renal or lung disease. Patients were selected based on the following criteria: lymphocytosis, lymphocyte morphology, clinical features, CD5+/CD19+ immunophenotype, and IGHV mutation status. Progressive CLL was characterized by expansion of the neoplastic clone and extravascular accumulation in lymphoid tissues, the bone marrow, and other organs, giving rise to immune dysfunction, lymphadenopathy, splenomegaly and hematopoietic failure. Clinical remission is when the bone marrow contains fewer than 5% blast cells, the blood cell counts return to within normal limits, and there are no signs or symptoms of the disease. Deletions of 17p13, 11q22, 13q14 and trisomy 12 were detected using fluorescence in situ hybridization (FISH) with the Metasystems XL DLEU/LAMP/12cen and XL ATM/TP53 Multi-Color Probe Kits (MetaSystems, Compiègne, France). The patients’ clinical characteristics are summarized in Table 1.

2.2. CLL Cell Separation and Flow Cytometry

Peripheral blood mononuclear cells were isolated from blood using Ficoll-Hypaque density gradient (1.077 g/mL) centrifugation. Freshly isolated cells were stained and analyzed immediately in the flow cytometry assay, using anti-CD19 (clone 4G7, mIgG1; Santa-Cruz, Heidelberg, Germany), anti-CD5 (clone 205919, mIgG1; R&D Systems, Abingdon, UK), anti-CD38 (clone HB7, mouse IgG1; BD Biosciences, LePont de Claix, France), anti-NGAL-R (clone 16315, rabbit IgG; CliniSciences, Nanterre, France), and appropriate isotype controls (Santa-Cruz). Stained cells were analyzed with a Coulter Epics XL flow (Beckman-Coulter, Les Ullis, France) or a FACSCanto II flow (BD Biosciences, Le Pont de Claix, France) cytometer. Data were analyzed using LYSYS (Beckman-Coulter) or FloJo (version 7.0, BD Biosciences, Le Pont de Claix, France) software.

2.3. Statistics

Mann–Whitney tests, Student’s t-tests and one-way ANOVA were performed using GraphPad Prism software (version 7.0, GraphPad Software, La Jolla, CA, USA). For greater stringency, all tests were two-tailed. Significance levels were defined as * p < 0.05, ** p < 0.01, and **** p < 0.0001.

2.4. Ethics Statement

In line with the ethical tenets of the Declaration of Helsinki, the study protocol was approved by the ethical board on human experimentation at the Pitié-Salpêtrière Hospital on 21 May 2014 (CPPIDF6, Paris, France). The patients provided their written and informed consent to participation in the study.

3. Results

3.1. Prognostic Relevance of NGAL-R and CD38 Co-Expression

We measured surface levels of NGAL-R and CD38 (at the same time point for each patient) in CD19+/CD5+ blood samples from 37 untreated CLL patients (including 10 newly diagnosed patients). A significant correlation was observed between the percentage positivity of CD38 and NGAL-R within the total cohort (r = 0.88, p < 0.0001) (Figure 1a). Analysis of NGAL-R/CD38 co-expression showed no significant differences between the group of newly diagnosed patients and the group of CLL follow-up patients (Figure 1a). In addition to the interpatient relationship, a positive correlation between CD38 and NGAL-R was observed on CLL cells within individual patients, as illustrated in Figure 1b by two representative experiments: patient P31 exhibited low NGAL-R/CD38 co-expression (<10%), while patient P22 showed high NGAL-R/CD38 co-expression (>80%), which strongly suggests the colocalization of the two proteins at the cell level. When a 30% cut-off for NGAL-R/CD38 co-expression was used, no significant difference was observed between (NGAL-R/CD38)low and (NGAL-R/CD38)high groups regarding sex, age, lymphocyte count, Binet stage, and IGHV mutational status. Enhanced CD38 expression is associated with CLL cells bearing trisomy 12 or del(17p) [20,21,22]. Our analysis confirmed and completed these data with significantly elevated NGAL-R/CD38 levels found in trisomy 12 cells compared to CLL cells without or with del(11q) and del(13q) (Figure 1c,d). A similar increase in NGAL-R/CD38 levels was also observed in del(17p) CLL cells from patient P5 (Figure 1c,d). The association between NGAL-R/CD38 co-expression and time-to-progression was evaluated in three untreated patients (P16, P17 and P30) with progressive CLL. A longitudinal analysis of CLL cell samples showed that NGAL-R/CD38 levels had increased over time with disease progression, independently of lymphocyte count (Figure 1e).

3.2. Correlation of NGAL-R/CD38 Co-Expression with Remission Rate and Outcome

As for the untreated patients, we measured a significant positive correlation between the expression of NGAL-R and CD38 in CLL-treated patients, both in the relapsed and clinical remission groups (Figure 2a). Both antigens remained co-expressed at the cell level (Figure 2b); as exemplified in Figure 2b, a high level of NGAL-R+/CD38+ CLL cells (>80%) was observed for relapsed patient P44, while the proportion of NGAL-R+/CD38+ CLL cells was low from patient P52 in clinical remission (<10%). The levels of NGAL-R/CD38 were significantly higher in the majority of tumor cell samples from the relapsed group (CD38: mean 40.57% ± 8.85; NGAL-R: mean 41.0% ± 9.63) than in the remission group (CD38: mean 14.88% ± 2.45; NGAL-R: mean 13% ± 4.21) (2.7 and 3.1 times more, respectively; p = 0.005 and p = 0.019, respectively) (Figure 2c). In contrast, and as expected, the levels of NGAL-R/CD38 in the relapsed group were close to NGAL-R/CD38 levels in the untreated group (CD38: mean 28.86% ± 4.53; mean NGAL-R: 29.97% ± 4.22) (p = 0.299 and p = 0.303, respectively).
The surface levels of NGAL-R/CD38 were quantified in CLL cells from two patients (P5 and P10) before treatment and at clinical remission (Figure 2d). The level of NGAL-R/CD38 co-expression increased with time in P10′s CLL cells before treatment, and was markedly decreased at remission (month 16 corresponding to 10 months after FCR treatment) (Figure 2d); similarly, the elevated NGAL-R/CD38 levels in untreated patient P5′s CLL cells fell at remission (months 12 and 15, corresponding to 1 and 4 months, respectively, after RCb treatment) (Figure 2d). Thus, the diminution of NGAL-R/CD38 expression appears to be associated with a good treatment response. At the time of our study, we had no patients who had undergone treatment and then relapsed.
NGAL-R/CD38 levels for each treated patient were measured at different time points after the start of the treatment (Table 1). Our data indicate that the remission duration did not seem to influence the rate of NGAL-R/CD38 expression in the interpatient group; for instance, patient P51, in remission for 17 months, exhibited lower NGAL-R/CD38 CLL cell levels (0%/5%) than patient P46, in remission for 3 months (30%/25%). Moreover, in the relapsed group, expression of NGAL-R/CD38 in CLL cells (84%/85% at 24 months after treatment) from patient P44 was markedly higher than in leukemic cells from patients P39 (20%/25% at 48 months after treatment) and P43 (26%/22% at 72 months after treatment). This may be explained by the extreme clinical heterogeneity of CLL. The kinetics of expression of NGAL-R/CD38 on CLL cells during the course of chemotherapy and at relapse would have to be analyzed in individual CLL patients.

4. Discussion

The present study establishes the association of NGAL-R with CD38 on the surface of primary CLL cells. The high CD38/NGAL-R co-expression is associated with a poor clinical outcome and represents a potential therapeutic target in CLL.
Reports in the literature highlight common biochemical and functional features for NGAL-R and CD38. First, CD38 and NGAL-R promoters contain CpG islands that can be methylated [23,24]. DNA hypomethylation enhances the expression of CD38 in multiple myeloma [23] and NGAL-R in esophageal squamous cell carcinoma [24]. In CLL, alterations in DNA methylation affect a large number of genes, including CD38 and SLC22 members [25,26], and hypomethylation programming is targeted to regions marked as enhancers and binding sites for various transcription factors, including NF-κB, AP-1 and Runx3 [26]. The CD38 promoter exhibits binding sites for NF-κB and AP-1 which increase the transcription of CD38 via binding to its promoter [27,28]. NGAL-R expression in murine bone marrow cells is upregulated by Runx3 [29]. High Runx3 expression is associated with poor survival in leukemic patients [30]. NGAL-R upregulation depending on Runx3 transcriptional activity in CLL is therefore plausible. Of importance, DNA methylation profiles are modulated during CLL progression, time to first treatment, and response to treatment [31]. These changes in DNA methylation may affect CpG sites of CD38 and NGAL-R genes, including transcription-factor binding sites implicated in the regulation of these proteins. Second, CD38 and NGAL-R mediate—at least in part—convergent prosurvival signalings in CLL. The anti-apoptotic protein Mcl-1 is constitutively expressed in CLL cells, and is involved in the cells’ ability to prevent death [32]. CD38 favors survival in CLL cells by activating an AKT/SYK/Mcl-1 signaling pathway [11], while the binding of NGAL to NGAL-R induces the survival of CLL cells by activating an SRC/STAT3/Mcl-1 signaling [18]. Many signaling pathways related to survival and proliferation are associated with lipid rafts in tumor cells [33]. Since CD38 has a short cytoplasmic tail suggesting it is unable to initiate a signal cascade, its involvement in CD19 and BCR survival signaling has been proposed, as CD38 associates with these two receptors in lipid rafts and enhances BCR signaling in CLL cells [34,35]. Although not yet shown for NGAL-R, several SLC22 receptors associate with lipid rafts [36,37]. As NGAL-R co-exists with CD38 on CLL cells (possibly in lipid rafts), the involvement of NGAL-R in CD38 signaling (and vice versa) might be hypothesized. The neutralizing mAbs daratumumab and GTX85032 targeting CD38 and NGAL-R, respectively, induce ex vivo the death of primary CLL cells [12,18,38]. Therefore, we propose that neutralizing CD38 and NGAL-R Abs may be therapeutically used to interrupt survival-promoting pathways. Moreover, our study endorses a potential therapeutic relevance of CD38/NGAL-R Bi-Abs to target poor-prognosis CLL. Blockade therapies with CD38/NGAL-R Bi-Abs in combination with BTK or Bcl-2 inhibitors already available in clinics might optimize the landscape of CLL therapy.
The consideration of CD38 and NGAL-R as druggable co-targets for CLL immunotherapy is reinforced by the fact that both proteins are involved in tumor metabolism [39,40]. Metabolic changes in tumor cells represent a novel opportunity for therapy approaches [41]. In CLL, CD38, through its enzymatic activity, regulates NAD metabolism which leads to an increase in cytoplasmic Ca2+ concentrations, positively influencing proliferation and signaling mediated via chemokine receptors or integrins [42]. Iron, by modulating metabolism, is implicated in the proliferation and survival mechanisms of various cancer cell types, including CLL bearing a TP53 abnormality [43,44]. By capturing circulating and intracellular free iron [40], NGAL bound to its receptor can regulate iron traffic in tumors [40,45]. For instance, NGAL decreases intracellular iron levels and stimulates the expression of glutathione peroxidase 4 to prevent membrane lipid peroxidation in colon cancer cell lines [46].
To our knowledge, the potential value of CD38/NGAL-R association in the pathogenesis of tumors has not previously been assessed. Our study provides the first evidence that the co-expression of NGAL-R and CD38 is a high-value target in CLL. Beyond CLL, the expression rates of CD38 and NGAL-R are high in other tumors such as acute and chronic myeloid leukemias [47,48,49,50], epithelial carcinomas (esophagus, liver, kidney, endometrial carcinomas) [13,24,51,52,53,54,55] and gliomas [51,56]. Although these studies separately showed elevated expressions of CD38 and NGAL-R, it is very likely that both antigens are co-expressed in these tumors, and therefore suggest that a combination of anti-CD38/NGAL-R Abs may offer new therapeutic options for the management of these cancers. The efficacy of anti-CD38 Abs including daratumumab is already being evaluated in preclinical and clinical trials in these malignancies [13,52,57,58]. Based on this, advances in the area of CD38/NGAL-R Bi-Abs might be of significant consideration for future targeted therapies in these types of cancer, which abnormally express CD38 and NGAL-R.
Finally, the limitations of this study are that a small number of treated patients were studied and relapsed patients were not followed up during the progression of CLL. Moreover, our study was single-centered. Therefore, CD38 and NGAL-R markers should be simultaneously studied on other CLL cohorts in centers which extend diagnostic and therapeutic facilities to CLL patients. However, this pilot study can already form a basis for larger studies on the subject.

5. Conclusions

CD38 and NGAL-R represent co-molecular targets with high value in CLL and give good perspectives for the development of innovative therapeutic strategies using Bi-Abs in CLL. CD38/NGAL-R Bi-Abs might have therapeutic potential in CLL patients in relapse or not responding to current therapies.

Author Contributions

Methodology, experimental work, formal analysis and writing, B.B.; lymphocytosis and mutational status analyses, C.Q.; cytogenetic and karyotype analyses, E.C. and F.N.-K.; CLL sample resources, K.M. and F.N.-K.; review and editing, all authors. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study protocol was conducted according to the Declaration of Helsinki and approved by the Ethics Committee on human experimentation of Pitié-Salpêtrière Hospital (21 May 2014, CPPIDF6, Paris, France).

Informed Consent Statement

In line with the ethical tenets of the Declaration of Helsinki, informed consent was obtained from all the patients diagnosed with CLL and involved in the study.

Acknowledgments

This work was supported by the French National Institute of Health and Medical Research (INSERM). We thank Delphine Garnier for critical reading and suggestions for this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Pleyer, L.; Egle, A.; Hartmann, T.N.; Greil, R. Molecular and cellular mechanisms of CLL: Novel therapeutic approaches. Nat. Rev. Clin. Oncol. 2009, 6, 405–418. [Google Scholar] [CrossRef]
  2. Tannoury, M.; Garnier, D.; Susin, S.A.; Bauvois, B. Current Status of Novel Agents for the Treatment of B Cell Malignancies: What’s Coming Next? Cancers 2022, 14, 6026. [Google Scholar] [CrossRef]
  3. Svanberg, R.; Janum, S.; Patten, P.E.M.; Ramsay, A.G.; Niemann, C.U. Targeting the tumor microenvironment in chronic lymphocytic leukemia. Haematologica 2021, 106, 2312–2324. [Google Scholar] [CrossRef]
  4. Wright, S.C.E.; Vasilevski, N.; Serra, V.; Rodon, J.; Eichhorn, P.J.A. Mechanisms of Resistance to PI3K Inhibitors in Cancer: Adaptive Responses, Drug Tolerance and Cellular Plasticity. Cancers 2021, 13, 1538. [Google Scholar] [CrossRef]
  5. Diepstraten, S.T.; Anderson, M.A.; Czabotar, P.E.; Lessene, G.; Strasser, A.; Kelly, G.L. The manipulation of apoptosis for cancer therapy using BH3-mimetic drugs. Nat. Rev. Cancer 2022, 22, 45–64. [Google Scholar] [CrossRef]
  6. Lew, T.E.; Seymour, J.F. Clinical experiences with venetoclax and other pro-apoptotic agents in lymphoid malignancies: Lessons from monotherapy and chemotherapy combination. J. Hematol. Oncol. 2022, 15, 75. [Google Scholar] [CrossRef]
  7. Ran, F.; Liu, Y.; Wang, C.; Xu, Z.; Zhang, Y.; Liu, Y.; Zhao, G.; Ling, Y. Review of the development of BTK inhibitors in overcoming the clinical limitations of ibrutinib. Eur. J. Med. Chem. 2022, 229, 114009. [Google Scholar] [CrossRef]
  8. Skånland, S.S.; Brown, J.R. PI3K inhibitors in chronic lymphocytic leukemia: Where do we go from here? Haematologica 2023, 108, 9–21. [Google Scholar] [CrossRef]
  9. Dürig, J.; Naschar, M.; Schmücker, U.; Renzing-Köhler, K.; Hölter, T.; Hüttmann, A.; Dührsen, U. CD38 expression is an important prognostic marker in chronic lymphocytic leukaemia. Leukemia 2002, 16, 30–35. [Google Scholar] [CrossRef]
  10. Burgler, S. Role of CD38 Expression in Diagnosis and Pathogenesis of Chronic Lymphocytic Leukemia and Its Potential as Therapeutic Target. Crit. Rev. Immunol. 2015, 35, 417–432. [Google Scholar] [CrossRef]
  11. Benkisser-Petersen, M.; Buchner, M.; Dörffel, A.; Dühren-von-Minden, M.; Claus, R.; Kläsener, K.; Leberecht, K.; Burger, M.; Dierks, C.; Jumaa, H.; et al. Spleen Tyrosine Kinase Is Involved in the CD38 Signal Transduction Pathway in Chronic Lymphocytic Leukemia. PLoS ONE 2016, 11, e0169159. [Google Scholar] [CrossRef] [PubMed]
  12. Manna, A.; Aulakh, S.; Jani, P.; Ahmed, S.; Akhtar, S.; Coignet, M.; Heckman, M.; Meghji, Z.; Bhatia, K.; Sharma, A.; et al. Targeting CD38 Enhances the Antileukemic Activity of Ibrutinib in Chronic Lymphocytic Leukemia. Clin. Cancer Res. 2019, 25, 3974–3985. [Google Scholar] [CrossRef] [PubMed]
  13. Szlasa, W.; Czarny, J.; Sauer, N.; Rakoczy, K.; Szymańska, N.; Stecko, J.; Kołodziej, M.; Kaźmierczak, M.; Barg, E. Targeting CD38 in Neoplasms and Non-Cancer Diseases. Cancers 2022, 14, 4169. [Google Scholar] [CrossRef] [PubMed]
  14. Aurran-Schleinitz, T.; Tomowiak, C.; Roos-Weil, D.; Ferrant, E.; Mahé, B.; Molina, L.; Ysebaert, L.; Fornecker, L.M.; Michallet, A.S.; Levy, V.; et al. Combined Treatment with Ibrutinib and Anti-CD38 Monoclonal Antibody Daratumumab in Relapsed/Refractory Chronic Lymphocytic Leukemia with TP53 Aberrations: Results of the Filo Phase II Study IDA53. Blood 2022, 140, 7030–7031. [Google Scholar] [CrossRef]
  15. Morandi, F.; Airoldi, I.; Marimpietri, D.; Bracci, C.; Faini, A.C.; Gramignoli, R. CD38, a Receptor with Multifunctional Activities: From Modulatory Functions on Regulatory Cell Subsets and Extracellular Vesicles, to a Target for Therapeutic Strategies. Cells 2019, 8, 1527. [Google Scholar] [CrossRef] [PubMed]
  16. Labrijn, A.F.; Janmaat, M.L.; Reichert, J.M.; Parren, P. Bispecific antibodies: A mechanistic review of the pipeline. Nat. Rev. Drug Discov. 2019, 18, 585–608. [Google Scholar] [CrossRef] [PubMed]
  17. Cabedo Martinez, A.I.; Weinhaupl, K.; Lee, W.K.; Wolff, N.A.; Storch, B.; Zerko, S.; Konrat, R.; Kozminski, W.; Breuker, K.; Thevenod, F.; et al. Biochemical and Structural Characterization of the Interaction between the Siderocalin NGAL/LCN2 (Neutrophil Gelatinase-associated Lipocalin/Lipocalin 2) and the N-terminal Domain of Its Endocytic Receptor SLC22A17. J. Biol. Chem. 2016, 291, 2917–2930. [Google Scholar] [CrossRef]
  18. Bauvois, B.; Pramil, E.; Jondreville, L.; Chapiro, E.; Quiney, C.; Maloum, K.; Susin, S.A.; Nguyen-Khac, F. Relation of Neutrophil Gelatinase-Associated Lipocalin Overexpression to the Resistance to Apoptosis of Tumor B Cells in Chronic Lymphocytic Leukemia. Cancers 2020, 12, 2124. [Google Scholar] [CrossRef]
  19. Hallek, M.; Cheson, B.D.; Catovsky, D.; Caligaris-Cappio, F.; Dighiero, G.; Dohner, H.; Hillmen, P.; Keating, M.; Montserrat, E.; Chiorazzi, N.; et al. iwCLL guidelines for diagnosis, indications for treatment, response assessment, and supportive management of CLL. Blood 2018, 131, 2745–2760. [Google Scholar] [CrossRef]
  20. El-Kinawy, N.S.; Sharaf, H.M.; Abd El-Hammid, M. Prognostic significance of del 17p, ZAP70 and CD38 as independent indicators for B-CLL: Correlation to response to treatment and disease outcome. Egypt. J. Med. Hum. Genet. 2012, 13, 173–181. [Google Scholar] [CrossRef]
  21. Chevallier, P.; Penther, D.; Avet-Loiseau, H.; Robillard, N.; Ifrah, N.; Mahé, B.; Hamidou, M.; Maisonneuve, H.; Moreau, P.; Jardel, H.; et al. CD38 expression and secondary 17p deletion are important prognostic factors in chronic lymphocytic leukaemia. Br. J. Haematol. 2002, 116, 142–150. [Google Scholar] [CrossRef] [PubMed]
  22. Riches, J.C.; O’Donovan, C.J.; Kingdon, S.J.; McClanahan, F.; Clear, A.J.; Neuberg, D.S.; Werner, L.; Croce, C.M.; Ramsay, A.G.; Rassenti, L.Z.; et al. Trisomy 12 chronic lymphocytic leukemia cells exhibit upregulation of integrin signaling that is modulated by NOTCH1 mutations. Blood 2014, 123, 4101–4110. [Google Scholar] [CrossRef] [PubMed]
  23. Choudhry, P.; Mariano, M.C.; Geng, H.; Martin, T.G., 3rd; Wolf, J.L.; Wong, S.W.; Shah, N.; Wiita, A.P. DNA methyltransferase inhibitors upregulate CD38 protein expression and enhance daratumumab efficacy in multiple myeloma. Leukemia 2020, 34, 938–941. [Google Scholar] [CrossRef] [PubMed]
  24. Cui, L.; Xu, L.Y.; Shen, Z.Y.; Tao, Q.; Gao, S.Y.; Lv, Z.; Du, Z.P.; Fang, W.K.; Li, E.M. NGALR is overexpressed and regulated by hypomethylation in esophageal squamous cell carcinoma. Clin. Cancer Res. 2008, 14, 7674–7681. [Google Scholar] [CrossRef]
  25. Rahmatpanah, F.B.; Carstens, S.; Hooshmand, S.I.; Welsh, E.C.; Sjahputera, O.; Taylor, K.H.; Bennett, L.B.; Shi, H.; Davis, J.W.; Arthur, G.L.; et al. Large-scale analysis of DNA methylation in chronic lymphocytic leukemia. Epigenomics 2009, 1, 39–61. [Google Scholar] [CrossRef]
  26. Oakes, C.C.; Seifert, M.; Assenov, Y.; Gu, L.; Przekopowitz, M.; Ruppert, A.S.; Wang, Q.; Imbusch, C.D.; Serva, A.; Koser, S.D.; et al. DNA methylation dynamics during B cell maturation underlie a continuum of disease phenotypes in chronic lymphocytic leukemia. Nat. Genet. 2016, 48, 253–264. [Google Scholar] [CrossRef]
  27. Deshpande, D.A.; Guedes, A.G.P.; Graeff, R.; Dogan, S.; Subramanian, S.; Walseth, T.F.; Kannan, M.S. CD38/cADPR Signaling Pathway in Airway Disease: Regulatory Mechanisms. Mediat. Inflamm. 2018, 2018, 8942042. [Google Scholar] [CrossRef]
  28. Zuo, W.; Liu, N.; Zeng, Y.; Liu, Y.; Li, B.; Wu, K.; Xiao, Y.; Liu, Q. CD38: A Potential Therapeutic Target in Cardiovascular Disease. Cardiovasc. Drugs Ther. 2021, 35, 815–828. [Google Scholar] [CrossRef]
  29. Sheng, Z.; Wang, S.Z.; Green, M.R. Transcription and signalling pathways involved in BCR-ABL-mediated misregulation of 24p3 and 24p3R. Embo J. 2009, 28, 866–876. [Google Scholar] [CrossRef]
  30. Sun, C.C.; Li, S.J.; Chen, Z.L.; Li, G.; Zhang, Q.; Li, D.J. Expression and Prognosis Analyses of Runt-Related Transcription Factor Family in Human Leukemia. Mol. Ther. Oncolytics 2019, 12, 103–111. [Google Scholar] [CrossRef]
  31. Tsagiopoulou, M.; Papakonstantinou, N.; Moysiadis, T.; Mansouri, L.; Ljungström, V.; Duran-Ferrer, M.; Malousi, A.; Queirós, A.C.; Plevova, K.; Bhoi, S.; et al. DNA methylation profiles in chronic lymphocytic leukemia patients treated with chemoimmunotherapy. Clin. Epigenetics 2019, 11, 177. [Google Scholar] [CrossRef] [PubMed]
  32. Packham, G.; Stevenson, F.K. Bodyguards and assassins: Bcl-2 family proteins and apoptosis control in chronic lymphocytic leukaemia. Immunology 2005, 114, 441–449. [Google Scholar] [CrossRef] [PubMed]
  33. Li, B.; Qin, Y.; Yu, X.; Xu, X.; Yu, W. Lipid raft involvement in signal transduction in cancer cell survival, cell death and metastasis. Cell Prolif. 2022, 55, e13167. [Google Scholar] [CrossRef]
  34. Deaglio, S.; Capobianco, A.; Bergui, L.; Dürig, J.; Morabito, F.; Dührsen, U.; Malavasi, F. CD38 is a signaling molecule in B-cell chronic lymphocytic leukemia cells. Blood 2003, 102, 2146–2155. [Google Scholar] [CrossRef] [PubMed]
  35. Camponeschi, A.; Kläsener, K.; Sundell, T.; Lundqvist, C.; Manna, P.T.; Ayoubzadeh, N.; Sundqvist, M.; Thorarinsdottir, K.; Gatto, M.; Visentini, M.; et al. Human CD38 regulates B cell antigen receptor dynamic organization in normal and malignant B cells. J. Exp. Med. 2022, 219, e20220201. [Google Scholar] [CrossRef]
  36. Srimaroeng, C.; Cecile, J.P.; Walden, R.; Pritchard, J.B. Regulation of renal organic anion transporter 3 (SLC22A8) expression and function by the integrity of lipid raft domains and their associated cytoskeleton. Cell Physiol. Biochem. 2013, 31, 565–578. [Google Scholar] [CrossRef]
  37. Engelhart, D.C.; Granados, J.C.; Shi, D.; Saier, M.H., Jr.; Baker, M.E.; Abagyan, R.; Nigam, S.K. Systems Biology Analysis Reveals Eight SLC22 Transporter Subgroups, Including OATs, OCTs, and OCTNs. Int. J. Mol. Sci. 2020, 21, 1791. [Google Scholar] [CrossRef]
  38. Matas-Céspedes, A.; Vidal-Crespo, A.; Rodriguez, V.; Villamor, N.; Delgado, J.; Giné, E.; Roca-Ho, H.; Menéndez, P.; Campo, E.; López-Guillermo, A.; et al. The Human CD38 Monoclonal Antibody Daratumumab Shows Antitumor Activity and Hampers Leukemia-Microenvironment Interactions in Chronic Lymphocytic Leukemia. Clin. Cancer Res. 2017, 23, 1493–1505. [Google Scholar] [CrossRef]
  39. Gao, L.; Du, X.; Li, J.; Qin, F.X. Evolving roles of CD38 metabolism in solid tumour microenvironment. Br. J. Cancer 2023, 128, 492–504. [Google Scholar] [CrossRef]
  40. Chakraborty, S.; Kaur, S.; Guha, S.; Batra, S.K. The multifaceted roles of neutrophil gelatinase associated lipocalin (NGAL) in inflammation and cancer. Biochim. Biophys. Acta 2012, 1826, 129–169. [Google Scholar] [CrossRef]
  41. 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, 113. [Google Scholar] [CrossRef] [PubMed]
  42. Vaisitti, T.; Audrito, V.; Serra, S.; Buonincontri, R.; Sociali, G.; Mannino, E.; Pagnani, A.; Zucchetto, A.; Tissino, E.; Vitale, C.; et al. The enzymatic activities of CD38 enhance CLL growth and trafficking: Implications for therapeutic targeting. Leukemia 2015, 29, 356–368. [Google Scholar] [CrossRef] [PubMed]
  43. Hsu, M.Y.; Mina, E.; Roetto, A.; Porporato, P.E. Iron: An Essential Element of Cancer Metabolism. Cells 2020, 9, 2591. [Google Scholar] [CrossRef] [PubMed]
  44. Laubach, K.; Zhang, J.; Chen, X. The p53 Family: A Role in Lipid and Iron Metabolism. Front. Cell Dev. Biol. 2021, 9, 715974. [Google Scholar] [CrossRef] [PubMed]
  45. Bauvois, B.; Susin, S.A. Revisiting Neutrophil Gelatinase-Associated Lipocalin (NGAL) in Cancer: Saint or Sinner? Cancers 2018, 10, 336. [Google Scholar] [CrossRef]
  46. Chaudhary, N.; Choudhary, B.S.; Shah, S.G.; Khapare, N.; Dwivedi, N.; Gaikwad, A.; Joshi, N.; Raichanna, J.; Basu, S.; Gurjar, M.; et al. Lipocalin 2 expression promotes tumor progression and therapy resistance by inhibiting ferroptosis in colorectal cancer. Int. J. Cancer 2021, 149, 1495–1511. [Google Scholar] [CrossRef]
  47. Herrmann, H.; Sadovnik, I.; Eisenwort, G.; Rülicke, T.; Blatt, K.; Herndlhofer, S.; Willmann, M.; Stefanzl, G.; Baumgartner, S.; Greiner, G.; et al. Delineation of target expression profiles in CD34+/CD38- and CD34+/CD38+ stem and progenitor cells in AML and CML. Blood Adv. 2020, 4, 5118–5132. [Google Scholar] [CrossRef]
  48. Naik, J.; Themeli, M.; de Jong-Korlaar, R.; Ruiter, R.W.J.; Poddighe, P.J.; Yuan, H.; de Bruijn, J.D.; Ossenkoppele, G.J.; Zweegman, S.; Smit, L.; et al. CD38 as a therapeutic target for adult acute myeloid leukemia and T-cell acute lymphoblastic leukemia. Haematologica 2019, 104, e100–e103. [Google Scholar] [CrossRef]
  49. Nadarajan, V.S.; Ang, C.H.; Bee, P.C. Lipocalin-2 is associated with modulation of disease phenotype in a patient with concurrent JAK2-V617F and BCR-ABL mutation. Eur. J. Haematol. 2012, 88, 175–178. [Google Scholar] [CrossRef]
  50. Bauvois, B. Overexpression of Neutrophil Gelatinase-Associated Lipocalin Receptor in Human Acute Myeloid Leukemias. INSERM UMRS1138, Paris, France. 2023; Unpublished Work. [Google Scholar]
  51. Konen, J.M.; Fradette, J.J.; Gibbons, D.L. The Good, the Bad and the Unknown of CD38 in the Metabolic Microenvironment and Immune Cell Functionality of Solid Tumors. Cells 2019, 9, 52. [Google Scholar] [CrossRef]
  52. Dwivedi, S.; Rendón-Huerta, E.P.; Ortiz-Navarrete, V.; Montaño, L.F. CD38 and Regulation of the Immune Response Cells in Cancer. J. Oncol. 2021, 2021, 6630295. [Google Scholar] [CrossRef]
  53. Zhang, Y.; Fan, Y.; Mei, Z. NGAL and NGALR overexpression in human hepatocellular carcinoma toward a molecular prognostic classification. Cancer Epidemiol. 2012, 36, e294–e299. [Google Scholar] [CrossRef] [PubMed]
  54. Liu, F.; Li, N.; Yang, W.; Wang, R.; Yu, J.; Wang, X. The expression analysis of NGAL and NGALR in clear cell renal cell carcinoma. Gene 2018, 676, 269–278. [Google Scholar] [CrossRef] [PubMed]
  55. Miyamoto, T.; Asaka, R.; Suzuki, A.; Takatsu, A.; Kashima, H.; Shiozawa, T. Immunohistochemical detection of a specific receptor for lipocalin2 (solute carrier family 22 member 17, SLC22A17) and its prognostic significance in endometrial carcinoma. Exp. Mol. Pathol. 2011, 91, 563–568. [Google Scholar] [CrossRef] [PubMed]
  56. Liu, M.F.; Jin, T.; Shen, J.H.; Shen, Z.Y.; Zheng, Z.C.; Zhang, Z.L.; Xu, L.Y.; Li, E.M.; Xu, H.X. NGAL and NGALR are frequently overexpressed in human gliomas and are associated with clinical prognosis. J. Neurooncol. 2011, 104, 119–127. [Google Scholar] [CrossRef]
  57. Cui, Q.; Liang, P.; Dai, H.; Cui, W.; Cai, M.; Ding, Z.; Ma, Q.; Yin, J.; Li, Z.; Liu, S.; et al. Case report: CD38-directed CAR-T cell therapy: A novel immunotherapy targeting CD38- positive blasts overcomes TKI and chemotherapy resistance of chronic myeloid leukemia in blastic phase. Front. Immunol. 2022, 13, 1012981. [Google Scholar] [CrossRef]
  58. Zhong, X.; Ma, H. Targeting CD38 for acute leukemia. Front. Oncol. 2022, 12, 1007783. [Google Scholar] [CrossRef]
Figure 1. Correlation of surface expression of CD38 and NGAL-R in CLL cells from untreated patients. (a) The plot compares the percentage surface CD38 and NGAL-R co-expression on CLL cells derived from 37 untreated CLL patients (10 newly diagnosed and 27 in follow-up). Spearman’s correlation coefficient (r) and the p value are shown. (b) Representative cytograms of CLL cells from 2 untreated patients (P31, P22), stained with rabbit IgG-FITC/mIgG1-PE, and NGAL-R-FITC/CD38-PE Abs. (c) CD38 and (d) NGAL-R surface levels were determined in CLL cells from patients without (normal, 4 patients) or with del(11q) (4 patients), del(13q) (27 patients), trisomy 12 (4 patients) and del(17p) (1 patient). The data are presented as mean ± SEM; ANOVA values are shown. * p < 0.05; not significant (ns). (e) Levels of NGAL-R and CD38, and lymphocyte count (G/L) in 3 untreated, stage A CLL patients (P16, P17, P30) over months after first analysis. Patient P16 progressed to stage B at month 16.
Figure 1. Correlation of surface expression of CD38 and NGAL-R in CLL cells from untreated patients. (a) The plot compares the percentage surface CD38 and NGAL-R co-expression on CLL cells derived from 37 untreated CLL patients (10 newly diagnosed and 27 in follow-up). Spearman’s correlation coefficient (r) and the p value are shown. (b) Representative cytograms of CLL cells from 2 untreated patients (P31, P22), stained with rabbit IgG-FITC/mIgG1-PE, and NGAL-R-FITC/CD38-PE Abs. (c) CD38 and (d) NGAL-R surface levels were determined in CLL cells from patients without (normal, 4 patients) or with del(11q) (4 patients), del(13q) (27 patients), trisomy 12 (4 patients) and del(17p) (1 patient). The data are presented as mean ± SEM; ANOVA values are shown. * p < 0.05; not significant (ns). (e) Levels of NGAL-R and CD38, and lymphocyte count (G/L) in 3 untreated, stage A CLL patients (P16, P17, P30) over months after first analysis. Patient P16 progressed to stage B at month 16.
Biomedicines 11 02335 g001
Figure 2. Co-expression profiles of surface NGAL-R and CD38 on CLL cells from patients after therapeutic treatment. (a) The plot compares the percentage surface CD38 and NGAL-R co-expression on CLL cells derived from 15 treated CLL patients (8 in remission and 7 relapsed). Spearman’s correlation coefficient (r) and the p value are shown. (b) Representative cytograms of CLL cells from 2 treated patients (P44/relapse, P52/remission), stained with rabbit IgG-FITC/mIgG1-PE, and NGAL-R-FITC/CD38-PE Abs. (c) NGAL-R/CD38 levels were determined in CLL cells from patients at clinical remission (n = 8) or relapse (n = 7). The data are shown as mean ± SEM, and p values are calculated using a Mann–Whitney U-test. * p < 0.05; ** p < 0.01. (d) Levels of NGAL-R/CD38 in CLL cells from patient P10 (stage B) at months 0 and 2, treated with FCR at month 6, and at remission (month 16 (patient P49)); levels of NGAL-R/CD38 in CLL cells from patient P5 (stage C) at month 0, treated with RCb at month 11, and at clinical remission (month 12 (patient P45) and month 15). The arrow indicates the beginning of the treatment T. Lymphocyte count (G/L) over months is shown.
Figure 2. Co-expression profiles of surface NGAL-R and CD38 on CLL cells from patients after therapeutic treatment. (a) The plot compares the percentage surface CD38 and NGAL-R co-expression on CLL cells derived from 15 treated CLL patients (8 in remission and 7 relapsed). Spearman’s correlation coefficient (r) and the p value are shown. (b) Representative cytograms of CLL cells from 2 treated patients (P44/relapse, P52/remission), stained with rabbit IgG-FITC/mIgG1-PE, and NGAL-R-FITC/CD38-PE Abs. (c) NGAL-R/CD38 levels were determined in CLL cells from patients at clinical remission (n = 8) or relapse (n = 7). The data are shown as mean ± SEM, and p values are calculated using a Mann–Whitney U-test. * p < 0.05; ** p < 0.01. (d) Levels of NGAL-R/CD38 in CLL cells from patient P10 (stage B) at months 0 and 2, treated with FCR at month 6, and at remission (month 16 (patient P49)); levels of NGAL-R/CD38 in CLL cells from patient P5 (stage C) at month 0, treated with RCb at month 11, and at clinical remission (month 12 (patient P45) and month 15). The arrow indicates the beginning of the treatment T. Lymphocyte count (G/L) over months is shown.
Biomedicines 11 02335 g002
Table 1. Clinical characteristics of chronic lymphocytic leukemia (CLL) patients.
Table 1. Clinical characteristics of chronic lymphocytic leukemia (CLL) patients.
CLLSex/
Age
Stage at DiagnosisLymphocyte
Count G/L at Analysis Time
IGHV FISHKaryotype
at Diagnosis
Therapy (Analysis Time Point after the Start of Treatment)
1M/71A12.4MTri 121 abnormalityNone
2M/63A5.51nd13q-ComplexNone
3M/90A16.02MndndNone
4M/76A48.91nd13q-NormalNone
5M/87C21.89UM17p-, 13q-NormalNone
6M/66B37.22M13q-2 abnormalitiesNone
7F/74A78UM13q-NormalNone
8F/78A26.84M13q-NormalNone
9F/68A16.15nd13q-1 abnormalityNone
10F/62B179.86UM13q-NormalNone
11F/83A64ndNormalNormalNone
12M/82A4.69nd11q-, 13q-NormalNone
13F/77A34.93UM13q-FailureNone
14M/74A30.08nd13q-2 abnormalitiesNone
15F/64A5.77M13q-NormalNone
16M/70A9.04nd13q-NormalNone
17M/75A13.36M13q-NormalNone
18F/67A20.08nd13q-NormalNone
19F/70A9nd13q-NormalNone
20F/79A10.12nd13q-NormalNone
21F/54B41.02ndTri 121 abnormalityNone
22M/76A5.99M13q-NormalNone
23M/68B70.97nd11q-ComplexNone
24F/84A20.44ndNormalNormalNone
25M/75B184.01UMTri 121 abnormalityNone
26F/82A8.8nd13q-NormalNone
27F/85A71,98ndNormal2 abnormalitiesNone
28F/74A3.44nd13q-1 abnormalityNone
29M/76A14.46nd13q-NormalNone
30F/85A19.51M13q-NormalNone
31F/52A11.9nd13q-NormalNone
32F/68B70.14nd11q-, 13q- ComplexNone
33F/57A10.49nd13q-NormalNone
34F/80B35.01nd11q-1 abnormalityNone
35M/62A11.42ndNormalNormalNone
36M/61B78nd13q-, Tri 12ComplexNone
37F/53A15.58UM13q-2 abnormalitiesNone
38F/80B30.97nd11q-1 abnormalityBR/relapse
(month 12)
39F/78A10.38M13q-1 abnormalityRCb/relapse
(month 48)
40M/76B68.25nd17p-, 13q-2 abnormalitiesIbrutinib/relapse
(month 12)
41M/80B11.06UM17p-, 13q-ComplexIbrutinib/relapse
(month 24)
42M/74B7.5ndndndFCR/relapse
(month 96)
43M/81C7.02M11q-, 13q-ndAlemtuzumab/relapse
(month 72)
44M/82C6.42UM11q-, 13q-ComplexBR/relapse
(month 24)
45M/88C0.87UM17p-, 13q-NormalRCb/remission
(month 8)
46M/79B1.3UMNormalNormalBR/remission
(month 8)
47M/64A1.15UMTri 121 abnormalityFCR/remission
(month 54)
48F/75B1.39M13q-, Tri 121 abnormalityFCR/remission
(month 36)
49F/64B0.26UM13q-NormalFCR/remission
(month 10)
50M/48C43.64UM17p-ComplexIbrutinib/remission
(month 2)
51F/72B20.2UM17p-, Tri 12ComplexIbrutinib/remission
(month17)
52M/66C7.64UM13q-, Tri 12ComplexIbrutinib/remission
(month 2)
Patients were diagnosed according to the IWCLL criteria and staged according to the Binet system [19]. Newly diagnosed patients P28-P37. The patient cohort listed in this table is based in part on the cohort previously published by our group [18]. Fluorescent in situ hybridization (FISH) detecting 13q14 deletion (13q-), 11q22 deletion (11q-), 17p13 deletion (17p-) and trisomy 12 (Tri 12); chromosomal abnormalities are defined by conventional karyotyping [18,19]. IGHV mutational status data were not available for 26 patients, FISH not available for 2 patients, karyotype not available for 3 patients; not done (nd). Bendamustine and rituximab (BR). Rituximab and chlorambucil (RCb). Fludarabine, cyclophosphamide and rituximab (FCR). Alemtuzumab is a CD52 mAb. G/L, giga per liter; unmutated (UM); mutated (M).
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

Bauvois, B.; Chapiro, E.; Quiney, C.; Maloum, K.; Susin, S.A.; Nguyen-Khac, F. The Value of Neutrophil Gelatinase-Associated Lipocalin Receptor as a Novel Partner of CD38 in Chronic Lymphocytic Leukemia: From an Adverse Prognostic Factor to a Potential Pharmacological Target? Biomedicines 2023, 11, 2335. https://doi.org/10.3390/biomedicines11092335

AMA Style

Bauvois B, Chapiro E, Quiney C, Maloum K, Susin SA, Nguyen-Khac F. The Value of Neutrophil Gelatinase-Associated Lipocalin Receptor as a Novel Partner of CD38 in Chronic Lymphocytic Leukemia: From an Adverse Prognostic Factor to a Potential Pharmacological Target? Biomedicines. 2023; 11(9):2335. https://doi.org/10.3390/biomedicines11092335

Chicago/Turabian Style

Bauvois, Brigitte, Elise Chapiro, Claire Quiney, Karim Maloum, Santos A. Susin, and Florence Nguyen-Khac. 2023. "The Value of Neutrophil Gelatinase-Associated Lipocalin Receptor as a Novel Partner of CD38 in Chronic Lymphocytic Leukemia: From an Adverse Prognostic Factor to a Potential Pharmacological Target?" Biomedicines 11, no. 9: 2335. https://doi.org/10.3390/biomedicines11092335

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