Mac-1 (CD11b/CD18) is crucial for effective Fc receptor–mediated immunity to melanoma

Blood ◽  
2003 ◽  
Vol 101 (1) ◽  
pp. 253-258 ◽  
Author(s):  
Annemiek B. van Spriel ◽  
Heidi H. van Ojik ◽  
Annie Bakker ◽  
Marco J. H. Jansen ◽  
Jan G. J. van de Winkel

Abstract Antibody-reliant destruction of tumor cells by immune effector cells is mediated by antibody-dependent cellular cytotoxicity, in which Fc receptor (FcR) engagement is crucial. This study documents an important role for the β2 integrin Mac-1 (CD11b/CD18) in FcR-mediated protection against melanoma. CD11b-deficient mice, those that lack Mac-1, were less protected by melanoma-specific monoclonal antibody TA99 than wild-type (WT) mice. Significantly more lung metastases and higher tumor loads were observed in Mac-1−/− mice. Histologic analyses revealed no differences in neutrophil infiltration of lung tumors between Mac-1−/− and WT mice. Importantly, Mac-1−/−phagocytes retained the capacity to bind tumor cells, implying that Mac-1 is essential during actual FcR-mediated cytotoxicity. In summary, this study documents Mac-1 to be required for FcR-mediated antimelanoma immunity in vivo and, furthermore, supports a role for neutrophils in melanoma rejection.

Nanoscale ◽  
2021 ◽  
Author(s):  
Youshi Zheng ◽  
Cuilin Zhang ◽  
Zisen Lai ◽  
Yongyi Zeng ◽  
Juan Li ◽  
...  

In vivo modulating interactions between immune effector cells and tumor cells by bi-specific aptamer (Ap) is a promising strategy for cancer immunotherapy recently. However, it remains a technical challenge due...


2002 ◽  
Vol 30 (4) ◽  
pp. 507-511 ◽  
Author(s):  
M. Peipp ◽  
T. Valerius

In recent years, antibody therapy has become a new treatment modality for tumour patients, although the majority of responses are only partial and not long lasting. Based on evidence that effector-cell-mediated mechanisms significantly contribute to antibody efficacy in vivo, several approaches are currently persued to improve the interaction between Fc receptor-expressing effector cells and tumour target antigens. These approaches include application of Fc receptor-directed bispecific antibodies, which contain one specificity for a tumour-related antigen and another for a cytotoxic Fc receptor on immune effector cells. Thereby, bispecific antibodies selectively engage cytotoxic trigger molecules on killer cells, avoiding, for example, interaction with inhibitory Fc receptors. In vitro, chemically linked bispecific antibodies directed against the Fcγ receptors FcγRIII (CD16) and FcγRI (CD64), and the Fc α receptor FcαRI (CD89), were significantly more effective than conventional IgG antibodies. Recent animal studies confirmed the therapeutic potential of these constructs. However, results from clinical trials have been less promising so far and have revealed clear limitations of these molecules, such as short plasma half-lives compared with conventional antibodies. In this review, we briefly summarize the scientific background for bispecific antibodies, and describe the rationale for the generation of novel recombinant molecules. These constructs may allow us to more specifically tailor pharmacokinetic properties to the demands of clinical applications.


Cancers ◽  
2021 ◽  
Vol 13 (20) ◽  
pp. 5203
Author(s):  
Mostafa Jarahian ◽  
Faroogh Marofi ◽  
Marwah Suliman Maashi ◽  
Mahnaz Ghaebi ◽  
Abdolrahman Khezri ◽  
...  

Glycans linked to surface proteins are the most complex biological macromolecules that play an active role in various cellular mechanisms. This diversity is the basis of cell–cell interaction and communication, cell growth, cell migration, as well as co-stimulatory or inhibitory signaling. Our review describes the importance of neuraminic acid and its derivatives as recognition elements, which are located at the outermost positions of carbohydrate chains linked to specific glycoproteins or glycolipids. Tumor cells, especially from solid tumors, mask themselves by re-expression of hypersialylated neural cell adhesion molecule (NCAM), neuropilin-2 (NRP-2), or synaptic cell adhesion molecule 1 (SynCAM 1) in order to protect themselves against the cytotoxic attack of the also highly sialylated immune effector cells. More particularly, we focus on α-2,8-linked polysialic acid chains, which characterize carrier glycoproteins such as NCAM, NRP-2, or SynCam-1. This characteristic property correlates with an aggressive clinical phenotype and endows them with multiple roles in biological processes that underlie all steps of cancer progression, including regulation of cell–cell and/or cell–extracellular matrix interactions, as well as increased proliferation, migration, reduced apoptosis rate of tumor cells, angiogenesis, and metastasis. Specifically, re-expression of poly/oligo-sialylated adhesion molecules on the surface of tumor cells disrupts their interaction with immune-effector cells and contributes to pathophysiological immune escape. Further, sialylated glycoproteins induce immunoregulatory cytokines and growth factors through interactions with sialic acid-binding immunoglobulin-like lectins. We describe the processes, which modulate the interaction between sialylated carrier glycoproteins and their ligands, and illustrate that sialic acids could be targets of novel therapeutic strategies for treatment of cancer and immune diseases.


2017 ◽  
Vol 2017 ◽  
pp. 1-14 ◽  
Author(s):  
Shiming Ye ◽  
Melvin I. Fox ◽  
Nicole A. Belmar ◽  
Mien Sho ◽  
Debra T. Chao ◽  
...  

Enavatuzumab is a humanized IgG1 anti-TWEAK receptor monoclonal antibody that was evaluated in a phase I clinical study for the treatment of solid malignancies. The current study was to determine whether and how myeloid effector cells were involved in postulated mechanisms for its potent antitumor activity in xenograft models. The initial evidence for a role of effector cells was obtained in a subset of tumor xenograft mouse models whose response to enavatuzumab relied on the binding of Fc of the antibody to Fcγ receptor. The involvement of effector cells was further confirmed by immunohistochemistry, which revealed strong infiltration of CD45+ effector cells into tumor xenografts in responding models, but minimal infiltration in nonresponders. Consistent with the xenograft studies, human effector cells preferentially migrated toward in vivo-responsive tumor cells treated by enavatuzumab in vitro, with the majority of migratory cells being monocytes. Conditioned media from enavatuzumab-treated tumor cells contained elevated levels of chemokines, which might be responsible for enavatuzumab-triggered effector cell migration. These preclinical studies demonstrate that enavatuzumab can exert its potent antitumor activity by actively recruiting and activating myeloid effectors to kill tumor cells. Enavatuzumab-induced chemokines warrant further evaluation in clinical studies as potential biomarkers for such activity.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A263-A263
Author(s):  
Jonathan Nyce

BackgroundAntisense oligonucleotides function by targeting the messenger RNA coding for a target protein, rather than the protein itself. This laboratory previously introduced Respirable Antisense Oligonucleotides (RASONs) into human clinical trials for asthma.1–5 In that work we demonstrated that RASONs delivered by inhalation are absorbed into surfactant lining the surface of the lung; are distributed with high efficiency throughout the bronchial epithelium; and are taken up with therapeutic effect by both bronchial epithelial cells and immune effector cells resident throughout the bronchial epithelium, as well as in bronchial-associated lymphoid tissue (BALT). We have now re-engineered this technology to adapt it to the treatment of primary and metastatic lung tumors via immune checkpoint inhibition. While immune checkpoints expressed on lung tumors are not amenable to RASON inhibition, immune cells resident in the bronchial epithelium and BALT represent good targets for the RASON approach to checkpoint inhibition. E.g., SIRP-alpha is a receptor expressed by myeloid lineage cells such as dendritic cells (DCs), tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs). When CD47, found on the surface of tumor cells, binds to SIRP-alpha on immune effector cells, the anti-tumor action of such immune effector cells becomes significantly diminished. We hypothesized that RASONs targeting mRNA of immune checkpoint proteins found on immune effector cells would eliminate checkpoint proteins from their surface, such that when they were signaled to home in on lung tumors, they would arrive in the tumor-associated microenvironment in a state impervious to checkpoint ligands expressed on the surface of tumor cells. To test this hypothesis, we applied a RASON protocol to dogs with spontaneous lung tumors presenting to their veterinarians.MethodsIn this preliminary, proof-of-principal study, two dogs with histologically confirmed metastatic lung tumors were administered RASONs targeting PD1, CTLA-4 and SIRP-alpha, by inhalation, twice weekly for eight weeks.ResultsX-ray analysis performed two weeks after the conclusion of RASON treatment showed dramatic results. One dog showed complete tumor dissolution (figure 1), and the second dog showed near total tumor dissolution, with faint shadows remaining (figure 2).Abstract 244 Figure 1Canine 2 presented with a 3-cm spherical tumor (circled, left). After RASON treatment (right), tumor underwent complete regressionAbstract 244 Figure 2Canine 1 presented with one 9-cm tumor and four smaller tumors ranging from 1–2 cm (tumors are marked with dot at their center). Left, before RASON treatment). Right, after RASON treatment near complete tumor resolutionConclusionsWhile these are preliminary results, and need to be dramatically expanded, they provide an initial indication that the RASON approach might prove to be an effective addition to immune checkpoint inhibition. It possesses certain advantages over small molecule or antibody approaches to checkpoint inhibition. For example, rather than being delivered systemically, RASONs are delivered by inhalation directly to the target tissues-- the bronchial epithelium and BALT. Furthermore, it may be possible to reduce the toxicity of systemic treatments targeting checkpoint proteins on tumor cells, by reducing or eliminating their ligands on immune effector cells. In as much as the RASON approach to the treatment of human asthma failed in clinical trials as a result of its induction of an influx of macrophages into the lung, the ability to render TAMs impervious to the presence of tumor-associated immune checkpoints suggests that the RASON approach may hold considerable promise for the treatment of lung tumors.Ethics ApprovalAll research reported here involved informed consent by owners of dogs with spontaneous lung neoplasms, for which no satisfactory alternative treatment was available, and was performed in strict compliance with both the Basle Declaration, to which the laboratory is a signatory member, as well as guidelines published by the International Council for Laboratory Animal Science (ICLAS).ConsentN/AReferencesNyce JW, & Metzger, W. J. DNA antisense therapy for asthma in an animal model. Nature 1997; 385(6618), 721–725. https://doi.org/10.1038/385721a0Metzger, W. J., & Nyce, J. W. Respirable antisense oligonucleotide (RASON) therapy for allergic asthma. BioDrugs 1999; clinical immunotherapeutics, biopharmaceuticals and gene therapy 12(4), 237–243. https://doi.org/10.2165/00063030-199912040-00001Nyce J. W. ( 1997). Respirable antisense oligonucleotides as novel therapeutic agents for asthma and other pulmonary diseases. Expert opinion on investigational drugs;6(9):1149–1156. https://doi.org/10.1517/13543784.6.9.1149Nyce J. Respirable antisense oligonucleotides: a new, third drug class targeting respiratory disease. Current opinion in allergy and clinical immunology ( 2002);2(6):533–536. https://doi.org/10.1097/00130832-200212000-00009Sandrasagra, A., Tang, L., Leonard, S. A., Teng, K., Li, Y., Mannion, J. C., & Nyce, J. W. ( 2001). RASONs: a novel antisense oligonucleotide therapeutic approach for asthma. Expert opinion on biological therapy, 1(6), 979–983. https://doi.org/10.1517/14712598.1.6.979


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Faroogh Marofi ◽  
Heshu Sulaiman Rahman ◽  
Lakshmi Thangavelu ◽  
Aleksey Dorofeev ◽  
Favian Bayas-Morejón ◽  
...  

AbstractIn recent decades, a new method of cellular immunotherapy was introduced based on engineering and empowering the immune effector cells. In this type of immunotherapy, the immune effector cells are equipped with chimeric antigen receptor (CAR) to specifically target cancer cells. In much of the trials and experiments, CAR-modified T cell immunotherapy has achieved very promising therapeutic results in the treatment of some types of cancers and infectious diseases. However, there are also some considerable drawbacks in the clinical application of CAR-T cells although much effort is in progress to rectify the issues. In some conditions, CAR-T cells initiate over-activated and strong immune responses, therefore, causing unexpected side-effects such as systemic cytokine toxicity (i.e., cytokine release syndrome), neurotoxicity, on-target, off-tumor toxicity, and graft-versus-host disease (GvHD). To overcome these limitations in CAR-T cell immunotherapy, NK cells as an alternative source of immune effector cells have been utilized for CAR-engineering. Natural killer cells are key players of the innate immune system that can destroy virus-infected cells, tumor cells, or other aberrant cells with their efficient recognizing capability. Compared to T cells, CAR-transduced NK cells (CAR-NK) have several advantages, such as safety in clinical use, non-MHC-restricted recognition of tumor cells, and renewable and easy cell sources for their preparation. In this review, we will discuss the recent preclinical and clinical studies, different sources of NK cells, transduction methods, possible limitations and challenges, and clinical considerations.


Sign in / Sign up

Export Citation Format

Share Document