T-cell activation induced by anti-CD3 × anti-B-cell lymphoma monoclonal antibody is enhanced by pretreatment of lymphoma cells with soluble CD40 ligand

1997 ◽  
Vol 45 (3-4) ◽  
pp. 174-179 ◽  
Author(s):  
James E. Wooldridge ◽  
Chris E. Dahle ◽  
George J. Weiner
2012 ◽  
Vol 33 (1) ◽  
pp. 22-29 ◽  
Author(s):  
David Eric Ouedraogo ◽  
Alain Makinson ◽  
Nils Kuster ◽  
Nicolas Nagot ◽  
Pierre-Alain Rubbo ◽  
...  

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3512-3512 ◽  
Author(s):  
Heather A Huet ◽  
Casey Judge ◽  
R. Anthony Barnitz ◽  
Ryan Boomer ◽  
Kathleen McGinness ◽  
...  

Abstract The Antibody-Coupled T-cell Receptor (ACTR) platform is a universal engineered T cell therapy developed to mediate anti-tumor activity in combination with tumor-targeting antibodies. The ACTR construct is composed of the extracellular domain of CD16 (FCGR3A; high-affinity V158 variant) linked to CD3ζ signaling and 41BB co-stimulatory domains. ACTR-expressing T cells recognize antibodies bound to antigen on the surface of target cells, and this receptor engagement results in T cell activation, proliferation, and cytotoxic attack of target cells. Previous preclinical research has demonstrated that ACTR T cells elicit tumor cell cytotoxicity in combination with rituximab, trastuzumab, and hu14.18K322A against CD20, HER2, and GD2 expressing tumor cell lines, respectively (Kudo et al., Cancer Res 2014; 74:93-103). The safety of ACTR T cells in combination with rituximab for subjects with B-cell malignancies is currently being explored in a pilot phase 1 clinical study (ATTCK-20) sponsored by National University Hospital, Singapore. Electroporation of mRNA is being used to deliver the ACTR transgene to study subjects' T-cells, creating a transiently active form of the therapy. In this study, we present non-clinical data supporting the first-in-human clinical trial of ACTR087, the ACTR transgene (CD16V-41BB-CD3ζ) delivered by γ-retrovirus to human T cells, in combination with rituximab in relapsed refractory B cell lymphoma (NCT02776813). In vitro data demonstrate that in the absence of rituximab or in the presence of non-targeting antibodies, no increase in cytotoxicity is observed, demonstrating that ACTR signaling is target- and antibody-specific. ACTR T cells proliferate in response to rituximab-bound lymphoma cells, but this proliferation is antibody-dependent and self-limiting in the absence of antibody. In vivo efficacy and pharmacokinetic studies further support the dependence on adequate rituximab exposure for ACTR T cell response, suggesting that ACTR T cell activity can be modulated by antibody dose and schedule, which is a potential advantage over other chimeric antigen receptor (CAR) T cell therapies. Additional in vitro studies were conducted to explore the effects of potential interventional antibody therapies used to mitigate adverse events reported for CAR-T therapies in similar patient populations. These include tocilizumab, used to treat cytokine release syndrome (CRS), and intravenous immunoglobulin (IVIG), used to treat hypogammaglobulinemia complicated by recurrent sinopulmonary infections. At therapeutically relevant doses, rituximab was able to compete with substantial excess IVIG to induce a CD20-specific ACTR T cell response. Importantly, IVIG did not induce greater cytokine release from ACTR T cells. Likewise, tocilizumab did not mediate ACTR T cell activation or cytotoxicity of IL-6R+ target cells, consistent with reports that tocilizumab is ADCC-deficient. These data illustrate an important principal of Fc receptor biology: not all IgG1 antibodies mediate ADCC despite binding to CD16, and support the considered use in the presence of ACTR T cells under certain circumstances. Taken together, these non-clinical data demonstrate the specificity and versatility of the ACTR T cell therapeutic approach to target diverse cancer antigens. The Phase 1 study (ATTCK20-2, ClinicalTrial.gov No. NCT02776813) of ACTR087 plus rituximab in subjects with relapsed or refractory CD20-positive B cell lymphoma is a multi-center, single-arm, open-label study evaluating the safety and efficacy of an autologous T-cells, culture expanded and transduced ex vivo with a γ-retrovirus containing an ACTR expression construct (CD16V-41BB-CD3ζ). Subjects who meet eligibility requirements with histologically-confirmed relapsed or refractory CD20+ B-cell lymphoma of one of the following types are eligible for the study: DLBCL, MCL, PMBCL, Gr3b-FL, TH-FL. Following a conditioning regimen with fludarabine and cyclophosphamide, and prior to ACTR087 infusion, subjects will receive rituximab at the standard IV clinical dose, 375 mg/m2. Subjects may receive up to 7 additional cycles of rituximab 375 mg/m2 at 3-week intervals and continuing as long as they show evidence of response. Disclosures Huet: Unum Therapeutics: Employment. Judge:Unum Therapeutics: Employment. Barnitz:Unum Therapeutics: Employment. Boomer:Unum Therapeutics: Employment. McGinness:Unum Therapeutics: Employment. Shin:Unum Therapeutics: Employment. Cheema:Unum Therapeutics: Employment. Whiteman:Unum Therapeutics: Employment. Schultes:Unum Therapeutics: Employment. Ranger:Unum Therapeutics: Employment. Cao:Unum Therapeutics: Employment. Hodge:Unum Therapeutics: Employment. Vasconcelles:Unum Therapeutics: Employment. Ettenberg:Unum Therapeutics: Employment.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4637-4637
Author(s):  
Gerald G. Wulf ◽  
Anita Boehnke ◽  
Bertram Glass ◽  
Lorenz Truemper

Abstract Anti-CD45 mediated cytoreduction is an effective means for T-cell depletion in rodents and humans. In man, the CD45-specific rat monoclonal antibodies YTH24 and YTH54 are IgG2b subclass, exert a predominantly complement-dependent cytolytic activity against normal T-lymphocytes, and have been safely given to patients as part of conditioning therapies for allogeneic stem cell transplantation. The efficacy of such antibodies against human lymphoma is unknown. Therefore, we evaluated the cytolytic activity of YTH24 and YTH54 by complement-dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity (ADCC), as well as by direct apoptotic and antiproliferative effects, against a panel of Hodgkin disease (HD) and non-Hodgkin lymphoma (NHL) cell lines, and against primary specimens. Significant CDC activity (>50% cytolysis) of the antibodies YTH54 and YTH24 was observed against three of five T-cell lymphoma lines, but against only one of nine B-cell lymphoma lines and none of four HD cell lines. The combination of YTH54 and YTH24 induced ADCC in all T-cell lymphoma cell lines and three primary leukemic T-cell lymphoma specimens, but were ineffective in B-cell lymphoma and HD cell lines.There were only minor effects of either antibody or the combination on lymphoma cell apoptosis or cell cycle arrest. In summary, anti-CD45 mediated CDC and ADCC via the antibodies YTH24 and YTH54 are primarily effective against lymphoma cells with T-cell phenotype, and may be an immunotherapeutic tool for the treatment of human T-cell lymphoma.


Blood ◽  
1998 ◽  
Vol 92 (12) ◽  
pp. 4750-4757 ◽  
Author(s):  
Peter T. Daniel ◽  
Arne Kroidl ◽  
Joachim Kopp ◽  
Isrid Sturm ◽  
Gerhard Moldenhauer ◽  
...  

Bispecific antibodies (CD3x19) against the CD3ɛ-chain of the T-cell–receptor/CD3 complex and the CD19 antigen on B cells can target polyclonal, nontumor-specific T cells to B lymphoma cells. This induces T-cell activation, and generation of cytotoxic T cells (CTLs). These polyclonal CTLs, targeted by the CD3x19 bispecific antibodies, can lyse CD19+ B-lymphoma cells. In a xenotransplant model in severe combined immunodeficiency deficient (SCID) mice, we and others observed that CD28 triggering is required for efficient elimination of B-lymphoma cells and cure from the tumor in addition to CD3x19 administration. We also showed that the activation and targeting of CTLs to the target cell by signal one alone, ie, the CD3x19 mab, induces T-cell death by apoptosis. In blocking experiments we showed that this “veto” apoptosis is mediated by the CD95/Fas ligand. Addition of anti-CD28 (signal 2) renders the T cells resistant for veto apoptosis both in vitro and in vivo. We therefore conclude that the role of costimulation in immunotherapy with bispecific antibodies or other T-cell–based immune strategies is not only to facilitate T-cell activation but also to prevent T-cell deletion by apoptosis.


1998 ◽  
Vol 103 (1) ◽  
pp. 270-275 ◽  
Author(s):  
Katharina Clodi ◽  
Zahra Asgary ◽  
Shourong Zhao ◽  
Kay-Oliver Kliche ◽  
Fernando Cabanillas ◽  
...  

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1671-1671
Author(s):  
Harbani Malik ◽  
Ben Buelow ◽  
Brian Avanzino ◽  
Aarti Balasubramani ◽  
Andrew Boudreau ◽  
...  

Abstract Introduction Along with CD20 and CD22, the restricted expression of CD19 to the B-cell lineage makes it an attractive target for the therapeutic treatment of B-cell malignancies. Many monoclonal antibodies and antibody drug conjugates specific to CD19 have been described, including bispecific T-cell redirecting antibodies (T-BsAbs). In addition, anti-CD19 chimeric antigen receptor T-cells (CAR-Ts) have been approved to treat leukemia. To date, toxicity from over-activation of T-cells and large-scale production of CAR-Ts still hinder this approach. Bispecific T-cell engaging antibodies redirecting T cells to CD19 circumvent the latter problem but to date have shown similar T-cell over-activation, as well as significant neurotoxicity. Utilizing TeneoSeek, a next generation sequencing (NGS)-based discovery pipeline that uses in silico analysis of heavy chain only/fixed light chain antibody (HCA/Flic, respectively) sequences to enrich for antigen specific antibodies, we made a high affinity αCD19 HCA and a library of αCD3 Flic antibodies that showed a >2 log range of EC50s for T cell activation in vitro. Of note, the library contained a selectively-activating αCD3 that induced potent T-cell dependent lysis of lymphoma cells (when paired with an αCD19 HCA) with minimal cytokine secretion. To characterize the relative efficacy and potential therapeutic window of this unique molecule, we compared the low-activating (and Fc-containing) CD19 x CD3 to two pan T-cell activating bispecific CD19 x CD3 antibodies (blinatumomab and another developed in-house) in vitro and in vivo for T-cell activation, efficacy in killing lymphoma cells, and toxicity. Methods T-cell activation was measured via flow cytometry (CD69 and CD25 expression) and cytokine ELISA (IL-2, IL-6, IL-10, INF-ɣ, and TNFα) in vitro. Lysis of B-cell tumor cell lines (Raji, Ramos, and Nalm6) was measured via calcein release in vitro. In vivo, NOG mice were engrafted with human peripheral blood mononuclear cells (huPBMC) and human lymphoma cell lines, and the mice treated with weekly injections of T-BsAbs. Tumor burden was evaluated via caliper measurement. Pharmacokinetic (PK) studies were performed in NOG mice using ELISA. Results EC50s for cytotoxicity were in the single-digit nanomolar range for the selective T cell activating T-BsAb and sub-nanomolar for the pan T-cell activating controls. The selective T cell activator showed markedly reduced cytokine release for all cytokines tested compared to the pan T-cell controls even at saturating concentrations. In vivo, established CD19 positive B-cell tumors were cleared in NOG mice in the presence of huPBMC. PK profiles of both molecules generated in-house (selective and pan T-cell activators) were consistent with those of an IgG in mice. No activation of T-cells was observed in vitro or in vivo in the absence of CD19 expressing target cells. Conclusions Both the selectively-activating and the pan T-cell activating control bispecific antibodies killed lymphoma cells in vitro and in vivo in a CD19-dependent manner. While the pan T-cell activating controls showed T-cell activation comparable to other CD3-engaging bispecifics, the selective activator induced significantly reduced cytokine secretion by T-cells and demonstrated a half-life consistent with other IgG antibodies. In summary, our selectively activating CD19 x CD3 T-BsAb shows promise as a lymphoma therapeutic differentiated from current T-cell targeted therapies currently in the clinic and in clinical trials. Disclosures Malik: Teneobio, Inc.: Employment. Buelow:Teneobio Inc.: Employment. Avanzino:Teneobio, Inc.: Employment. Balasubramani:Teneobio, Inc.: Employment. Boudreau:Teneobio, Inc.: Employment. Clarke:Teneobio, Inc.: Employment. Dang:Teneobio, Inc.: Employment. Davison:Teneobio, Inc.: Employment. Force Aldred:Teneobio Inc.: Employment. Harris:Teneobio, Inc.: Employment. Jorgensen:Teneobio, Inc.: Employment. Li:Teneobio, Inc.: Employment. Medlari:Teneobio, Inc.: Employment. Narayan:Teneobio, Inc.: Employment. Ogana:Teneobio, Inc.: Employment. Pham:Teneobio Inc.: Employment. Prabhakar:Teneobio, Inc.: Employment. Rangaswamy:Teneobio, Inc.: Employment. Sankaran:Teneobio, Inc.: Employment. Schellenberger:Teneobio, Inc.: Employment. Ugamraj:Teneobio, Inc.: Employment. Trinklein:Teneobio, Inc.: Employment. Van Schooten:Teneobio, Inc.: Employment.


Blood ◽  
1991 ◽  
Vol 77 (4) ◽  
pp. 809-817 ◽  
Author(s):  
ML Hansmann ◽  
HH Wacker ◽  
J Gralla ◽  
H Lumbeck ◽  
M Kossmahl ◽  
...  

In the search for immunoreagents appropriate for the histopathologic diagnosis of malignant B-cell lymphomas in routinely processed paraffin sections, a new monoclonal antibody, Ki-B5, was generated using a high- grade B-cell lymphoma as the immunogene. Ki-B5 is a mouse IgG1/kappa that recognizes five protein fractions of about 84, 82, 55, 48, and 27 Kd after biosynthetic radiolabeling and immunoprecipitation. Protein fractions with the molecular weights of approximately 84 and 82 Kd were expressed on the cell surface and show that Ki-B5 is probably unrelated to CD45. It was possible through electron microscopy to visualize the membrane-bound portion of Ki-B5. Extensive immunohistologic studies on normal human tissue and various neoplasias demonstrated the high specificity of Ki-B5 to normal human B cells and a minor subgroup of plasma cells. Except for ML-2, which is a myelomonocytic human cell line, Ki-B5 exclusively recognized the B-cell lineage, including EB-3, BALL-1, and NALM-1. All carcinomas, sarcomas, and malignant melanomas tested with Ki-B5 were negative. Although normal granulocytes and monocytes were constantly negative, three of eight myelomonocytic leukemias coreacted with this antibody. Eight of the 57 T-cell lymphomas studied were positive to Ki-B5. Five were classified as lymphoblastic, two represented T8-CLL, and one was classified as immunoblastic T-cell lymphoma. Only 3 of 126 cases of B-cell lymphoma, including rare types not considered in the current classifications, were negative to Ki-B5. Plasmacytomas were also negative, except for one case. Irrespective of the cases of lymphoblastic lymphoma and plasmacytoma, Ki-B5 represents a new monoclonal antibody appropriate for the diagnosis and immunophenotyping of malignant lymphomas in routinely processed paraffin sections.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4832-4832
Author(s):  
Lisa Hicks ◽  
J. Mena ◽  
D. E. Spaner

Abstract Background: B cells express toll-like-receptor-7 (TLR7), a powerful modulator of inate immunity. We hypothesized that the immunogenicity of indolent B cell lymphomas and leukemias could be increased through activation of the TLR7 pathway. This might lead to increased immune clearance of malignant cells. Imiquimod 5% (an imidazoquinolone) has recently been identified as a TLR 7/8 agonist. It is available as a topical formulation and has demonstrated activity against condylomata as well as skin carcinomas. A small number of case reports also suggest activity against cutaneous T cell lymphoma. Case: A 71 year old Caucasian man presented with Rai Stage 0 CLL. For approximately 8 years prior to presentation, he suffered recurrent, erythematous, nodular lesions on his hands, arms and torso. The lesions were removed with liquid nitrogen but recurred. At the time of diagnosis with CLL, several lesions were present. Biopsy demonstrated a diffuse infiltrate of small round lymphocytes without epidermotropism. CD20 staining was positive. Molecular analysis confirmed a monoclonal B cell population consistent with B cell lymphoma. The patient was otherwise asymptomatic and did not warrant conventional therapy for CLL. Based on the safety and efficacy of imidazoquinoline in other skin cancers, he was offered and consented to a trial of this therapy. The drug was applied to the affected area 3x per week as recommended for treatment of genital warts. After 8 weeks, an area of hypopigmentation had formed, however the size of the lesion had not decreased. The frequency of application was increased to 1x per day. Over a 6 week period the lesion gradually disappeared. Six months after stopping the drug, the lesion has not recurred. No side effects were noted. Untreated lymphomatous lesions and peripheral blood lymphocyte counts did not change over the course of treatment. In vitro Data: We hypothesized that Imiquimod might increase the immunogenicity of CLL cells by increasing their surface expression of costimulatory molecules. To investigate this hypothesis, the above patient’s CLL cells were isolated (with his consent) directly through negative selection and then incubated for 48h with an active soluble imidazoquinalone or with a control. Expression of co-stimulatory molecules was determined by flow cytometry pre and post-incubation. CD80, 83, 86 and 54 surface expression were found to be increased significantly by TLR-7 agonists in vitro. Conclusion: We report the first case of Imiquimod activity against a cutaneous B cell lymphoma. We propose that the drug increases co-stimulatory molecule expression by malignant B cells, thereby facilitating cytotoxic T cell activation and killing of CLL cells. We believe that further study of TLR7 agonists is warranted in B cell malignancies.


Sign in / Sign up

Export Citation Format

Share Document