Abstract

Double Hit Lymphomas (DHL) constitute 6-9% of all newly-diagnosed Diffuse Large B-cell Lymphomas (DLBCL) and are defined by MYC translocation in concert with BCL2 (80%) or BCL6 (20%) rearrangement. DHL is characterized by poor upfront response to standard chemoimmunotherapies. The development of more effective treatment for DHL has been limited by the lack of models that accurately recapitulate the biology of this disease. To faithfully model bona fide human DHL, we generated two patient-derived xenografts (PDXs) in NSG mice from patients with DHL who relapsed after or were refractory to R-CHOP. Both PDXs had low/negative expression of surface CD20, in line with previous reports of DHL, and high surface expression of CD52. Importantly, these features recapitulated the original tumor phenotypes. We reasoned that immunologic approaches that target DHLs may help overcome their resistance to cytotoxic chemotherapy. Given the surface expression of CD52 on both PDXs, we treated xenografted mice with the anti-CD52 antibody Alemtuzumab (2x5mg/kg) when >2% tumor was detected in peripheral blood, a time corresponding to marked splenomegaly and ~30% bone marrow involvement. In both models, Alemtuzumab markedly reduced tumor involvement in the peripheral blood and spleen; however, the bone marrow was largely refractory to Alemtuzumab treatment. This is in line with previous results (Pallasch et al. Cell 2014) who also observed bone marrow-specific resistance to Alemtuzumab using a humanized transgenic model of DHL. In the latter model, Alemtuzumab resistance was successfully overcome by high-dose cyclophosphamide (CTX, 100mg/kg), which induced a tumor secretory response that induced macrophage-mediated phagocytosis of tumor cells. Thus, we treated both PDX models with vehicle, CTX 100mg/kg, Alemtuzumab or CTX+Alemtuzumab. While CTX monotherapy was able to partially reduce bone marrow tumor burden, in both PDXs, a single dose of CTX+Alemtuzumab was highly synergistic and eradicated nearly all bone marrow disease (Figure 1), which resulted in significantly extend murine survival compared to either CTX or Alemtuzumab alone alone (p<0.01). In contrast, high-dose Doxorubicin (DOX, 5mg/kg) had minimal effect on bone marrow DHL and DOX+Alemtuzumab was not synergistic. Strikingly, CD11b+F4/80+CD11c- macrophages comprised 20-40% of bone marrow cellularity 8 days after CTX treatment of xengorafted mice. DHL cells isolated from treated mice were next examined for changes in surface protein expression. At 16 hours after treatment, bone marrow DHL cells in mice receiving CTX (but not those receiving DOX or vehicle) had significantly reduced levels of surface CD47, the “don’t eat me” signal that suppresses macrophage phagocytosis. By 48 hours after treatment, bone marrow DHL cells in mice receiving CTX (but not those receiving DOX or vehicle) had significantly increased expression of the pro-phagocytic proteins CD31 and Calreticulin. Next, we performed cytokine array analysis on bone marrow from mice engrafted with DHL PDXs at 16 hours after treatment. CTX-treated (but not vehicle- or DOX-treated) bone marrow had significantly increased levels of human IL-16 and CCL4, cytokines known to promote monocyte recruitment, as well as VEGF, PDGF and TNFα. Ex-vivo phagocytosis assays using murine bone marrow-derived macrophages confirmed that recombinant human VEGF and TNFα each enhanced macrophage phagocytosis of Alemtuzumab coated Raji lymphoma cells. Findings from RNA-seq studies on tumor cells and macrophages from the spleen and bone marrow as well as therapy induced changes in these respective niches will be further discussed at the meeting. In summary, we have demonstrated that high-dose CTX induces a phenotypic and secretory response from human DHL cells in vivo that promotes phagocytosis of Alemtuzumab-opsinized cells. As a result, we are initiating a clinical trial of this combination in patients with relapsed/refractory DHL. [Display omitted] DisclosuresNo relevant conflicts of interest to declare.

Highlights

  • While in vitro culture systems can be used to assess sensitivity of cancer cells to therapy, in vivo drug administration is necessary to study the effect of therapeutics that depend on cell extrinsic induction of apoptosis such as immune mediated clearance

  • Immunoblotting NSG mice were injected with 106 tumor cells intravenously and treated with PBS, Dox or CTX. 16 (DFBL-20954) or 48 hours (DFBL-69487) after treatment bone marrow or splenic cells were harvested as above and human cells purified using an EasySep Mouse/Human Chimera Isolation Kit (StemCell Technologies)

  • Chromatin Immunoprecipitation and quantitative PCR (ChIP-qPCR) NSG mice were injected with 106 tumor cells intravenously and treated with PBS, Dox or CTX. 16 (DFBL-20954) or 48 (DFBL-69487) hours after treatment bone marrow or splenic cells were harvested as above and human cells purified using an EasySep Mouse/Human Chimera Isolation Kit (StemCell Technologies)

Read more

Summary

Chapter 1: Introduction

Non-Hodgkin’s Lymphoma and Diffuse Large B-cell Lymphomas Non-Hodgkin’s Lymphomas (NHLs) are the sixth most common cancer in the United. 1) whether the effects of high-dose CTX extend to bona fide human DHL; 2) whether other alkylating agents in the CTX drug class could recapitulate synergy with alemtuzumab; 3) the mechanisms of bone marrow antibody resistance in the “humaneered DHL,” and how this resistance was established; 4) the requirement of macrophages for CTXmediated killing; 5) whether human DHL exhibits the ASAP and how this is mechanistically promoted by alkylating agents and 6) the transcriptional and phenotypic effects of chemoimmunotherapies on lymphoma-associated macrophages that promote synergy with alemtuzumab. CTX treatment in lymphoma-bearing mice lead to an expansion of a unique macrophage subset characterized by surface CD36 and Fcgr[4] expression that was “super phagocytic” for antibody-coated lymphoma cells Together these findings illuminate the synergistic activity seen in the bone marrow and provide the rationale for employing the combination of high-dose cyclophosphamide and alemtuzumab in DHL and possibly other diseases with low macrophage: lymphoma cell ratios. Chapter 2: DHL bone marrow antibody resistance can be overcome by high-dose alkylating agent therapy

Background
Results
Chapter 7: Discussion
Chapter 8: Experimental Methods
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call