scholarly journals Exosomes Derived from Cancer Associated Fibroblasts Elicit Survival and Drug Resistance of Primary Lymphoma Cells

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4128-4128
Author(s):  
Shunsuke Kunou ◽  
Kazuyuki Shimada ◽  
Tomoya Hikita ◽  
Tomohiro Aoki ◽  
Akihiko Sakamoto ◽  
...  

Abstract Background Cancer associated fibroblasts (CAFs) are one of the major components constituting the tumor microenvironment and known to be deeply involved in the growth and metastasis of solid tumors. We previously reported that CAFs supported survival of primary lymphoma cells via the increased glycolytic metabolism (Aoki et al. Oncotarget 2017). Recent findings indicate that exosomes secreted from tumor cells play an important role of the survival and metastasis. Although the information about exosomes secreted from tumor cells has been accumulated, the role of them secreted from cells constituting the tumor microenvironment has been largely unexplored. Purpose To uncover the role of exosomes in lymphoma microenvironment, we investigated the function of them derived from CAFs those were isolated from primary lymphoma samples. Methods CAFs were successfully isolated from primary lymph node samples of various types of non-Hodgkin lymphoma including follicular lymphoma, diffuse large B-cell lymphoma, angioimmunoblastic T-cell lymphoma, mantle cell lymphoma, and T lymphoblastic lymphoma (N=20), and subsequently exosomes secreted from the representative 4 CAFs were obtained by a standard procedure using ultracentrifugation. Exosomes were confirmed by immunoblotting, electron microscope, and the nano tracking analysis. The functional role of CAFs and exosomes in an interaction between lymphoma cells and its microenvironment were investigated. Written informed consent for the experimental use of patient lymph node samples was obtained, and all experimental procedures were approved by the institutional review board of Nagoya University Hospital. Results We isolated CAFs from different types of non-Hodgkin lymphoma samples, and studied whether the survival of patient lymphoma cells could be supported in co-culture with CAFs. The survival of cells differed depending on CAFs indicating the diversity of the ability of CAFs to support lymphoma cells. Then we analyzed cellular glycolysis and ATP production of lymphoma cells in co-culture with CAFs. As expected, the increase of glycolysis and production of ATP differed among 4 CAFs probably due to the extent of the Warburg effect. Next, we investigated whether CAFs secreted extracellular vesicles into culture supernatant. We found that vesicles with CD9 positive and matched exosomes in size were accumulated in culture media (Figure A, B), and the amount of released exosomes differed among CAFs in line with the ability to support the survival of lymphoma cells. Exosomes displayed survival support of lymphoma cells in a dose-dependent manner(Figure C), and cellular glycolysis and ATP production were increased in the presence of exosomes as well as CAFs, which indicated that the ability of CAFs to support lymphoma cells was, at least in part, elicited by exosomes. While exosomes secreted from CAFs with the strong ability to support lymphoma cells displayed the support of the survival, the survival of lymphoma cells was not observed in the presence of exosomes derived from CAFs with the weak support of lymphoma cells even in higher dose of exosomes, indicating that not only the quantity but also the quality of exosomes could be a determinant of the survival effect. Next, we focused on exosomes secretion-related proteins of nSMase2 and Rab27b to uncover the underlying mechanism of secretion of exosomes. NSMase2 required for exosomes formation and Rab27b involved in the migration of multivesicular body from the pericule to the membrane were strongly expressed in CAFs with higher ability to support lymphoma cells. Using nSMase2- and Rab27b- specific siRNA, the amount of exosomes secretion was reduced resulting in the decreased survival support. Finally, we studied the presence of CAFs or exosomes associated with the drug resistance. As expected, primary lymphoma cells demonstrated resistance to gemcitabine and cytarabine in the presence of CAFs or exosomes, and the resistance was restored in the presence of CAFs transduced with Rab27b-specific siRNA. Conclusion Our results suggest that CAFs and exosomes secreted from them are involved in the survival and drug resistance of patient lymphoma cells and play a pivotal role in the microenvironment of non-Hodgkin lymphoma. Exosomes would be a novel attractive therapeutic target. Disclosures Kiyoi: Sanofi K.K.: Research Funding; Kyowa Hakko Kirin Co., Ltd.: Research Funding; Otsuka Pharmaceutical Co., Ltd.: Research Funding; Nippon Shinyaku Co., Ltd.: Research Funding; Celgene Corporation: Research Funding; Zenyaku Kogyo Co., Ltd.: Research Funding; Eisai Co., Ltd.: Research Funding; FUJIFILM Corporation: Research Funding; Chugai Pharmaceutical Co., Ltd.: Research Funding; Takeda Pharmaceutical Co., Ltd.: Research Funding; Phizer Japan Inc.: Research Funding; Novartis Pharma K.K.: Research Funding; Astellas Pharma Inc.: Research Funding; Sumitomo Dainippon Pharma Co., Ltd.: Research Funding; Bristol-Myers Squibb: Honoraria.

Blood ◽  
2010 ◽  
Vol 116 (24) ◽  
pp. 5228-5236 ◽  
Author(s):  
Tint Lwin ◽  
Jianhong Lin ◽  
Yong Sung Choi ◽  
Xinwei Zhang ◽  
Lynn C. Moscinski ◽  
...  

AbstractFollicular dendritic cells (FDCs), an essential component of the lymph node microenvironment, regulate and support B-lymphocyte differentiation, survival, and lymphoma progression. Here, we demonstrate that adhesion of mantle cell lymphoma and other non-Hodgkin lymphoma cells to FDCs reduces cell apoptosis and is associated with decreased levels of the proapoptotic protein, Bim. Bim down-regulation is posttranscriptionally regulated via up-regulation of microRNA-181a (miR-181a). miR-181a overexpression decreases, whereas miR-181a inhibition increases Bim levels by directly targeting Bim. Furthermore, we found that cell adhesion–up-regulated miR-181a contributes to FDC-mediated cell survival through Bim down-regulation, implicating miR-181a as an upstream effector of the Bim-apoptosis signaling pathway. miR-181a inhibition and Bim upregulation significantly suppressed FDC-mediated protection against apoptosis in lymphoma cell lines and primary lymphoma cells. Thus, FDCs protect B-cell lymphoma cells against apoptosis, in part through activation of a miR-181a–dependent mechanism involving down-regulation of Bim expression. We demonstrate, for the first time, that cell-cell contact controls tumor cell survival and apoptosis via microRNA in mantle cell and other non-Hodgkin lymphomas. Regulation of microRNAs by B-cell–FDC interaction may support B-cell survival, representing a novel molecular mechanism for cell adhesion–mediated drug resistance and a potential therapeutic target in B-cell lymphomas.


Cancers ◽  
2020 ◽  
Vol 12 (2) ◽  
pp. 396
Author(s):  
Estíbaliz Tamayo-Orbegozo ◽  
Laura Amo ◽  
Javier Díez-García ◽  
Elena Amutio ◽  
Marta Riñón ◽  
...  

Mature B-cell non-Hodgkin lymphoma (B-NHL) constitutes a group of heterogeneous malignant lymphoproliferative diseases ranging from indolent to highly aggressive forms. Although the survival after chemo-immunotherapy treatment of mature B-NHL has increased over the last years, many patients relapse or remain refractory due to drug resistance, presenting an unfavorable prognosis. Hence, there is an urgent need to identify new prognostic markers and therapeutic targets. Podocalyxin (PODXL), a sialomucin overexpressed in a variety of tumor cell types and associated with their aggressiveness, has been implicated in multiple aspects of cancer progression, although its participation in hematological malignancies remains unexplored. New evidence points to a role for PODXL in mature B-NHL cell proliferation, survival, migration, drug resistance, and metabolic reprogramming, as well as enhanced levels of PODXL in mature B-NHL. Here, we review the current knowledge on the contribution of PODXL to tumorigenesis, highlighting and discussing its role in mature B-NHL progression.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 5087-5087 ◽  
Author(s):  
Takashi Tokunaga ◽  
Akihiro Tomita ◽  
Kazuyuki Shimada ◽  
Junji Hiraga ◽  
Takumi Sugimoto ◽  
...  

Abstract Abstract 5087 Background Rituximab is an anti-CD20 chimeric-monoclonal antibody, and its effectiveness for treatment of CD20-positive B-cell lymphomas has been proven over the past 10 years. Although rituximab is now a key molecular targeting drug for CD20-positive lymphomas, some patients with rituximab resistance have emerged. We previously reported that the CD20-protein-negative phenotypic change after using rituximab is one of the critical mechanisms in rituximab resistance (Hiraga J, Tomita A, et al., Blood, 2009., Sugimoto T, Tomita A, et al., Biochem Biophys Res Commun, 2009.). Recently, we have recognized that some newly-diagnosed B-cell lymphomas show CD20-protein-positive in immunohistochemistry (IHC) but -negative in flow cytometry (FCM) analyses. For these patients, so far, neither the molecular mechanisms of CD20 IHC(+)/FCM(−) phenotype, nor the relationship between this phenotype and rituximab resistance are clear. Thus, the clinical significance of introducing rituximab therapy for these patients must be elucidated. Aims Analyses of the molecular backgrounds of CD20 IHC(+)/FCM(−) phenotype in primary B-lymphoma cells, and confirmation of the effectiveness of rituximab therapy for the patients who show CD20 IHC(+)/FCM(−) phenotype. Results Primary B-cell lymphoma (diffuse large B-cell (DLBCL), follicular, MALT, mantle cell, and Burkitt) tissues and cells were analyzed by IHC and FCM. Four newly-diagnosed B-cell lymphoma patients showed IHC CD79(+)/CD20(+) and FCM CD19(+)/CD20(−) phenotype using anti-CD20 antibodies L26 for IHC and B1 for FCM, and all were diagnosed as DLBCL. Chromosomal analysis showed complex karyotypes in 3 out of 3 patients analyzed, and no shared abnormalities were confirmed. Primary lymphoma cells from 3 patients were available for further molecular analyses, and the genomic DNA, the total RNA, and the protein from whole cell lysate were obtained from these lymphoma cells. DNA sequencing analysis indicated no significant genetic mutations on the coding sequences (CDS) of MS4A1 (CD20) gene. Semi-quantitative and quantitative RT-PCR indicated that CD20 mRNA expression was almost normal in 2 patients and ≂~f10 times lower in 1 patient compared to the positive control B-lymphoma/leukemia cells. Almost the same expression tendency with RT-PCR was confirmed in immunoblot analysis using whole cell lysate and the two different anti-CD20 antibodies. The molecular weight of the CD20 protein in immunoblotting corresponded to the wild type in these patients. Rituximab binding assay in vitro was performed using primary lymphoma cells from a patient and the fluorescent-labeled rituximab (Alexa488-rituximab). Interestingly, rituximab binding on the surface of the CD19 positive lymphoma cells was confirmed in vitro. Rituximab containing combination chemotherapy was performed, resulting in complete response in all 4 cases after completing 4 to 8 courses. Conclusions and Discussion CD20 IHC(+)/FCM(−) phenotype was confirmed in newly-diagnosed DLBCL patients. Significant abnormalities in CD20 protein and mRNA expression in immunoblotting and RT-PCR were not confirmed, and genetic mutations on CDS of MS4A1 gene, resulting in the conformation change of CD20 protein, were not detected. The possibility of abnormal post-translational modification or aberrant localization of CD20 protein, leading to interference with antibody binding, can not be excluded. Rituximab binding with CD19-positive primary lymphoma cells was confirmed in a patient, suggesting that CD20 IHC(+)/FCM(-) phenotype does not directly indicate the ineffectiveness of rituximab for these cells. Further investigations, performing in vitro CDC and ADCC assay using primary lymphoma cells, are still warranted to show rituximab effectiveness and sensitivity to those cells. Disclosures: Kinoshita: Zenyaku Kogyo Co.: Research Funding; Chugai Pharmaceutical Co., Ltd.: Research Funding. Naoe:Zenyaku Kogyo Co.: Research Funding; Chugai Pharmaceutical Co., Ltd.: Research Funding.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3692-3692
Author(s):  
Julie M Vose ◽  
Fausto R. Loberiza ◽  
R. Gregory Bociek ◽  
Philip Bierman ◽  
James O. Armitage

Abstract Abstract 3692 Introduction: Lenalidomide and ofatumumab have demonstrated clinical activity as single agents in a variety of types of non-Hodgkin lymphoma (NHL). This trial is a phase I/II trial combining these two agents for treatment of patients with relapsed and refractory B-cell NHL. Methods: Patients with relapsed and refractory B-cell NHL of any histology were enrolled on a phase I/II trial combining lenalidomide and ofatumumab. Nine patients were on the phase I part of the trial and received a fixed dose of ofatumumab 1000 mg weekly × 8 doses along with lenalidomide 15 mg (N=3), or10 mg (N=6) for 21/28 days until the time of progression. The phase II portion of the study has 28 patients on the study with adequate follow-up at the time of analysis. The phase II doses were ofatumumab 1000 mg weekly × 8 along with lenalidomide 10 mg on 21/28 days. The lenalidomide was dose adjusted according to standard dose reduction criteria. All patients were on either a daily aspirin or other anticoagulation for thrombosis (DVT) prophylaxis. Results: Thirty seven evaluable patients had adequate follow-up at the time of the analysis. The patients had a median age of 65 years (range 36–81), 76% were male, and 89% have an ECOG performance status of 0–1. The majority of patients had a relapsed indolent lymphoma with 12/37 (32%) follicular lymphoma (FL), 6/37 (16%) chronic lymphocytic leukemia (CLL)/small lymphocytic lymphoma (SLL), 7/37 (19%) mantle cell lymphoma (MCL), one unclassifiable indolent lymphoma (3%), and 11/37 (30%) diffuse large B-cell lymphoma (DLBLC). The median duration of follow-up of surviving patients was 13 months (range 4–24). The complete response (CR) rate was 2/37 (5%) (one each FL and DLBCL) and the partial response (PR) rate was 13/37 (35%) for an overall response rate (ORR) of 15/37 (40%). The 1 year progression-free survival (PFS) was 41% (95% CI; 23–58) and the 1-year overall survival (OS) was 68% (95% CI; 49–82). In an analysis of response by patient variables, those significant included the patients with an FL histology (ORR 83%) vs. DLBCL (ORR 18%) or other(SLL, MCL, unclassifiable) (ORR 21%) (p= 0.001) and lactic dehydrogenase (LDH) normal (ORR 56%) vs. elevated (ORR 14%) (p= 0.01). In an analysis of variables for PFS, the variables with significance include diagnosis of FL (1-year PFS 67%) vs. DLBCL (9%) and SLL, MCL, unclassifiable (45%) (p=0.002), LDH normal (1-year PFS 55%) vs. elevated LDH (1-year PFS 19%), and number of prior chemotherapies 1–2 (1-year PFS 58%) vs. > 3 (1-year PFS 19%). Higher grade toxicities included grade 4 neutropenia in 9/37 (24%), one each of grade 4 bacteremia, one grade 4 DVT, stroke, and acute renal failure. Conclusions: The combination of lenalidomide and ofatumumab was well tolerated by most patients. The patients with indolent NHL had a high response rate of 83% and a 1-year PFS of 67%. Disclosures: Vose: Glaxo Smith Kline: Research Funding; Celgene: Research Funding. Off Label Use: Lenalidamide and Ofatumumab will be discussed for use in indolent and aggressive non-Hodgkin lymphoma.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1279-1279
Author(s):  
Matthew J. Matasar ◽  
Coral L. Atoria ◽  
Elena B. Elkin ◽  
Chadi Nabhan

Abstract Background: The introduction of rituximab has improved outcomes in B-cell non-Hodgkin lymphoma (BCL) across all histologies. Extended use of rituximab, or maintenance rituximab, improves progression-free survival in follicular lymphoma (FL) patients who achieve a response to induction rituximab with or without chemotherapy, but there is no evidence of an overall survival benefit. There is currently little evidence to support extended use of rituximab in other histologic subgroups, and older patients in particular may be at risk of adverse events. Our objective was to characterize patterns and predictors of extended rituximab therapy in a population-based cohort of older BCL patients in the United States. Methods: In the Surveillance, Epidemiology and End Results (SEER)-Medicare dataset,we identified patients 66 years and older diagnosed with BCL in 2000-2010. Histology was classified as diffuse large B-cell lymphoma (DLBCL), Burkitt lymphoma (BL), FL, mantle cell lymphoma (MCL), other indolent BCL, and BCL not otherwise specified (NOS). We identified Medicare claims for rituximab starting at any point following diagnosis. Extended rituximab therapy was defined as a duration of greater than 7 months with no gap in rituximab claims greater than 6 months. Demographic and clinical characteristics associated with extended rituximab were evaluated in multivariable logistic regression. Results: There were 24,232 BCL patients who received rituximab during the study period. The cohort was predominantly white (91%), half were men, 15% had a Charlson comorbidity score ≥2, and 12% were 85 years or older. DLBCL was the most common histology (44%), followed by FL (21%), other indolent BCL (17%), BCL-NOS (13%), MCL (6%), and BL (1%). Overall, most patients (85%) received rituximab for ≤7 months, but duration varied by histology (Table 1). More than a quarter of FL patients had extended therapy, including 7% who had rituximab for more than 24 months. Among patients with other histologies, receipt of extended therapy varied from 20% (other indolent BCL) to 8% (BL). Compared with FL patients and controlling for demographic and disease characteristics, patients with other histologies were less likely to receive extended rituximab therapy (p<0.0001). Adjusted odds ratios were 0.91 (95% CI 0.78-1.05) for MCL, 0.83 (0.75-0.91) for other indolent BCL, 0.67 (0.60-0.75) for BCL-NOS, 0.32 (0.29-0.36) for DLBCL, and 0.28 (0.15-0.53) for BL. However, 75% of patients who had extended rituximab, and 63% of those who had rituximab >24 months, were of non-FL histology. Controlling for histology and other characteristics, extended rituximab therapy was more likely among women (adjusted OR 1.09, 95% CI 1.01-1.18), and less likely among unmarried patients (0.92, 0.85-0.99) and those in rural areas (0.84, 0.75-0.94). There was significant regional variation (p<0.0001), with patients in the West (adjusted OR 0.86, 95% CI 0.79-0.95), and Midwest (0.75, 0.66-0.86) less likely to receive extended rituximab than those in the Northeast. There was no significant relationship between extended therapy and age, race, or comorbidity. Conclusions: While FL patients were more likely than others to receive extended rituximab, the majority of patients receiving extended rituximab had other diagnoses across the entire spectrum of B-cell lymphoma, for which extended rituximab is neither indicated nor supported by guidelines or prospective data. After controlling for histology, several demographic characteristics significantly influenced the duration of therapy. Extended use of rituximab – particularly in patients for whom it is not clearly indicated – may have important implications for clinical outcomes, toxicity, and costs. Table 1 Duration of rituximab use across B-cell lymphoma histologic subgroups Histology Duration of Rituximab N ≤7 mos >7-24 mos >24 mos DLBCL 10,567 91% 7% 2% FL 5,001 76% 17% 7% BL 127 92% 6% 2% MCL 1,339 79% 16% 5% Other indolent 4,095 80% 15% 5% BCL NOS 3,103 83% 13% 4% Total 24,232 80% 15% 5% Disclosures Matasar: Merck: Research Funding; GlaxoSmithKline: Research Funding; Genentech: Honoraria; Spectrum: Honoraria. Nabhan:Celgene: Honoraria, Research Funding.


Oncogene ◽  
2021 ◽  
Author(s):  
Shunsuke Kunou ◽  
Kazuyuki Shimada ◽  
Mika Takai ◽  
Akihiko Sakamoto ◽  
Tomohiro Aoki ◽  
...  

AbstractThe tumor microenvironment is deeply involved in the process of tumor growth and development. In this study, we focused on cancer-associated fibroblasts (CAFs) and their derived exosomes on the lymphoma microenvironment to uncover their clinical significance. CAFs were established from primary lymphoma samples, and exosomes secreted from CAFs were obtained by standard procedures. We then investigated the roles of CAFs and their derived exosomes in the survival and drug resistance of lymphoma cells. CAFs supported the survival of lymphoma cells through increased glycolysis, and the extent differed among CAFs. Exosomes were identified as a major component of the extracellular vesicles from CAFs, and they also supported the survival of lymphoma cells. The suppression of RAB27B, which is involved in the secretion of exosomes, using a specific siRNA resulted in reduced exosome secretion and decreased survival of lymphoma cells. Moreover, anti-pyrimidine drug resistance was induced in the presence of exosomes through the suppression of the pyrimidine transporter, equilibrative nucleoside transporter 2 (ENT2), and the suppression of ENT2 was significant in in vivo experiments and clinical samples. RNA sequencing analysis of miRNAs in exosomes identified miR-4717-5p as one of the most abundant miRNAs in the exosome, which suppressed the expression of ENT2 and induced anti-pyrimidine drug resistance in vitro. Our results suggest that exosomes including miR-4717-5p secreted from CAFs play a pivotal role in the lymphoma microenvironment, indicating that they are a promising therapeutic target.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2874-2874
Author(s):  
Akihiro Tomita ◽  
Takashi Tokunaga ◽  
Chisako Iriyama ◽  
Kazuyuki Shimada ◽  
Junji Hiraga ◽  
...  

Abstract Abstract 2874 Background: Rituximab is an anti-CD20 mouse/human chimeric monoclonal antibody. Recent reports indicate that combination chemotherapy with rituximab for CD20(+) B-cell lymphoma has improved the clinical outcomes compared to conventional chemotherapy without rituximab. However, patients showing rituximab resistance have been also recognized. We previously reported observation of relapse (RD)/progression (PD) with CD20(-) phenotype after using rituximab, and CD20(-) phenotype is closely related to ritxuximab resistance (Hiraga J, et al., Blood, 2009., Sugimoto T, et al., Biochem Biophys Res Commun, 2009., Tomita A, et al., Int J Hematol, 2007.). We concluded that aberrant down-regulation of MS4A1 (CD20) gene expression was one of the critical mechanisms of CD20(-) phenotypic change. Moreover, all those patients died of disease progression within one year after CD20(-) change, indicating the tendency for a relatively poor prognosis. Since such patients occur less frequently, more patients are definitely needed to confirm the clinical features and molecular basis. Aims: To confirm the clinical features of patients who show CD20-protein-negative phenotypic change after using rituximab. Analyses of the molecular backgrounds of CD20(-) phenotypes will be conducted using CD20(-)-transformed primary lymphoma cells. Results: CD20(+) B-cell lymphoma patients (DLBCL, FL, MALT, MCL, CLL/SLL, and Burkitt) treated with rituximab containing chemotherapy, were used for the analyses. CD20 protein expression was confirmed by immunohistchemistry (IHC) and flow cytometry (FCM) using primary lymphoma cells at the time points of the initial diagnosis and the RD/PD. All patients analyzed showed CD20 IHC(+)/FCM(+) phenotype at the initial diagnosis, and the CD20(-) change was confirmed by IHC(-)/FCM(-) at RD/PD period. Ten patients showed CD20(-) phenotypic change after using rituximab. Pathological diagnosis of those patients was DLBCL (8 out of 10 cases; 8/10) and FL (2/10) at the initial diagnosis, and DLBCL (10/10) at CD20(-) change in RD/PD period. TdT was negative by IHC in 6 out of 6 analyzed cases. Bone marrow (BM) invasion at diagnosis was confirmed in 9/10, and extranodal involvement (BM, CNS, skin, and peripheral blood) at CD20(-) RD/PD period was confirmed in all patients (10/10). Leukemic transformation at the terminal stage was seen in 6/10 patients, and LDH elevation seems to reflect their disease status. All 10 patients died of disease progression within 1 year after the diagnosis of CD20(-) phenotypic change in spite of salvage chemotherapy. Molecular backgrounds were analyzed in patients whose primary lymphoma cells were available for molecular analyses. Quantitative RT-PCR indicated that the CD20 mRNA expression level was significantly lower than that of CD20 positive controls in all analyzed cases (5/5). Almost the same result was obtained by immunoblot analysis using anti-CD20 C-terminus antibody. Genetic mutations on the coding sequence (CDS) of MS4A1 gene were suggested in 2 out of 7 cases, but the frequency of the mutated clone was less than 20%. CD79b ITAM mutation was confirmed in 1 out of 5 cases. In vitro rituximab binding assay using fluorescent-labeled-rituximab and primary lymphoma cells indicated that the rituximab binding was significantly lower on CD20(-) cells compared to the control cells. Conclusions and Discussion: All the patients showing CD20(-) phenotypic change were diagnosed as DLBCL, and extranodal infiltration was confirmed in all cases. Leukemic transformation was relatively common at the terminal stage, and LDH elevation seemed to reflect the disease progression. CD20 mRNA expression level was generally lower than that of positive control cells, and clinically significant MS4A1 gene mutations were not confirmed. All the patients died within 1 year after diagnosis of CD20(-) transformation, suggesting that the CD20(-) change may closely correlate with the poor prognosis. From these findings, the phenotypic change after using rituximab may be considered as a particular clinicopathologic group such as “post-rituximab extranodal CD20-negative lymphoma.” Further accumulation of patients is warranted. Disclosures: Kinoshita: Zenyaku Kogyo Co.: Research Funding; Chugai Pharmaceutical Co., Ltd.: Research Funding. Naoe:Zenyaku Kogyo Co.: Research Funding; Chugai Pharmaceutical Co., Ltd.: Research Funding.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3929-3929
Author(s):  
Liang Zhang ◽  
Jing Yang ◽  
Jianfei Qian ◽  
Sattva Neelapu ◽  
Larry Kwak ◽  
...  

Abstract Abstract 3929 Mantle cell lymphoma (MCL) is a type of aggressive B-cell non-Hodgkin lymphoma characterized by frequent resistance to conventional chemotherapy. Currently, there is no standard of care for the treatment of MCL, and patient prognosis is poor. It is well known that the tumor microenvironment plays an important role in tumor cell growth and resistance to chemotherapy. However, little is known about the microenvironment and its influence in MCL. In this study we investigated the role of IL-6 and its signaling in the growth, survival, and development of drug resistance in MCL, as IL-6 is an important cytokine for B cells and multiple myeloma. We found that the membrane IL-6 receptor gp130 is generally expressed in established MCL cell lines and in primary lymphoma cells from patients, which can be upregulated by stress such as serum starvation or low-dose chemotherapy drug treatment. Some but all not MCL cells also secrete IL-6 and/or IL-6 soluble receptor gp80. Although IL-6 and its signaling pathway do not affect MCL growth in vitro, they play an important role in MCL survival and resistance to chemotherapy drugs. Neutralizing IL-6 and/or blocking IL-6 receptors in IL-6- or gp80-secreting MCL cells increased their sensitivity to chemotherapy drug- and serum starvation-induced apoptosis. For MCL cells that do not secrete IL-6 or gp80, low doses of exogenous IL-6 or gp80 protected them from chemotherapy drug-induced apoptosis. Because T cells, macrophages, and bone marrow stromal cells (BMSCs) secrete IL-6 and/or gp80, coculture of MCL cells with peripheral blood mononuclear cells or BMSCs protected MCL cells from chemotherapy drug-induced growth inhibition and apoptosis, which can be abrogated by anti-IL-6, anti-gp80, and/or anti-gp130 antibodies. Knocking down gp80 in gp80high MCL cells rendered the cells more sensitive to chemotherapy drug-induced apoptosis, even in the presence of exogenous IL-6. Overexpression of gp80 in gp80low IL-6+ MCL cells provided protection of MCL cells from chemotherapy drug-induced apoptosis. Next, Jak2 or STAT3 inhibitors completely abrogated IL-6-mediated protection of MCL cells from apoptosis, whereas PI3K or MEK inhibitors partially abrogated IL-6-mediated protection of MCL cells from apoptosis. Furthermore, gp80-overexpressing, gp80low IL-6+ MCL cells grew faster than vector control or parental cells in the SCID mouse model, and immunohistochemistry staining showed strong surface gp80 and nuclear phosphorylated STAT3 in gp80-overexpressing MCL tumor cells. Thus, these results clearly show that IL-6 and gp80, derived from MCL cells themselves or from cells in the microenvironment, play a pivotal role in MCL cell survival and drug resistance. Although IL-6 activates Jak2/STAT3, PI3K/AKT, and MEK/Erk signaling pathways, STAT3 signaling may play an important role in mediating IL-6-induced protection of MCL against chemotherapy drug-induced apoptosis. This study suggests that targeting IL-6 and its signaling pathway may improve the efficacy of chemotherapy in MCL patients. Disclosures: Wang: Celgene: Honoraria, Research Funding; Onyx: Research Funding; Millenium: Research Funding; Novartis: Research Funding.


2021 ◽  
pp. 20210576
Author(s):  
Elizabeth H Phillips ◽  
Rohan Iype ◽  
Andrew Wirth

FDG-PET scanning has a central role in lymphoma staging and response assessment. There is a growing body of evidence that PET response assessment during and after initial systemic therapy can provide useful prognostic information, and PET response has an evolving role in guiding patient care. This review provides a perspective on the role of PET response assessment for individualised management of patients with the most common aggressive lymphomas, Hodgkin lymphoma and diffuse large B-cell lymphoma.


Sign in / Sign up

Export Citation Format

Share Document