Drug-Induced Cytokine Release Sensitizes Protective Tumor Microenvironments to Antibody- Mediated Therapy

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1335-1335
Author(s):  
Christian P. Pallasch ◽  
Leskov Ilya ◽  
Yadira M Soto ◽  
Christian J. Braun ◽  
Nadine Kutsch ◽  
...  

Abstract Abstract 1335 Therapy-resistant microenvironments represent a major barrier to the effective elimination of disseminated malignancies. However, microenvironment dependent resistance mechanisms as potential synergistic drug interactions particularly for biologicals and monoclonal therapeutic antibodies are not completely understood. Here, we used the hMB humanized lymphoma mouse model as primary human B-ALL xenograft mice to address mechanisms of resistance and potential synergy of the clinical grade antibodies alemtuzumab and rituximab. Response to antibody therapy was shown to be mediated by macrophages as effector cells by direct phagocytosis - indicated by an abrogated response in macrophage-depleted mice. Antibody mono-therapy however still showed limited response in the bone marrow as a site of a primarily resistant microenvironment. In order to overcome therapeutic resistance and generate a functional state of the tumor microenvironment allowing effective antibody-mediated phagocytosis of lymphoma cells we applied a series of combinatorial regimens. Supplementing treatment with GM-CSF in order to improve the effector to target ratio slightly enhanced the therapeutic response as only modest additive affects were seen with total body irradiation. By combining alemtuzumab and cyclophosphamide in the hMB model as rituximab and cyclophosphamide respectively in CD20+ B-ALL xenografts we identified a striking synergy leading to profound depletion of malignant cells from bone marrow and spleen. Mice treated in the combinatorial arm survived significantly longer (Median survival 7 weeks vs. 28 weeks, p<0.001). Notably, synergy was exclusively demonstrated in cyclophosphamide while other alkylating agents, topoisomerase inhibitors, spindle poisons or steroids did not elicit a strong therapeutic synergy. Cyclophosphamide treatment induced an increased frequency of F4/80+ macrophages in the primarily resistant bone marrow in and multiphoton confocal microscopy of leukemia infiltrated and treated tumor tissue revealed enhanced phagocytic activity. Analyzing the underlying mechanisms of cyclophosphamide – antibody synergy using conditioned media from cyclophosphamide pretreated leukemia cells we identified an acute secretory response significantly enhancing macrophage mediated leukemia cell depletion upon antibody treatment in vivo. Analyzing a panel of human cytokines we could identify VEGF, CLL4, TNFα, and IL8 to be specifically induced by cyclophosphamide while not being induced by total body irradiation. Using recombinant cytokines or blocking antibodies in conditioned media we could recapitulate the significant influence of these cytokines on enhancing macrophage-dependent leukemia cell removal in vitro. Since the cyclophosphamide-induced acute secretory activating phenotype (ASAP) shows a rapid transient cytokine release synergistic chemo-immunotherapy of antibody and cyclophosphamide is limited to a 24h time-frame of simultaneous co-dosing to provide synergy. Here we could identify secretory phenotypes of malignant cells determining therapeutic outcome in antibody-based therapies. These findings underline the importance of tumor-microenvironment interactions for therapeutic outcome. Thus, the acute induction of stress-related cytokines represents a highly effective strategy to target cancer cells for targeted removal by the innate immune system. Here we identified so far unraveled mechanisms of synergy in chemo-immunotherapy and will thereby improve future design of clinical treatment regimens. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2012 ◽  
Vol 120 (13) ◽  
pp. 2600-2609 ◽  
Author(s):  
Maegan L. Capitano ◽  
Michael J. Nemeth ◽  
Thomas A. Mace ◽  
Christi Salisbury-Ruf ◽  
Brahm H. Segal ◽  
...  

Abstract Neutropenia is a common side effect of cytotoxic chemotherapy and radiation, increasing the risk of infection in these patients. Here we examined the impact of body temperature on neutrophil recovery in the blood and bone marrow after total body irradiation (TBI). Mice were exposed to either 3 or 6 Gy TBI followed by a mild heat treatment that temporarily raised core body temperature to approximately 39.5°C. Neutrophil recovery was then compared with control mice that received either TBI alone heat treatment alone. Mice that received both TBI and heat treatment exhibited a significant increase in the rate of neutrophil recovery in the blood and an increase in the number of marrow hematopoietic stem cells and neutrophil progenitors compared with that seen in mice that received either TBI or heat alone. The combination treatment also increased G-CSF concentrations in the serum, bone marrow, and intestinal tissue and IL-17, IL-1β, and IL-1α concentrations in the intestinal tissue after TBI. Neutralizing G-CSF or inhibiting IL-17 or IL-1 signaling significantly blocked the thermally mediated increase in neutrophil numbers. These findings suggest that a physiologically relevant increase in body temperature can accelerate recovery from neutropenia after TBI through a G-CSF–, IL-17–, and IL-1–dependent mechanism.


Cancer ◽  
1992 ◽  
Vol 69 (11) ◽  
pp. 2853-2865 ◽  
Author(s):  
Mahmuf Ozsahin ◽  
Francoise Pène ◽  
Emmanuel Touboul ◽  
Brigitte Gindrey-Vie ◽  
Claude Dominique ◽  
...  

1995 ◽  
Vol 31 ◽  
pp. S236
Author(s):  
V. Fayrel ◽  
Th. Schmitt ◽  
J.L. Stephan ◽  
D. Guyotat ◽  
G. Puel ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document