• In vivo imaging reveals that CLL cells bind to lymph node high endothelial venules via an L-selectin–dependent multistep adhesion cascade.

  • Interference with L-selectin–mediated trafficking in high endothelial venules could limit dissemination of CLL cells to lymph nodes.

B-cell chronic lymphocytic leukemia (CLL) is the most common leukemia in adults. Lymph nodes (LNs) are sites of malignant proliferation and LN enlargement is associated with poor prognosis in the clinics. The LN microenvironment is believed to favor disease progression by promoting CLL cell growth and drug resistance. A better understanding of the mechanisms regulating trafficking of CLL cells to LNs is thus urgently needed. Here, we studied the first step of CLL cell migration to LNs, their interaction with high endothelial venules (HEVs), specialized blood vessels for lymphocyte extravasation in lymphoid organs. We observed that the density of HEV blood vessels was increased in CLL LNs and that CD20+ CLL cells accumulated within HEV pockets, suggesting intense trafficking. We used intravital imaging to visualize the behavior of human CLL cells within the mouse LN microcirculation, and discovered that CLL cells bind to HEVs in vivo via a multistep adhesion cascade, which involves rolling, sticking, and crawling of the leukemic cells on the endothelium. Functional analyses revealed that the lymphocyte homing receptor L-selectin (CD62L) is the key factor controlling the binding of CLL cells to HEV walls in vivo. Interestingly, L-selectin expression was decreased on CLL cells from patients treated with idelalisib, a phosphoinositide-3-kinase δ inhibitor recently approved for CLL therapy. Interference with L-selectin–mediated trafficking in HEVs could represent a novel strategy to block dissemination of CLL cells to LNs and increase the efficacy of conventional therapy.

B-cell chronic lymphocytic leukemia (CLL) is the most common leukemia in adults with highly variable clinical progression, characterized by accumulation of malignant B cells in blood, bone marrow, lymph nodes (LNs), spleen, and liver.1,2  Accessory cells such as T cells, mesenchymal stromal cells, and nurse-like cells constitute a protective microenvironment for CLL cells in lymphoid organs.3-5  They prevent the apoptosis of CLL cells and support their clonal proliferation within specific structures known as proliferation centers. The tissue microenvironment is thus believed to play a critical role in CLL disease progression.3-5  Indeed, a bulky lymphadenopathy is a feature of advanced disease correlating with poor clinical outcome. Importantly, CLL cell migration to LNs provides protection from chemotherapy and immunotherapy.6  Interfering with CLL cell migration or retention in LNs could thus result in a greater efficacy of conventional therapy for advanced CLL patients.

Several chemokine receptors have been proposed to play important roles in CLL cell trafficking (ie, CXC chemokine receptors [CXCRs] CXCR4 and CXCR5, and CC chemokine receptor 7 [CCR7]).5,7  CXCR4, the receptor for CXC chemokine ligand 12 (CXCL12), was the first chemokine receptor shown to be expressed and functional on CLL cells.8  CLL cells were later found to express high levels of CCR79,10  and CXCR5,10-12  the receptors for chemokines CC chemokine ligands (CCLs) CCL19/CCL21 and CXCL13, respectively. In vitro chemotaxis assays have revealed that CLL cells efficiently migrate in response to all ligands of CCR7, CXCR4, and CXCR5.8-12  Together, these studies suggested that lymphoid chemokines and their receptors, which play critical roles in B-cell homing to LNs and other secondary lymphoid organs, are likely to be also important for CLL cell migration in vivo.8-12 

Despite these important advances on the role of chemokine receptors in CLL cell trafficking, the mechanisms regulating CLL cell migration in vivo remain incompletely characterized. For instance, it is currently unknown which molecules control the initial capture and trafficking of CLL cells in high endothelial venules (HEVs), specialized blood vessels for lymphocyte entry in LNs.13,14  B lymphocytes use a multistep adhesion cascade (rolling, sticking, crawling) to attach to HEV walls in vivo.13  Whether CLL cells exhibit a similar behavior has not yet been determined. Here, we used an in vivo imaging approach to address these unresolved and important issues. Intravital microscopy analysis of the mouse LN microcirculation14,15  allowed us to visualize for the first time the rolling, sticking, and crawling of human CLL cells on HEV endothelium in vivo, and to demonstrate the critical role of the lymphocyte homing receptor L-selectin (CD62L)16-18  in these processes. Interestingly, we observed downregulation of L-selectin on CLL cells from patients under treatment with the phosphoinositide 3-kinase δ inhibitor idelalisib,19,20  suggesting an inhibitory effect of this drug on CLL cell migration to LNs.

Study approval

Patient’s tissues and blood were obtained following standard ethical procedures (Helsinki principles), after informed written consent, and stored at the HIMIP collection. According to the French law, the HIMIP collection was declared to the Ministry of Higher Education and Research (DC 2008-307 collection 1); a transfer agreement (AC 2008-129) was obtained after approbation by the “Comité de Protection des Personnes Sud-Ouest et Outremer II” (ethical committee). Clinical and biological annotations of the samples have been declared to the Commission Nationale de l’Informatique et des Libertés (CNIL). Animal experiments were conducted according to institutional guidelines for animal handling and using protocols approved by the Institut de Pharmacologie et de Biologie Structurale and Région Midi-Pyrénées animal care committees.

Patient’s cells

Peripheral blood samples from 30 patients with CLL/small lymphocytic lymphoma (SLL) who underwent routine follow-up visits at Service d’Hématologie (Institut Universitaire du Cancer Toulouse [IUCT]–Oncopôle, Toulouse, France) were collected. Peripheral blood mononuclear cells (PBMCs) were isolated by density gradient centrifugation over Ficoll-Plaque (GE Healthcare) and used immediately. Purity of CD19+CD5+ B cells was assessed to be >90% by flow cytometry. CLL/SLL patient characteristics are described in Table 1. Patients were untreated at the time of blood collection, with the exception of patients treated with idelalisib (Table 1). Bulky disease was determined by a clinical examination and defined as presence of nodal mass ≥5 cm and/or spleen size ≥6 cm below left costal margin at physical examination. Peripheral blood samples from 6 second- to third-line patients (Table 1) who received idelalisib 150 mg twice a day (national patient drug access program) in combination with anti-CD20 immunotherapy were collected before treatment and at the indicated time under treatment.

Table 1

Patient characteristics

Patient no.CLL/ SLLBulkyBinet stageCirculating count, cells/mm3TreatmentsActive disease IWCLL 2008 criteriaIgVHMutations
Blood samples from untreated patients         
 BL01 SLL Yes 3 300 Untreated No Unmutated tri12 
 BL02 CLL Yes 30 000 Untreated No Unmutated del(11q), del(13q), del(14q), complex karyotype 
 BL03 CLL Yes 22 000 Untreated No Unmutated del(11q) 
 BL04 CLL No 120 000 Untreated No Mutated t(14;18) 
 BL05 CLL No 50 000 Untreated No Mutated Normal karyotype 
 BL06 CLL No 45 000 Untreated No Unmutated tri12, del(6q), simple karyotype 
 BL07 CLL No 106 000 Untreated Yes Unmutated del(13q) 
 BL08 CLL No 130 000 Untreated No Unmutated Normal karyotype 
 BL09 CLL No 21 000 Untreated No Mutated del(13q), tri12, tri19 
 BL10 CLL No 77 000 Untreated No Mutated Normal karyotype 
 BL11 CLL No 79 860 Untreated Yes Unmutated t(3;8), del6q 
 BL12 CLL No 65 710 Untreated No Mutated del13q, del11q 
 BL13 CLL No 111 710 Untreated No NA NA 
 BL14 CLL No 93 400 Untreated Yes Unmutated del1q, del6q, del13q 
 BL15 CLL Yes 52 000 Untreated Yes Unmutated Normal karyotype 
 BL16 CLL Yes 141 000 Untreated Yes Mutated del(13q) 
 BL17 CLL Yes 82 700 Untreated No Unmutated del(17p), tri12, del(13q) 
 BL18 CLL Yes 177 000 Untreated Yes Mutated NA 
 BL19 CLL Yes 40 000 Untreated Yes Mutated del(13q) 
 BL20 CLL Yes 140 000 Untreated Yes Mutated del(13q), del(17p), complex karyotype 
 BL21 CLL Yes 80 000 Untreated No Mutated del(11q) 
 BL22 CLL Yes 76 300 Untreated No Mutated tri12 
 BL23 CLL Yes 150 000 Untreated No Mutated Normal karyotype 
 BL24 CLL Yes 152 900 Untreated Yes Mutated del13q, del11q 
Blood samples from idelalisib-treated patients         
 BL25 CLL Yes 7 800 RF, R-Benda Yes Unmutated t(4;11) 
 BL26 CLL Yes 21 300 RFC, R-Benda Yes Unmutated del(11q) 
 BL27 CLL Yes 8 500 CLB, R-Benda Yes Unmutated del(5q), del(6q), del(13q), complex karyotype 
 BL28 CLL No 100 000 RFC, R-Benda Yes Unmutated del(17p), complex karyotype 
 BL29 CLL Yes 110 000 FC, R-Benda Yes Mutated tri12 
 BL30 CLL Yes 250 000 RF, R-Benda Yes Unmutated Normal karyotype 
LN biopsy         
 LN01 SLL No 2 000 R-CLB Yes NA NA 
 LN02 SLL No 1 200 Untreated No NA NA 
 LN03 SLL No 2 000 Untreated No NA NA 
 LN04 SLL No 1 400 Untreated No NA NA 
 LN05 SLL No 2 900 Untreated Yes NA NA 
 LN06 SLL No 1 800 Untreated Yes NA NA 
 LN07 SLL No 4 000 Untreated No NA NA 
 LN08 SLL No 4 000 RCD Yes NA NA 
 LN09 CLL No 7 500 Untreated No NA NA 
 LN10 CLL No 95 000 RFC Yes Mutated t(14;18) 
 LN11 CLL No 35 000 Untreated No NA NA 
 LN12 CLL No 20 000 CLB NA NA Normal karyotype 
 LN13 CLL No 300 000 CLB Yes NA NA 
 LN14 CLL No 11 200 Untreated No Mutated del(11q) 
 LN15 CLL No 7 650 Pento-RC Yes Unmutated del(17p) 
 LN16 CLL No 18 100 Untreated No NA NA 
 LN17 CLL No 90 000 Untreated No NA NA 
 LN18 CLL NA NA NA NA Na NA NA 
 LN19 CLL Yes 60 000 Untreated Yes Unmutated del(11q) 
 LN20 CLL Yes 20 000 CLB, R-mini-CHVP Yes NA del(13q), del(11q) 
 LN21 CLL Yes 50 000 CLB Yes Unmutated del(17p) 
 LN22 CLL Yes 138 000 RFC Yes Mutated Normal karyotype 
 LN23 CLL Yes 62 000 Mabcampath Yes Unmutated del(11q), del(17p), complex karyotype 
 LN24 CLL Yes 95 000 RFC Yes Unmutated del(11q) 
 LN25 CLL Yes 6 700 R-CVP Yes Mutated tri12, tri18, del(13q) 
 LN26 CLL Yes 61 000 R-Benda Yes Unmutated del(11q) 
 LN27 CLL Yes 13 300 Mini-CHVP Yes NA del(11q), complex karyotype 
 LN28 CLL Yes 16 000 Untreated Yes Unmutated del11q 
Patient no.CLL/ SLLBulkyBinet stageCirculating count, cells/mm3TreatmentsActive disease IWCLL 2008 criteriaIgVHMutations
Blood samples from untreated patients         
 BL01 SLL Yes 3 300 Untreated No Unmutated tri12 
 BL02 CLL Yes 30 000 Untreated No Unmutated del(11q), del(13q), del(14q), complex karyotype 
 BL03 CLL Yes 22 000 Untreated No Unmutated del(11q) 
 BL04 CLL No 120 000 Untreated No Mutated t(14;18) 
 BL05 CLL No 50 000 Untreated No Mutated Normal karyotype 
 BL06 CLL No 45 000 Untreated No Unmutated tri12, del(6q), simple karyotype 
 BL07 CLL No 106 000 Untreated Yes Unmutated del(13q) 
 BL08 CLL No 130 000 Untreated No Unmutated Normal karyotype 
 BL09 CLL No 21 000 Untreated No Mutated del(13q), tri12, tri19 
 BL10 CLL No 77 000 Untreated No Mutated Normal karyotype 
 BL11 CLL No 79 860 Untreated Yes Unmutated t(3;8), del6q 
 BL12 CLL No 65 710 Untreated No Mutated del13q, del11q 
 BL13 CLL No 111 710 Untreated No NA NA 
 BL14 CLL No 93 400 Untreated Yes Unmutated del1q, del6q, del13q 
 BL15 CLL Yes 52 000 Untreated Yes Unmutated Normal karyotype 
 BL16 CLL Yes 141 000 Untreated Yes Mutated del(13q) 
 BL17 CLL Yes 82 700 Untreated No Unmutated del(17p), tri12, del(13q) 
 BL18 CLL Yes 177 000 Untreated Yes Mutated NA 
 BL19 CLL Yes 40 000 Untreated Yes Mutated del(13q) 
 BL20 CLL Yes 140 000 Untreated Yes Mutated del(13q), del(17p), complex karyotype 
 BL21 CLL Yes 80 000 Untreated No Mutated del(11q) 
 BL22 CLL Yes 76 300 Untreated No Mutated tri12 
 BL23 CLL Yes 150 000 Untreated No Mutated Normal karyotype 
 BL24 CLL Yes 152 900 Untreated Yes Mutated del13q, del11q 
Blood samples from idelalisib-treated patients         
 BL25 CLL Yes 7 800 RF, R-Benda Yes Unmutated t(4;11) 
 BL26 CLL Yes 21 300 RFC, R-Benda Yes Unmutated del(11q) 
 BL27 CLL Yes 8 500 CLB, R-Benda Yes Unmutated del(5q), del(6q), del(13q), complex karyotype 
 BL28 CLL No 100 000 RFC, R-Benda Yes Unmutated del(17p), complex karyotype 
 BL29 CLL Yes 110 000 FC, R-Benda Yes Mutated tri12 
 BL30 CLL Yes 250 000 RF, R-Benda Yes Unmutated Normal karyotype 
LN biopsy         
 LN01 SLL No 2 000 R-CLB Yes NA NA 
 LN02 SLL No 1 200 Untreated No NA NA 
 LN03 SLL No 2 000 Untreated No NA NA 
 LN04 SLL No 1 400 Untreated No NA NA 
 LN05 SLL No 2 900 Untreated Yes NA NA 
 LN06 SLL No 1 800 Untreated Yes NA NA 
 LN07 SLL No 4 000 Untreated No NA NA 
 LN08 SLL No 4 000 RCD Yes NA NA 
 LN09 CLL No 7 500 Untreated No NA NA 
 LN10 CLL No 95 000 RFC Yes Mutated t(14;18) 
 LN11 CLL No 35 000 Untreated No NA NA 
 LN12 CLL No 20 000 CLB NA NA Normal karyotype 
 LN13 CLL No 300 000 CLB Yes NA NA 
 LN14 CLL No 11 200 Untreated No Mutated del(11q) 
 LN15 CLL No 7 650 Pento-RC Yes Unmutated del(17p) 
 LN16 CLL No 18 100 Untreated No NA NA 
 LN17 CLL No 90 000 Untreated No NA NA 
 LN18 CLL NA NA NA NA Na NA NA 
 LN19 CLL Yes 60 000 Untreated Yes Unmutated del(11q) 
 LN20 CLL Yes 20 000 CLB, R-mini-CHVP Yes NA del(13q), del(11q) 
 LN21 CLL Yes 50 000 CLB Yes Unmutated del(17p) 
 LN22 CLL Yes 138 000 RFC Yes Mutated Normal karyotype 
 LN23 CLL Yes 62 000 Mabcampath Yes Unmutated del(11q), del(17p), complex karyotype 
 LN24 CLL Yes 95 000 RFC Yes Unmutated del(11q) 
 LN25 CLL Yes 6 700 R-CVP Yes Mutated tri12, tri18, del(13q) 
 LN26 CLL Yes 61 000 R-Benda Yes Unmutated del(11q) 
 LN27 CLL Yes 13 300 Mini-CHVP Yes NA del(11q), complex karyotype 
 LN28 CLL Yes 16 000 Untreated Yes Unmutated del11q 

Benda, bendamustine; CLB, chlorambucil; CVP, cyclophosphamide; del, deletion; FC, fludarabine, cyclophosphamide; IgVH, immunoglobulin variable region heavy chain; IWCLL, International Workshop on Chronic Lymphocytic Leukaemia; mini-CHVP, cyclophosphamide, doxorubicin, vindesine, prednisone; NA, not available; Pento, pentostatine; R, rituximab; RC, rituximab, cyclophosphamide; RCD, rituximab, cyclophosphamide, dexamethasone; RF, rituximab, fludarabine; RFC, rituximab, fludarabine, cyclophosphamide; t, translocation; tri, trisomy.

Intravital microscopy and multiphoton in vivo imaging

Wide-field intravital microscopy was performed as previously described.14,15  In brief, calcein-labeled PBMCs were injected into the right femoral artery of C57BL/6 mice (Charles River Laboratories), and fluorescent events in the left inguinal LN microcirculation were visualized and recorded. Rolling fractions (the percentage of rolling cells in the total flux of cells in each venule), sticking fractions (the percentage of rolling cells that subsequently arrested for >30 seconds), and rolling speed (rolling velocity [Vroll]) were determined as previously described.14,15  When indicated, cells were pretreated for 30 minutes with a function blocking anti-L-selectin (CD62L) antibody (Ab) (6 μg/mL, DREG56; eBioscience) or with an isotype control. Multiphoton imaging was performed on a 7MP upright microscope (Carl Zeiss). Excitation was provided by a Ti-Sapphire femtosecond laser, Chameleon Ultra 2 (Coherent Inc) tuned at 800 nm; emitted fluorescence was collected with 2 non-descanned detectors: Channel 2 (500-550 nm) for carboxyfluorescein diacetate succinimidyl ester (CFSE)-labeled PBMCs and Channel 3 (565-610 nm) for Qtracker-565 (Molecular Probes)-labeled vessels. Mice were anesthetized and placed on a customized stage for securing mice and immobilizing the inguinal LN on a heating pad with temperature feedback to an mTCII micro-Temperature Controller (Cell MicroControls). The crawling speed (crawling velocity [Vcrawl]) was determined with Imaris (Bitplane) on image sequences. See supplemental Methods (available on the Blood Web site) for additional information.

Histology and confocal microscopy

LNs were either fixed in 10% formalin and embedded in paraffin or embedded in optimal cutting-temperature compounds and snap-frozen in liquid nitrogen. For histology, 3-µm-thick sections were used; for confocal microscopy, 5-µm-thick sections were used. HEV density and HEV pockets were analyzed using the HEV-specific antibody MECA-79 (ATCC). The detailed procedures and antibodies used are described in supplemental Methods. Panoramic Viewer and HistoQuant software were used for, respectively, viewing and analyzing the digitalized slides (RTM 1.5.053; 3D Histech).

Flow cytometry

Freshly isolated PBMCs were incubated with the following fluorochrome-conjugated antibodies (Abs), CD19-phycoerythrin (PE; HIB19), CD5-fluorescein isothiocyanate (4CHT2), CD62L-PECy5 (DREG-56), CXCR4-allophycocyanin (12G5), or CD11a-PE (HI-111) (all from BD Biosciences), and analyzed with a FACSCalibur flow cytometer (BD Biosciences) and FlowJo software (Tree Star). The mean fluorescence intensity (MFI) ratio (MFIR) was determined by dividing the MFI of the analyzed antigens by the MFI of the isotype control. In some experiments, freshly isolated PBMCs were incubated ex vivo for the indicated time at 37°C with idelalisib (5 µM; Selleckchem) or dimethylsulfoxide (untreated conditions) before analysis by flow cytometry.

Statistics

Data are shown as mean ± standard deviation (SD) or mean ± standard error of the mean (as indicated) and were analyzed using a 2-tailed unpaired Student t test. When indicated, a 2-tailed paired Student t test or Mann-Whitney test were instead applied. Differences were considered statistically significant when P < .05.

Table 1 presents the characteristics of the different groups of patients analyzed in this study. Blood samples (BLs) were obtained from untreated CLL/SLL patients (with or without bulky disease) and idelalisib-treated CLL patients. For the idelalisib-treated group, blood was collected both before and under treatment of each patient. LN biopsies were obtained from a different group of CLL/SLL patients with various clinical characteristics.

CD20+ CLL cells accumulate within HEV pockets in CLL LNs

To analyze the HEV network, we stained human CLL and control LNs with the HEV-specific Ab MECA-79 which recognizes sulfated ligands for lymphocytes (Figure 1A). HEV density was significantly increased in CLL LNs (n = 13) and SLL LNs (n = 5) in comparison with control LNs (n = 8) (Figure 1B). HEVs were located both outside and inside proliferation centers (Figure 1C-D), and expressed the 6-sulfosialyl Lewis X oligosaccharides ligands for the lymphocyte homing receptor L-selectin, recognized by the HEV-specific Abs G72 and G15221  (Figure 1E). L-selectin+ cells were located inside, around (<10 μm), or at a distance (>10 μm) from MECA-79+ HEVs (Figure 1F). However, there was no specific enrichment of L-selectin+ cells around MECA-79+ HEVs (Figure 1G), probably because L-selectin shedding occurs during extravasation through HEVs.22 

Figure 1

Analysis of the HEV network in CLL/SLL LNs. (A) Representative images showing the density and the distribution of HEV blood vessels (MECA-79+, green) in a reactive LN (control; left) and a CLL LN (CLL; right). Scale bar, 500 µm. (B) HEV density (number of HEVs/mm2) was quantified on control (n = 8), SLL (n = 5), and CLL (n = 13) LNs. Unpaired t test; *P < .05. (C) CLL LNs were double-stained with MECA-79 (brown) and Abs directed against Ki-67 (red). Scale bar, 100 µm. (D) Relative MECA-79+ area was quantified inside proliferation centers (PCs) (sum of MECA-79+ area inside all PC/sum of PC area × 100) and outside area (sum of MECA-79+ area outside all PC/overall area − sum of PC area × 100) in CLL/SLL LNs (n = 19). PCs were revealed by Ki-67 staining. Paired t test. (E) Representative image showing staining of HEV blood vessels in CLL LNs with HEV-specific Abs G72 and G152 (brown). Scale bar, 20 µm. (F) Double staining of a CLL LN section with anti-L-selectin Ab (brown) and HEV-specific Ab MECA-79 (red), showing L-selectin+ cells (arrows) inside, around, or at a distance from MECA-79+ HEVs. Scale bar, 20 µm. (G) Quantification of the percentage of L-selectin+ cells inside (inside HEV), around (<10 µm from HEV), or at a distance (>10 µm from HEV) from MECA-79+ HEVs. A total of 362 L-selectin+ cells were counted on 5 different sections from 2 different patients. DAPI, 4′,6 diamidino-2-phenylindole; ns, not significant.

Figure 1

Analysis of the HEV network in CLL/SLL LNs. (A) Representative images showing the density and the distribution of HEV blood vessels (MECA-79+, green) in a reactive LN (control; left) and a CLL LN (CLL; right). Scale bar, 500 µm. (B) HEV density (number of HEVs/mm2) was quantified on control (n = 8), SLL (n = 5), and CLL (n = 13) LNs. Unpaired t test; *P < .05. (C) CLL LNs were double-stained with MECA-79 (brown) and Abs directed against Ki-67 (red). Scale bar, 100 µm. (D) Relative MECA-79+ area was quantified inside proliferation centers (PCs) (sum of MECA-79+ area inside all PC/sum of PC area × 100) and outside area (sum of MECA-79+ area outside all PC/overall area − sum of PC area × 100) in CLL/SLL LNs (n = 19). PCs were revealed by Ki-67 staining. Paired t test. (E) Representative image showing staining of HEV blood vessels in CLL LNs with HEV-specific Abs G72 and G152 (brown). Scale bar, 20 µm. (F) Double staining of a CLL LN section with anti-L-selectin Ab (brown) and HEV-specific Ab MECA-79 (red), showing L-selectin+ cells (arrows) inside, around, or at a distance from MECA-79+ HEVs. Scale bar, 20 µm. (G) Quantification of the percentage of L-selectin+ cells inside (inside HEV), around (<10 µm from HEV), or at a distance (>10 µm from HEV) from MECA-79+ HEVs. A total of 362 L-selectin+ cells were counted on 5 different sections from 2 different patients. DAPI, 4′,6 diamidino-2-phenylindole; ns, not significant.

Close modal

Confocal microscopy analysis revealed that CD20+ CLL cells are frequently found within HEV pockets in human CLL LNs (Figure 2). HEV pockets are highly dynamic structures that are closely associated with lymphocyte extravasation through HEVs.13,23,24  Indeed, they have been shown to disappear when lymphocyte migration to LNs is inhibited.23,24  CD20+ cells nested in HEV pockets were separated from the HEV lumen by the membrane of the HEV endothelial cells stained with MECA-79 and CD31 antibodies (Figure 2). As previously described in murine LNs,23  HEV pockets in human CLL LNs were able to house up to 4 to 5 CD20+ cells. Together, these observations suggested intense trafficking of CLL cells through HEVs in CLL LNs.

Figure 2

CD20+ CLL cells accumulate within HEV pockets in CLL LNs. Four-color confocal images showing the accumulation of CD20+ CLL cells (light blue) within HEV pockets (arrowheads) in CLL LNs. HEV endothelial cells were stained with MECA-79 (green) and CD31 or von Willebrand factor (vWF; red). Counterstaining was done with DAPI (dark blue). HEV pockets were found to house up to 4 CD20+ CLL cells. 63×, zoom 3; scale bar, 5 µm.

Figure 2

CD20+ CLL cells accumulate within HEV pockets in CLL LNs. Four-color confocal images showing the accumulation of CD20+ CLL cells (light blue) within HEV pockets (arrowheads) in CLL LNs. HEV endothelial cells were stained with MECA-79 (green) and CD31 or von Willebrand factor (vWF; red). Counterstaining was done with DAPI (dark blue). HEV pockets were found to house up to 4 CD20+ CLL cells. 63×, zoom 3; scale bar, 5 µm.

Close modal

Human CLL cells roll, stick, and crawl on HEV endothelium in vivo

To visualize the interaction of human CLL cells with HEV endothelium in vivo, we next performed intravital microscopy analyses in LNs of wild-type mice, making use of the evolutionary conservation of the vasculature and the unique capacity of human leukemic cells to migrate to murine bone marrow and lymphoid organs.25  The venular tree of the inguinal LN consists of up to 5 branching orders (I-V), and higher order venules (III-V) correspond to HEVs13-15  (Figure 3A). Human CLL cells, obtained from freshly isolated samples to avoid any phenotypic alteration after culture or freezing, were fluorescently labeled ex vivo, and injected in recipient mice for in vivo imaging of their behavior in the LN microcirculation. Human CLL cells were observed to roll and stick (arrest) within LN venules with numerous interactions in order III to V HEVs (Figure 3B). More interactions were observed for CLL cells from patients with bulky disease and high circulating lymphocyte counts (≥40 000 cells/mm3; Table 1; supplemental Videos 1-2) than for CLL cells from patients without bulky disease (Table 1; supplemental Video 3). Indeed, 60% of the CLL cells from patients with bulky disease rolled in order V HEVs and >10% of these cells arrested, compared with <30% and 3%, respectively, for CLL cells from patients with no bulky disease (Figure 3B). In addition, CLL cells from patients with bulky disease rolled more slowly in HEVs than cells from patients without bulky disease (Figure 3C). Intravital 2-photon laser-scanning microscopy revealed that after arrest, CLL cells were able to crawl on the HEV endothelium, some of them in the opposite direction of blood flow (Figure 3D; supplemental Video 4). The average intraluminal velocity of the CLL cells was ∼6 μm per minute (Figure 3E). This velocity is similar to that of murine B cells in LN HEVs.26  We concluded that human CLL cells are able to roll, stick, and crawl on HEV endothelium in vivo.

Figure 3

CLL cells roll, stick, and crawl within LN HEVs in vivo. (A) Visualization of the mouse inguinal LN microcirculation by intravital microscopy. HEV blood vessels were revealed by IV injection of Alexa 488–conjugated MECA-79 Ab. The different blood vessel orders are indicated (I-V). (B) Rolling and sticking of human CLL cells within mouse LN HEVs. The rolling fraction (left panel) and sticking fraction (right panel) of calcein-labeled CLL cells in the indicated venules are shown. Data represent the mean ± SD of independent experiments conducted with CLL cells from 7 CLL patients with no bulky disease (black) and 6 CLL patients with bulky disease and high circulating lymphocyte counts (≥40 000 cells/mm3; red). Unpaired t test; *P < .05, **P < .01, ***P < .001. (C) CLL cells from patients with bulky disease roll more slowly within LN HEVs. Data represent the percentages of rolling cells at or below a given velocity (Vroll) in the indicated venules. The median velocity of cells from CLL patients with no bulky disease was 220.7 and 124 µm.s−1 in order IV and V, respectively. The median velocity of cells from CLL patients with bulky disease was 79.6 and 48.3 µm.s−1 in order IV and V, respectively (50 cells/venular order from 5 patients were analyzed). Mann-Whitney test; ***P < .001. (D) Two-photon microscopy time-lapse images showing CFSE-labeled CLL cells from patients with bulky disease (arrowheads) crawling on HEV endothelium. Alexa 647–conjugated MECA-79 staining is shown on the first image. Yellow arrows indicate blood flow direction. Scale bar, 40 µm. (E) The median crawling speed (Vcrawl) was determined from 2 independent experiments (n = 21 cells).

Figure 3

CLL cells roll, stick, and crawl within LN HEVs in vivo. (A) Visualization of the mouse inguinal LN microcirculation by intravital microscopy. HEV blood vessels were revealed by IV injection of Alexa 488–conjugated MECA-79 Ab. The different blood vessel orders are indicated (I-V). (B) Rolling and sticking of human CLL cells within mouse LN HEVs. The rolling fraction (left panel) and sticking fraction (right panel) of calcein-labeled CLL cells in the indicated venules are shown. Data represent the mean ± SD of independent experiments conducted with CLL cells from 7 CLL patients with no bulky disease (black) and 6 CLL patients with bulky disease and high circulating lymphocyte counts (≥40 000 cells/mm3; red). Unpaired t test; *P < .05, **P < .01, ***P < .001. (C) CLL cells from patients with bulky disease roll more slowly within LN HEVs. Data represent the percentages of rolling cells at or below a given velocity (Vroll) in the indicated venules. The median velocity of cells from CLL patients with no bulky disease was 220.7 and 124 µm.s−1 in order IV and V, respectively. The median velocity of cells from CLL patients with bulky disease was 79.6 and 48.3 µm.s−1 in order IV and V, respectively (50 cells/venular order from 5 patients were analyzed). Mann-Whitney test; ***P < .001. (D) Two-photon microscopy time-lapse images showing CFSE-labeled CLL cells from patients with bulky disease (arrowheads) crawling on HEV endothelium. Alexa 647–conjugated MECA-79 staining is shown on the first image. Yellow arrows indicate blood flow direction. Scale bar, 40 µm. (E) The median crawling speed (Vcrawl) was determined from 2 independent experiments (n = 21 cells).

Close modal

The lymphocyte homing receptor L-selectin controls the adhesion of CLL cells to HEV walls in vivo

We then investigated the molecular mechanisms controlling the behavior of CLL cells within LN HEVs. We first analyzed the expression of the lymphocyte homing receptor L-selectin,17,18  which mediates the initial capture and rolling interactions of lymphocytes on HEV endothelium,13,16,27,28  and is known to be expressed on CLL cells.29,30  We found that L-selectin is expressed at significantly higher levels on CLL cells from patients with bulky disease compared with cells from patients without bulky disease (Figure 4A). In contrast, similar expression levels between the 2 subgroups of patients were observed for chemokine receptor CXCR4 and integrin LFA-1, which mediates the sticking of lymphocytes to HEV endothelial cells.13  In the course of these studies, we identified a third subgroup of patients with bulky disease but lower circulating lymphocyte counts (<40 000 cells/mm3; CLL/SLL patients BL01-03, Table 1). Interestingly, CLL/SLL cells from this subgroup expressed low levels of L-selectin and had a reduced capacity to roll and stick to HEVs (supplemental Figure 1), compared with CLL cells from patients with bulky disease and high circulating lymphocyte counts (≥40 000 cells/mm3; CLL patients BL15-BL24, Table 1). Together, these results suggested that differences in L-selectin expression could explain the different behavior of CLL cells in LNs HEVs in vivo. Functional analyses revealed that a blocking antibody to L-selectin had a strong inhibitory effect on CLL trafficking in LN HEVs; it reduced the rolling and completely abrogated the sticking of CLL cells to HEV walls (Figure 4B). We obtained similar results with circulating CLL cells from 3 patients with bulky disease (Figure 4B-D). Interestingly, reduction in rolling and sticking upon blockade of L-selectin also occurred with CLL cells from patients without bulky disease (3 patients shown in Figure 4C-D). We concluded that L-selectin is the critical determinant controlling the initial rolling and sticking interactions of CLL cells with HEV endothelium in vivo.

Figure 4

L-selectin is the key factor controlling the arrest of CLL cells within LN HEVs. (A) Flow cytometry analysis of the expression of L-selectin, LFA-1, and CXCR4 on CD19+CD5+ CLL cells. Graphs represent the MFIR of CLL cells from patients without (●, n = 7) or with (▪, n = 7) bulky disease. Mean ± SEM are shown. Unpaired t test; *P < .05. (B-D) Calcein-labeled CLL cells from patients were untreated (black) or treated with a function blocking anti-L-selectin Ab (white) or an isotype control Ab (gray). (B) The rolling and sticking fractions of CLL cells from a patient with bulky disease in the indicated venules are shown. Mean ± SD, venular order III (n = 3), order IV (n = 3), order V (n = 5). Unpaired t test; *P < .05; **P < .01. (C) The rolling fractions of CLL cells from 2 other patients with bulky disease and 3 patients with no bulky disease in venular order V are shown. Mean ± SD, venular order V (n = 3). (D) The sticking fractions of CLL cells from 1 representative patient (of 3) with bulky disease (BL18) and no bulky disease (BL11) in venular order V are shown. Mean ± SD, venular order V (n = 3).

Figure 4

L-selectin is the key factor controlling the arrest of CLL cells within LN HEVs. (A) Flow cytometry analysis of the expression of L-selectin, LFA-1, and CXCR4 on CD19+CD5+ CLL cells. Graphs represent the MFIR of CLL cells from patients without (●, n = 7) or with (▪, n = 7) bulky disease. Mean ± SEM are shown. Unpaired t test; *P < .05. (B-D) Calcein-labeled CLL cells from patients were untreated (black) or treated with a function blocking anti-L-selectin Ab (white) or an isotype control Ab (gray). (B) The rolling and sticking fractions of CLL cells from a patient with bulky disease in the indicated venules are shown. Mean ± SD, venular order III (n = 3), order IV (n = 3), order V (n = 5). Unpaired t test; *P < .05; **P < .01. (C) The rolling fractions of CLL cells from 2 other patients with bulky disease and 3 patients with no bulky disease in venular order V are shown. Mean ± SD, venular order V (n = 3). (D) The sticking fractions of CLL cells from 1 representative patient (of 3) with bulky disease (BL18) and no bulky disease (BL11) in venular order V are shown. Mean ± SD, venular order V (n = 3).

Close modal

Idelalisib treatment results in decreased L-selectin expression on circulating CLL cells in vivo

Idelalisib (formerly known as GS-1101 and CAL-101) is a drug, recently approved for CLL patients, which induces LN shrinkage and lymphocytosis as a consequence of CLL cell redistribution into the peripheral blood.5,19,20,31  To study the effect of idelalisib on CLL cell trafficking to LNs in vivo, we first measured the expression level of L-selectin at the surface of CLL cells, before and during idelalisib treatment. Interestingly, we observed that L-selectin expression is decreased on circulating CLL cells from patients treated with idelalisib (Figure 5A; supplemental Figure 2). Intravital microscopy analysis of CLL cells from a patient with bulky disease before treatment and after 3 or 7 weeks of treatment revealed that rolling and sticking interactions of CLL cells with HEV endothelium in vivo are reduced after treatment with idelalisib (Figure 5B). The inhibition of CLL cell sticking to HEV walls was associated with a fourfold increase in the rolling velocity of CLL cells after idelalisib treatment (Figure 5C), suggesting that cells rolled too quickly for stable arrest. To determine whether the effect of idelalisib on L-selectin expression is direct, we performed additional in vitro experiments with circulating CLL cells from 5 individual patients (Figure 5D). However, we did not observe any change in the expression of L-selectin on CLL cells after up to 4 hours of incubation with idelalisib, suggesting an indirect effect of the drug in vivo (Figure 5E). We concluded that idelalisib treatment induces a downregulation of L-selectin on CLL cells in vivo, that may impair HEV-mediated recruitment of CLL cells in LNs.

Figure 5

Idelalisib treatment is associated with decreased expression of L-selectin on circulating CLL cells in vivo. (A) Graph represents the MFIR of L-selectin on CLL cells from patients (n = 6) under idelalisib treatment (−, before treatment; +, under treatment). Paired t test; *P < .05. (B) Rolling and sticking fractions of calcein-labeled CLL cells from a patient with bulky disease are shown before treatment, and at 3 or 7 weeks under idelalisib treatment. Mean ± SD, venular order III (n = 3), order IV (n = 4), order V (n = 3-4). Unpaired t test; *P < .05, **P < .01. (C) Data represent the percentages of CLL cells rolling at or below a given velocity (Vroll). The median velocity of cells before treatment, and at week 3 or week 7 was 69.59, 475.1, and 299.8 µm.s−1, respectively, in order IV and 51.27, 247.2, and 185.9 µm.s−1, respectively, in order V (19 to 30 cells per venular order were analyzed). Mann-Whitney test; ***P < .001. (D) Graph represents the MFIR of L-selectin on CLL cells from patients (n = 5) incubated ex vivo with (+) or without (−) idelalisib during 4 hours. Paired t test. (E) MFIR of L-selectin on CLL cells from 2 different patients, incubated ex vivo with (+) or without (−) idelalisib, during the indicated time (hours). Mean ± SEM, n = 3 experimental replicates. Data are representative of 3 independent experiments.

Figure 5

Idelalisib treatment is associated with decreased expression of L-selectin on circulating CLL cells in vivo. (A) Graph represents the MFIR of L-selectin on CLL cells from patients (n = 6) under idelalisib treatment (−, before treatment; +, under treatment). Paired t test; *P < .05. (B) Rolling and sticking fractions of calcein-labeled CLL cells from a patient with bulky disease are shown before treatment, and at 3 or 7 weeks under idelalisib treatment. Mean ± SD, venular order III (n = 3), order IV (n = 4), order V (n = 3-4). Unpaired t test; *P < .05, **P < .01. (C) Data represent the percentages of CLL cells rolling at or below a given velocity (Vroll). The median velocity of cells before treatment, and at week 3 or week 7 was 69.59, 475.1, and 299.8 µm.s−1, respectively, in order IV and 51.27, 247.2, and 185.9 µm.s−1, respectively, in order V (19 to 30 cells per venular order were analyzed). Mann-Whitney test; ***P < .001. (D) Graph represents the MFIR of L-selectin on CLL cells from patients (n = 5) incubated ex vivo with (+) or without (−) idelalisib during 4 hours. Paired t test. (E) MFIR of L-selectin on CLL cells from 2 different patients, incubated ex vivo with (+) or without (−) idelalisib, during the indicated time (hours). Mean ± SEM, n = 3 experimental replicates. Data are representative of 3 independent experiments.

Close modal

In this study, we investigated the first step of CLL cell migration to LNs: their interaction with HEV blood vessels. We made use of the evolutionary conservation of the vasculature to analyze the behavior of human CLL cells in the mouse LN microcirculation using intravital microscopy and multiphoton in vivo imaging. This strategy allowed us to visualize for the first time the rolling, sticking, and crawling of CLL cells on HEV endothelium in vivo. Functional analyses revealed that the lymphocyte homing receptor L-selectin controls the interaction of CLL cells with HEVs in vivo. Interestingly, L-selectin was downregulated on CLL cells from patients treated with idelalisib, a phosphoinositide 3-kinase δ inhibitor recently approved in clinics for CLL treatment.

Our results show that, similar to B cells, CLL cells use a multistep adhesion cascade to bind to HEV walls in vivo. CLL cells circulating in the blood tether and roll on HEV walls through the binding of L-selectin to HEV endothelial cells. Rolling of CLL cells on the HEV endothelium is essential for subsequent arrest, as illustrated by the fact that a function-blocking antibody to L-selectin, which reduced rolling, completely blocked the arrest of CLL cells (Figure 4B,D). We observed a significantly increased rolling fraction and decreased rolling velocity for CLL cells from patients with bulky disease and high circulating lymphocyte counts (≥40 000 cells/mm3) (Figure 3C) which exhibit high expression levels of L-selectin (Figure 4A). The decreased rolling velocity is likely to facilitate chemokine-induced integrin-mediated arrest of CLL cells.13  In contrast, we observed a decreased sticking fraction and an increased rolling velocity (Figure 3B-C) for cells from CLL patients without bulky disease (including 3 patients with very high leukocytosis, ie, lymphocyte counts >100 000 cells/mm3), which express lower levels of L-selectin (Figure 4A). This suggested a possible defect for these cells in trafficking through LN HEVs in vivo. However, a certain level of rolling and sticking was still observed and it was dependent on L-selectin (Figure 4C-D). In addition, we observed low L-selectin expression and reduced rolling and sticking fractions for cells from patients with bulky disease but lower circulating lymphocytes counts (<40 000 cells/mm3, including 1 SLL patient; supplemental Figure 1). These patients may thus have a defect in CLL/SLL cell entry in LNs through HEVs. L-selectin expression was also decreased on CLL cells from patients after treatment with idelalisib in vivo (Figure 5A) and this was associated with increased rolling velocity (Figure 5C) and reduced sticking of CLL cells to HEV walls (Figure 5B). These observations are in agreement with previous findings showing that the number of L-selectin molecules at the cell surface dictates the rolling behavior of T and B lymphocytes within LN HEVs.32  Together, our findings indicated that L-selectin is a critical determinant of CLL cell trafficking in HEVs in vivo.

A major strength of our study is providing the first direct visualization and functional analysis of human CLL cell trafficking in the LN microcirculation in vivo. It is important to point out that the HEV ligands for L-selectin (6-sulfo-sialyl Lewis X) are sulfated sugars that are identical between human and mouse.13  These L-selectin ligands are expressed to high levels in both human and mouse HEVs, and they are recognized by the HEV-specific monoclonal Ab (mAb) MECA-79, both in human and mouse. L-selectin binding to HEV endothelial cells is thus evolutionarily conserved between human and mouse. Indeed, our intravital microscopy studies with function-blocking antibodies against human L-selectin (Figure 4B-D) convincingly demonstrate that L-selectin controls the rolling and firm arrest (sticking) of human CLL cells in LN HEVs in vivo. In the future, it would be interesting to further characterize the role of L-selectin in CLL trafficking through HEVs in mouse models of CLL, for instance by using the well-characterized Eμ-TCL1 model.33  Finally, it would have been nice to complement our in vivo imaging studies in mice with in vitro studies using human HEV endothelial cells. Unfortunately, it is not possible to culture human HEV endothelial cells in vitro because we showed previously that they rapidly de-differentiate outside of the lymphoid tissue microenvironment.34  In addition, no human HEV cell line or cultured human endothelial cells that exhibit the bona fide HEV phenotype (ie, expressing the sulfated ligands recognized by L-selectin and HEV-specific mAb MECA-79) have been described yet.

We found that the density of HEVs was increased in human CLL LNs and that CD20+ CLL cells frequently accumulated within HEV pockets (Figure 2). To the best of our knowledge, this is the first description of HEV pockets in human LNs. In mouse LNs, HEV pockets have been shown to function as “waiting areas” in which lymphocytes were held until space was made available to them for entry into the LN tissue.23  They were found to be highly dynamic structures that are continuously altered in size and location by lymphocyte migration. Interestingly, an antibody that blocks L-selectin–mediated lymphocyte entry in LNs through HEVs induced the disappearance of HEV pockets in vivo.23  Therefore, the presence of many HEV pockets occupied by CLL cells in human CLL LNs strongly suggests that HEVs mediate high levels of CLL cell entry in LNs.

In this study, we have focused on CLL cell entry (or re-entry) in LNs. However, another key aspect in CLL cell trafficking is the egress from LNs. In normal B cells, this process depends on the sphingosine-1 phosphate receptor S1PR1,13,26  a receptor which has been shown to be reduced on CLL cells from patients with unfavorable prognosis.35  Defective egress caused by impaired S1PR1 expression was proposed to contribute to the extended survival of CLL cells by prolonging their residency in the prosurvival niche of LNs.35  Egress occurs only minimally in SLL, and differential expression of S1PR1 may also explain some of the differences between CLL and SLL. Interestingly, idelalisib has recently been shown to increase S1PR1 expression on CLL cells,36  an effect that could contribute to the rapid resolution of lymphadenopathy (LN shrinkage) and redistribution of CLL cells into the blood (lymphocytosis) that have been observed after idelalisib treatment.19,31  In this study, we found that treatment with idelalisib results in decreased levels of L-selectin on CLL cells in vivo and reduced binding of CLL cells to HEV walls in LNs. We do not know the mechanism for this downregulation of L-selectin in vivo but it may be related to the profound CLL cell redistribution induced by idelalisib5,19,20,32  because our in vitro studies indicate that it is not a direct effect. Idelalisib has been shown to interfere with integrin-mediated adhesion of CLL cells to endothelial cells in vitro37  and to inhibit chemotaxis of CLL cells toward lymphoid chemokines.20  Idelalisib could thus not only induce exit of CLL cells from LNs,5,20  but also inhibit their re-entry through HEVs via L-selectin downregulation and interference with integrin-mediated adhesion and chemotaxis. Both effects, induction of CLL cell exit and inhibition of re-entry, could contribute to LN shrinkage and redistribution of CLL cells into the blood.

Together, our findings suggest that interfering with L-selectin–mediated CLL cell trafficking in HEVs could represent a novel strategy to block dissemination of CLL cells to LNs. This strategy could be particularly useful to increasing the efficacy of conventional therapy for advanced CLL patients with bulky lymphadenopathy and high levels of L-selectin on their leukemic cells. Targeting L-selectin might prove to be better than targeting chemokine receptors because multiple chemokine receptors (ie, CXCR4, CXCR5, CCR7) contribute to CLL cell migration,8-12  whereas a single molecule (ie, L-selectin) controls the initial capture and rolling interaction of CLL cells with HEV endothelium in vivo.

The online version of this article contains a data supplement.

There is an Inside Blood Commentary on this article in this issue.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The authors thank the Institut de Pharmacologie et de Biologie Structurale (IPBS)TRI facility, the IPBSANEXPLO facility, and the INSERM Unité Mixte de Recherche 1043 cellular imaging facility. The authors are grateful to R. Kannagi for gift of Abs G72 and G152, V. Thuries, L. Jalabert, and G. Perez for help with immunohistochemistry, and F. X. Frenois for whole-slide imaging studies (Service d’Anatomie et de Cytologie Pathologique, IUCT-Oncopôle); they are also grateful to all patients and their families and to the SILLC Association.

This work was supported by Laboratoire d’Excellence Toulouse Cancer (LABEX TOUCAN, “Integrative Analysis of Resistance in Hematological Cancers”), Fondation Recherche Innovation Thérapeutique Cancérologie (Fondation RITC), Région Midi-Pyrénées, and Fondation ARC Pour la Recherche sur le Cancer (ARC Equipment No. 8505).

Contribution: F.L. performed the experiments, analyzed the results, and was involved in study design and manuscript preparation; E.B., C.L., and C.M. performed the experiments and analyzed the results; J.-J.F. was involved in study design; L.Y. was involved in study design and analyzed the results; and J.-P.G. designed the study, analyzed the results, and wrote the paper.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Jean-Philippe Girard, IPBS-CNRS, 205 Route de Narbonne, 31077 Toulouse cedex 4, France; e-mail: jean-philippe.girard@ipbs.fr.

1
Chiorazzi
 
N
Rai
 
KR
Ferrarini
 
M
Chronic lymphocytic leukemia.
N Engl J Med
2005
, vol. 
352
 
8
(pg. 
804
-
815
)
2
Gaidano
 
G
Foà
 
R
Dalla-Favera
 
R
Molecular pathogenesis of chronic lymphocytic leukemia.
J Clin Invest
2012
, vol. 
122
 
10
(pg. 
3432
-
3438
)
3
Burger
 
JA
Ghia
 
P
Rosenwald
 
A
Caligaris-Cappio
 
F
The microenvironment in mature B-cell malignancies: a target for new treatment strategies.
Blood
2009
, vol. 
114
 
16
(pg. 
3367
-
3375
)
4
Caligaris-Cappio
 
F
Bertilaccio
 
MT
Scielzo
 
C
How the microenvironment wires the natural history of chronic lymphocytic leukemia.
Semin Cancer Biol
2014
, vol. 
24
 (pg. 
43
-
48
)
5
Burger
 
JA
Montserrat
 
E
Coming full circle: 70 years of chronic lymphocytic leukemia cell redistribution, from glucocorticoids to inhibitors of B-cell receptor signaling.
Blood
2013
, vol. 
121
 
9
(pg. 
1501
-
1509
)
6
Hayden
 
RE
Pratt
 
G
Roberts
 
C
Drayson
 
MT
Bunce
 
CM
Treatment of chronic lymphocytic leukemia requires targeting of the protective lymph node environment with novel therapeutic approaches.
Leuk Lymphoma
2012
, vol. 
53
 
4
(pg. 
537
-
549
)
7
Burger
 
JA
Chemokines and chemokine receptors in chronic lymphocytic leukemia (CLL): from understanding the basics towards therapeutic targeting.
Semin Cancer Biol
2010
, vol. 
20
 
6
(pg. 
424
-
430
)
8
Burger
 
JA
Burger
 
M
Kipps
 
TJ
Chronic lymphocytic leukemia B cells express functional CXCR4 chemokine receptors that mediate spontaneous migration beneath bone marrow stromal cells.
Blood
1999
, vol. 
94
 
11
(pg. 
3658
-
3667
)
9
Till
 
KJ
Lin
 
K
Zuzel
 
M
Cawley
 
JC
The chemokine receptor CCR7 and alpha4 integrin are important for migration of chronic lymphocytic leukemia cells into lymph nodes.
Blood
2002
, vol. 
99
 
8
(pg. 
2977
-
2984
)
10
López-Giral
 
S
Quintana
 
NE
Cabrerizo
 
M
, et al. 
Chemokine receptors that mediate B cell homing to secondary lymphoid tissues are highly expressed in B cell chronic lymphocytic leukemia and non-Hodgkin lymphomas with widespread nodular dissemination.
J Leukocyte Biol
2004
, vol. 
76
 
2
(pg. 
462
-
471
)
11
Trentin
 
L
Cabrelle
 
A
Facco
 
M
, et al. 
Homeostatic chemokines drive migration of malignant B cells in patients with non-Hodgkin lymphomas.
Blood
2004
, vol. 
104
 
2
(pg. 
502
-
508
)
12
Bürkle
 
A
Niedermeier
 
M
Schmitt-Gräff
 
A
Wierda
 
WG
Keating
 
MJ
Burger
 
JA
Overexpression of the CXCR5 chemokine receptor, and its ligand, CXCL13 in B-cell chronic lymphocytic leukemia.
Blood
2007
, vol. 
110
 
9
(pg. 
3316
-
3325
)
13
Girard
 
JP
Moussion
 
C
Förster
 
R
HEVs, lymphatics and homeostatic immune cell trafficking in lymph nodes.
Nat Rev Immunol
2012
, vol. 
12
 
11
(pg. 
762
-
773
)
14
Moussion
 
C
Girard
 
JP
Dendritic cells control lymphocyte entry to lymph nodes through high endothelial venules.
Nature
2011
, vol. 
479
 
7374
(pg. 
542
-
546
)
15
von Andrian
 
UH
Intravital microscopy of the peripheral lymph node microcirculation in mice.
Microcirculation
1996
, vol. 
3
 
3
(pg. 
287
-
300
)
16
Rosen
 
SD
Ligands for L-selectin: homing, inflammation, and beyond.
Annu Rev Immunol
2004
, vol. 
22
 (pg. 
129
-
156
)
17
Gallatin
 
WM
Weissman
 
IL
Butcher
 
EC
A cell-surface molecule involved in organ-specific homing of lymphocytes.
Nature
1983
, vol. 
304
 
5921
(pg. 
30
-
34
)
18
Arbonés
 
ML
Ord
 
DC
Ley
 
K
, et al. 
Lymphocyte homing and leukocyte rolling and migration are impaired in L-selectin-deficient mice.
Immunity
1994
, vol. 
1
 
4
(pg. 
247
-
260
)
19
Furman
 
RR
Sharman
 
JP
Coutre
 
SE
, et al. 
Idelalisib and rituximab in relapsed chronic lymphocytic leukemia.
N Engl J Med
2014
, vol. 
370
 
11
(pg. 
997
-
1007
)
20
Hoellenriegel
 
J
Meadows
 
SA
Sivina
 
M
, et al. 
The phosphoinositide 3′-kinase delta inhibitor, CAL-101, inhibits B-cell receptor signaling and chemokine networks in chronic lymphocytic leukemia.
Blood
2011
, vol. 
118
 
13
(pg. 
3603
-
3612
)
21
Mitsuoka
 
C
Sawada-Kasugai
 
M
Ando-Furui
 
K
, et al. 
Identification of a major carbohydrate capping group of the L-selectin ligand on high endothelial venules in human lymph nodes as 6-sulfo sialyl Lewis X.
J Biol Chem
1998
, vol. 
273
 
18
(pg. 
11225
-
11233
)
22
Faveeuw
 
C
Preece
 
G
Ager
 
A
Transendothelial migration of lymphocytes across high endothelial venules into lymph nodes is affected by metalloproteinases.
Blood
2001
, vol. 
98
 
3
(pg. 
688
-
695
)
23
Mionnet
 
C
Sanos
 
SL
Mondor
 
I
, et al. 
High endothelial venules as traffic control points maintaining lymphocyte population homeostasis in lymph nodes.
Blood
2011
, vol. 
118
 
23
(pg. 
6115
-
6122
)
24
Onder
 
L
Danuser
 
R
Scandella
 
E
, et al. 
Endothelial cell-specific lymphotoxin-β receptor signaling is critical for lymph node and high endothelial venule formation.
J Exp Med
2013
, vol. 
210
 
3
(pg. 
465
-
473
)
25
Kamel-Reid
 
S
Letarte
 
M
Sirard
 
C
, et al. 
A model of human acute lymphoblastic leukemia in immune-deficient SCID mice.
Science
1989
, vol. 
246
 
4937
(pg. 
1597
-
1600
)
26
Park
 
C
Hwang
 
IY
Sinha
 
RK
Kamenyeva
 
O
Davis
 
MD
Kehrl
 
JH
Lymph node B lymphocyte trafficking is constrained by anatomy and highly dependent upon chemoattractant desensitization.
Blood
2012
, vol. 
119
 
4
(pg. 
978
-
989
)
27
Uchimura
 
K
Gauguet
 
JM
Singer
 
MS
, et al. 
A major class of L-selectin ligands is eliminated in mice deficient in two sulfotransferases expressed in high endothelial venules.
Nat Immunol
2005
, vol. 
6
 
11
(pg. 
1105
-
1113
)
28
Warnock
 
RA
Askari
 
S
Butcher
 
EC
von Andrian
 
UH
Molecular mechanisms of lymphocyte homing to peripheral lymph nodes.
J Exp Med
1998
, vol. 
187
 
2
(pg. 
205
-
216
)
29
Spertini
 
O
Freedman
 
AS
Belvin
 
MP
Penta
 
AC
Griffin
 
JD
Tedder
 
TF
Regulation of leukocyte adhesion molecule-1 (TQ1, Leu-8) expression and shedding by normal and malignant cells.
Leukemia
1991
, vol. 
5
 
4
(pg. 
300
-
308
)
30
Drillenburg
 
P
Pals
 
ST
Cell adhesion receptors in lymphoma dissemination.
Blood
2000
, vol. 
95
 
6
(pg. 
1900
-
1910
)
31
Brown
 
JR
Byrd
 
JC
Coutre
 
SE
, et al. 
Idelalisib, an inhibitor of phosphatidylinositol 3-kinase p110δ, for relapsed/refractory chronic lymphocytic leukemia.
Blood
2014
, vol. 
123
 
22
(pg. 
3390
-
3397
)
32
Gauguet
 
JM
Rosen
 
SD
Marth
 
JD
von Andrian
 
UH
Core 2 branching beta1,6-N-acetylglucosaminyltransferase and high endothelial cell N-acetylglucosamine-6-sulfotransferase exert differential control over B- and T-lymphocyte homing to peripheral lymph nodes.
Blood
2004
, vol. 
104
 
13
(pg. 
4104
-
4112
)
33
Simonetti
 
G
Bertilaccio
 
MT
Ghia
 
P
Klein
 
U
Mouse models in the study of chronic lymphocytic leukemia pathogenesis and therapy.
Blood
2014
, vol. 
124
 
7
(pg. 
1010
-
1019
)
34
Lacorre
 
DA
Baekkevold
 
ES
Garrido
 
I
, et al. 
Plasticity of endothelial cells: rapid dedifferentiation of freshly isolated high endothelial venule endothelial cells outside the lymphoid tissue microenvironment.
Blood
2004
, vol. 
103
 
11
(pg. 
4164
-
4172
)
35
Capitani
 
N
Patrussi
 
L
Trentin
 
L
, et al. 
S1P1 expression is controlled by the pro-oxidant activity of p66Shc and is impaired in B-CLL patients with unfavorable prognosis.
Blood
2012
, vol. 
120
 
22
(pg. 
4391
-
4399
)
36
Till
 
KJ
Pettitt
 
AR
Slupsky
 
JR
Expression of functional sphingosine-1 phosphate receptor-1 is reduced by B cell receptor signaling and increased by inhibition of PI3 kinase δ but not SYK or BTK in chronic lymphocytic leukemia cells.
J Immunol
2015
, vol. 
194
 
5
(pg. 
2439
-
2446
)
37
Fiorcari
 
S
Brown
 
WS
McIntyre
 
BW
, et al. 
The PI3-kinase delta inhibitor idelalisib (GS-1101) targets integrin-mediated adhesion of chronic lymphocytic leukemia (CLL) cell to endothelial and marrow stromal cells.
PLoS One
2013
, vol. 
8
 
12
pg. 
e83830
 

Author notes

E.B. and C.L. contributed equally to this study.

Sign in via your Institution