Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
  • American Society of Hematology  (2)
  • 2005-2009  (2)
Type of Medium
Publisher
  • American Society of Hematology  (2)
Language
Years
  • 2005-2009  (2)
Year
Subjects(RVK)
  • 1
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 883-883
    Abstract: Monoclonal antibodies (mAb) directed to a range of cell surface antigens on malignant lymphoid cells are increasingly a key component of the treatment of a whole range of haematological malignancies and remain the focus of intense ongoing clinical investigation. The anti-CD20 mAb rituximab has over the last decade substantially increased clinical responses leading to a marked improvement in outcome for many patients with B cell malignancies. The pursuit of mAb to improve on the efficacy of rituximab is currently undergoing intensive research, with several new candidate mAb now undergoing clinical evaluation. These newer generation anti-CD20 mAb have largely focused on improvements in increasing the affinity of the Fc-FcgR interactions and the recruitment of immune effector cells as these are thought to underlie much of the mAb therapeutic effect. However the immune effector functions of mAb does not explain why certain mAb specificities are more potent than others in inducing tumour cell death. Previously, we have demonstrated that Type II anti-CD20 mAb (Tositumomab) were able to evoke direct cell death via a non-apoptotic mode of death that appeared to be linked with the induction of homotypic adhesion. In this study we have considerably extended these observations using both in vitro cell lines and primary CLL cells. Using time-lapse microscopy and the FRAP technique, we revealed that de novo F-actin synthesis within cell contact area is critical for both homotypic adhesion and cell death induced by both Type II anti-CD20 mAb (Tositumomab) and HLA DR Class II mAb (L243). We have demonstrated that the mode of cell death engaged is rapid, non-apoptotic and non-autophagic as detected by inability to inhibit this form of cell death with pan-caspase inhibitor QVD, overexpressed Bcl2 as well as with siRNA against key autophagy regulators Beclin 1 and ATG-12. Scanning and transmission electon microscopy studies revealed the cytoplasmic nature of cell death involving lysosomes which swell and then disperse their contents into the cytoplasm. The increase in the lysosomal compartment followed by lysosome membrane permeabilisation and subsequent cell death has also been confirmed by flow cytometry and fluorescence microscopy, using lysosome specific probe lysotracker. Furthermore, this cell death has been shown to be dependent on active V-type ATPase but not on chymotrypsinlike or trypsin-like serine proteases. Using time lapse microscopy, we show here for the first time that malignant B cells, undergoing homotypic adhesion, actively communicate via ~ 5 nm wide temporary inter-cytoplasmic bridges. The formation of these channels is accompanied by the exchange of plasma membrane components. Moreover, the extent of plasma membrane swapping correlates with the extent of cell death induced by both anti-CD20 and anti-HLA DR antibodies. This data provide new insights into the potential mechanisms underlying mAb-induced cell death and may guide the rational design of more effective mAb therapy in B cell malignancies.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 725-725
    Abstract: Abstract 725 The addition of the anti-CD20 monoclonal antibody (mAb) rituximab to chemotherapy has substantially improved the clinical outcome of patients with a wide range of B-cell malignancies. Despite this success, many patients are not cured by standard approaches and there is intense investigation into the development of new-generation anti-CD20 mAbs with further improved therapeutic efficacy. Although Fc-FcgR interactions appear to underlie much of the therapeutic success with Rituximab, certain Type II anti-CD20 mAbs, can directly induce programmed cell death (PCD), whereas rituximab-like Type I anti-CD20 mAbs do not (Chan et al. Cancer Res 63: 5480-5489, 2003). We have demonstrated that Type II mAbs are more effective at B-cell depletion in syngeneic human CD20 transgenic mice (Beers et al. Blood 112: 4170-4177, 2008). Recently, we elucidated the mechanism underlying PCD induced by the Type II anti-CD20 mAb Tositumomab, demonstrating a novel non-apoptotic mode of cell death, defined by homotypic adhesion, peripheral relocalization of actin and lysosomal activity (Ivanov et al. J Clin Invest, doi: 10.1172/JCI37884, 2009). Here we confirm that the humanized anti-CD20 mAb GA101 and derivatives harboring non-glycomodified human IgG1 or mouse IgG2a Fc regions are bone fide Type II reagents, lacking the ability to translocate CD20 into lipid rafts or initiate calcium flux. Furthermore, GA101 initiates extensive non-apoptotic cell death in a range of B-lymphoma cell lines in contrast to rituximab (e.g. in Raji cells 48 ± 1.8% versus 13 ± 0.2%, p 〈 0.001 by Student's t-test) quantified using the Annexin V/propidium iodide cell death assay. Inhibitors of actin polymerization (latrunculin B and cytochalasin D) inhibited cell death elicited by GA101 from 45 ± 1.5% to 15 ± 3.1% (p 〈 0.01). The importance of cell to cell contact in this form of antibody induced cell death was confirmed by the addition of low-melting point agarose which physically blocked cell to cell contact and markedly attenuated cell death induced by GA101. The role of lysosomal activity in GA101-induced PCD was assessed using an inhibitor of the lysosomal cysteine protease cathepsin B, which significantly inhibited cell death induced by GA101 from 53 ± 4.3% to 18 ± 1.9%, (p 〈 0.001). To confirm that this mode of death is non-apoptotic, we demonstrated that GA101-induced PCD occurred independently of BCL-2 over-expression and caspase activation. Complement-dependent cytotoxicity (CDC) assays using human serum as a source of complement reveal that GA101 has significantly weaker CDC activity than rituximab, consistent with our previous work on Type II anti-CD20 mAbs (Cragg et al Blood 101: 2738-2743, 2003). Taken together, these findings demonstrate that GA101 is the first humanized anti-CD20 mAb with Type II properties, potently eliciting a novel mode of cell death in B-cell malignancies, which potentially can lead to improved B-cell depletion over rituximab. Furthermore, we are currently investigating the relative ability of GA101 and rituximab to delete B cells in vivo using directly comparable versions of these mAb with human or mouse Fc regions in human CD20 transgenic mice and will present these data. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages