scholarly journals 869 Locally administered immunotherapy self-delivering RNAi PH-762 results in abscopal clearance of untreated distal tumors, suggesting systemic immune response, in a murine hepatocarcinoma model

2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A910-A910
Author(s):  
Benjamin Cuiffo ◽  
Melissa Maxwell ◽  
Dingxue Yan ◽  
Brianna Rivest ◽  
James Cardia ◽  
...  

BackgroundThe development of locally administered immune checkpoint inhibition (ICI) holds potential promise for enhanced activity and decreased systemic toxicity, but such an approach is challenging with the available ICI antibodies. We have previously shown that the intratumoral (IT) delivery of PH-762, a self-delivering RNAi compound targeting PD-1 based on proprietary INTASYL™ technology, can significantly inhibit tumor growth associated with changes in the immune cell population in the tumor microenvironment towards an anti-tumor phenotype. We present data showing that IT administration of PH-762 not only inhibits local tumor growth but can also elicit an abscopal effect in distal untreated tumors. The in vivo efficacy and in vitro mechanism of action support the generation of a PH-762 driven systemic anti-tumor immune response. Therefore, ICI using INTASYL is an alternative to antibody drugs for immunotherapy.MethodsTo assess in vivo efficacy, Hepa1–6 cells were implanted subcutaneously into the flanks of C57BL/6J mice. Vehicle (PBS) or murine targeting PH-762 (mPH-762) were administered IT on Days 1, 4, 7, 10 and 14. To determine an abscopal effect cells were also implanted into the opposite flank but left untreated. Tumor volumes and body weights were recorded. In addition, in vitro mechanism of action studies were performed with CD3-stimulated human pan T cells. PD-1 mRNA knockdown was assessed by qRT-PCR; PD-1 protein expression by flow cytometry; and T cell function by cytokine release.ResultsTreatment with IT administered mPH-762 significantly inhibited tumor growth compared with vehicle treated control tumors. Furthermore, the growth of the untreated bilateral tumor was significantly reduced with 80% of these tumors showing complete regression. Mechanism of action studies showed potent and durable silencing of PD-1. Increased release of IFN-γ, CXCL10, and IL-6 and suppression of IL-10 release were indicators of an enhanced immune response.ConclusionsThese data show that silencing PD-1 with IT administration of mPH-762 not only inhibits growth of treated tumors but elicits an abscopal effect leading to cure of distal tumors. This data and other recently published data showing evidence of a specific antitumor immune response in a tumor rechallenge model after prior treatment with INTASYL compounds, demonstrate the desired systemic immune response can be obtained with local administration of PH-762. INTASYL represent an alternative to antibody therapy for IT checkpoint blockade with potential for improved efficacy and reduced systemic toxicity which will be investigated in an upcoming clinical trial.

Author(s):  
С.В. Калиш ◽  
С.В. Лямина ◽  
А.А. Раецкая ◽  
О.П. Буданова ◽  
И.Ю. Малышев

Цель - представить доказательства правомерности гипотезы, что комбинированный пул репрограммированных in vitro макрофагов и лимфоцитов будет эффективно ограничивать пролиферацию опухолевых клеток in vitro , а при введении в организм будет существенно ограничивать развитие опухоли in vivo . Методика. Размножение опухолевых клеток инициировали in vitro путем добавления клеток карциномы Эрлиха (КЭ) в среду культивирования RPMI-1640. Развитие асцитной опухоли in vivo воспроизводили путем внутрибрюшной инъекции клеток КЭ мышам. Результаты. Установлено, что M3 макрофаги вместе с антиген-репрограммированными лимфоцитами оказывают выраженный противоопухолевый эффект и in vitro, и in vivo , который был существеннее противоопухолевого эффекта цисплатина. Заключение. Факты, свидетельствующие, что М3 макрофаги в сочетании с in vitro антиген-репрограммированными лимфоцитами значительно подавляют рост опухоли in vivo , делают перспективным разработку клинической версии биотехнологии ограничения роста опухоли путем предварительного программирования противоопухолевого иммунного ответа «в пробирке». Aim. To test a hypothesis that a combined pool of in vitro reprogrammed macrophages and lymphocytes will effectively limit growth of tumor cells in vitro , and injections of these cells into the body will considerably limit development of a tumor in vivo . Methods. Tumor growth was initiated in vitro by addition of Ehrlich carcinoma (EC) cells to the RPMI-1640 cell culture medium and in vivo by intraperitoneal injection of EC cells into mice. Results. M3 macrophages in combination with antigen-reprogrammed lymphocytes exerted a pronounced antitumor effect both in vitro and in vivo, which was superior to the effect of cisplatin. Conclusion. M3 macrophages in combination with in vitro antigen-reprogrammed lymphocytes significantly inhibited the tumor growth in vivo . This fact justifies development of a clinical version of the tumor growth restricting biotechnology using pre-programming of the antitumor immune response in vitro .


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. e13505-e13505
Author(s):  
Joline Sijing Lim ◽  
Todor Dimitrov ◽  
Kol Jia Yong ◽  
Chong Gao ◽  
Daniel G Tenen ◽  
...  

e13505 Background: Hepatocellular carcinoma (HCC) is the third leading cause of cancer related deaths worldwide, with chemotherapy or targeted therapy such as sorafenib achieving limited success. Recently stem cell factor SALL4 has emerged as a novel oncogene associated with leukemogenesis and is also implicated in many solid tumors. We have observed that SALL4 is not expressed in adult human liver tissues, but expressed in 30-40% of liver cancer, and this is associated with poorer prognosis and overall survival. We further tested whether inhibition of SALL4 function could be used for HCC treatment. Methods: A novel peptide blocking SALL4 function was designed and used to treat HCC lines with or without SALL4 expression. This is followed by evaluation for binding affinity, tumor growth inhibitory activity and mechanism of action. Treated cells were then transplanted in vivo into NOD/SCID mice and monitored for tumor growth. Comparison and/or combination of peptide with sorafenib were also carried out. Further modification of the peptide was done to allow for in vivo administration. Results: The peptide can effectively block SALL4 function. When used to treat HCC cell lines, it showed inhibitory effects in SNU398 cells (SALL4 expression), but not SNU387 cells (non-SALL4 expression). Post-xenotransplant, mice which received cells treated with peptide had slower rate of tumor growth (p=0.028) and lower tumor burden at dissection 26 days post transplant (p=0.048). Searching for its mechanism of action, we discovered that the peptide could affect the PTEN/AKT pathway, which was validated by western blot. When the peptide was combined with sorafenib, decreased cell viability was observed (p=0.03), suggestive of at least an additive effect between the peptide and sorafenib. Modification of peptide with TAT-protein showed similar inhibition of growth in vitro and was tested for further in-vivo usage through intraperitoneal injection. Conclusions: Our proof-of-principle studies have showed that a peptide blocking the function of stem cell factor SALL4 can be used as a novel approach for treating HCC. Combined with sorafenib, it may be able to enhance cell death and potentially lead to better outcomes in HCC patients.


2019 ◽  
Vol 21 (Supplement_6) ◽  
pp. vi269-vi270
Author(s):  
Victoria Sanchez ◽  
John Lynes ◽  
Stuart Walbridge ◽  
Xiang Wang ◽  
Nancy Edwards ◽  
...  

Abstract Preclinical models that reliably recapitulate the immunosuppressive properties of human gliomas are essential to assess immune-based therapies. Intracranially injected GL261 cells are widely used as an immunocompetent animal model of glioma, but it is common practice to transfect these with luciferase to facilitate tumor monitoring during treatment. Our group has previously shown that the luciferase-expressing GL261 Red-FLuc cells create an inflammatory response when implanted intracranially. Now, we additionally explore the inflammatory response of GL261-Luc2 cells and demonstrate a similar host immune response occurs with this model as well. In our in vivo evaluation, C57BL/6 mice underwent stereotaxic, intracranial implantation with GL261, GL261 Red-FLuc or GL261-Luc2 cells at doses of 5x104cells/5mL or 3x105cells/5uL.MRIs were performed to monitor relative tumor growth. To assess intrinsic differences between cell lines, in vitro cytokine profiles were evaluated by proteome microarray. Kaplan-Meier survival analyses demonstrated median survival for mice implanted with GL261 cells at 5x104cells was 18 to 21 days. The GL261-Red FLuc implanted mice cells did not reach median survival at either tumor dose with greater than 60% of mice termed long-term survivors. Finally, mice injected with GL261-Luc2 cells at 3x105cells reached median survival at 23 days, but median survival was significantly prolonged for mice implanted with GL261-Luc2 at a dose of 5x104cells (37 days, with 40% becoming long-term survivors) compared to GL261 implanted mice. MRIs reveal differences in tumor growth that correspond with the differences in median survival between groups. In addition, proteomic analyses revealed significantly elevated inflammatory cytokines such as IFN-gamma, IL-7 and TNF-alpha in the supernatants of the GL261 Red-FLuc cells and GL261-Luc2 cells. Further immune characterization is ongoing. Our data suggests that GL261 Red-FLuc and GL261-Luc2 murine models elicit an anti-tumor immune response by increasing pro-inflammatory modulators which stimulate the tumoricidal function of immune cells in the tumor microenvironment.


Author(s):  
John Kearney

There is a long trail of research studies testing the in vitro and in vivo efficacy of chloroquine and its derivatives in treating and preventing infection by various coronavirus species. More recent findings have highlighted the possibility of treating patients infected with the 2019 novel coronavirus, SARS-CoV-2. This review describes the mechanism of coronavirus infection, the mechanism of action of chloroquine, and summarizes the available literature highlighting the efficacy of chloroquine as an anti-coronavirus agent. These findings should encourage the wider scientific community to conduct thorough research on the possible efficacy of chloroquine and its derivatives in treating and preventing SARS-CoV-2 infection.


Cytokine ◽  
2010 ◽  
Vol 52 (1-2) ◽  
pp. 47
Author(s):  
Kyriaki Ioannou ◽  
Pinelopi Samara ◽  
Nadia Kavrochorianou ◽  
Christina Bega ◽  
George Thyphronitis ◽  
...  

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1837-1837
Author(s):  
Suresh Veeramani ◽  
George J. Weiner

Abstract Background Proteins within the complement system have complex effects on cellular immune responses. In previous studies, we found that active complement components, especially C5a, can dampen the development of antigen-specific immune responses following vaccination with a model antigen, in part by promoting generation of APC-induced T regulatory (Treg) cells. These studies also demonstrated that B lymphoma cell lines exposed to complement can induce Treg generation in vitro. The current study was designed to address whether depletion of C5a could enhance development of a cellular anti-lymphoma immune response in vivo. Methods Immunocompetent Balb/C mice were inoculated subcutaneously with syngeneic A20 B lymphoma cells mixed with either 10 μg of rat anti-mouse C5a monoclonal antibody (mAb) or 10 μg of isotype-matched Rat IgG2a control mAb. Tumor growth was followed. In select experiments, mice were sacrificed and analyzed for the percentage and activity of tumor-infiltrating T cells and A20-specific splenic T cell responses. Results 1. Tumor progression. Lymphoma grew more slowly in mice treated with anti-C5a mAb compared to mice treated with control mAb (p<0.05) {Fig. 1). 2. Intratumoral T cells. Tumors from mice treated with anti-C5a mAb had higher CD8+ T cell infiltration compared to mice treated with control mAb (p=0.002) (Fig. 2). Tumor-infiltrating CD8+ T cells showed a trend towards higher intracellular IFNg production in mice treated with anti-C5a mAb compared to control mAb (p=0.051). 3. Splenic T cells. Splenic T cells from mice treated with anti-C5a mAb produced IFNg to a greater degree than did splenic T cells from control mice when splenocytes were cultured with irradiated A20 cells in vitro (p=0.041) (Fig. 3). There was a trend towards decreased numbers of splenic CD4+CD25highFoxp3+ Tregs in C5a-depleted mice compared to control mice. Conclusions Depletion of C5a at the site of tumor inoculation slows tumor growth and increases the number of tumor infiltrating CD8 T cells in a syngenic immunocompetent model of lymphoma. A trend towards enhanced production of IFNg in the tumor infiltrating T cells, increased numbers of tumor-specific splenic T cells, and reduced numbers of splenic Tregs, suggests intratumoral C5a depletion can enhance tumor-specific immune responses both within the tumor and systemically. Ongoing studies are exploring the molecular mechanisms involved in C5a-promoted tumor progression and the use of C5a depletion as a novel strategy to improve anti-tumor immunity. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A936-A937
Author(s):  
Hui Yuwen ◽  
Tengteng Li ◽  
Yijing Ren ◽  
Dirk Hoenemann ◽  
Jay Mei ◽  
...  

BackgroundProgrammed death-ligand 1 (PD-L1) and programmed cell death protein 1(PD-1) blockade therapy has revolutionized the treatment landscape of malignancies. However, only a minority of patients are anticipated to experience a deep and durable response. In addition, successful therapeutic agonism of 4-1BB, a promising co-stimulatory immunologic target, has been limited by major safety concerns of hepatotoxicity or suboptimal agonistic potency. ATG-101, a novel PD-L1/4-1BB bispecific antibody, was designed to activate 4-1BB positive T cells in a PDL1-crosslinking dependent manner and to effectively treat tumors without on-target-off-tumor liver toxicity (figure 1).MethodsATG-101 was developed by introducing lower affinity 4-1BB scFv into a human IgG1 PD-L1 monoclonal antibody. The N297A mutation on CH2 abolishes the binding capacity to most FcγRs but retains the binding to FcγRn. A series of in vitro and in vivo studies were performed to evaluate the potency, safety and specific mechanism of action.ResultsATG-101 simultaneous binds to 4-1BB and PD-L1 with higher affinity to PD-L1, and potently activates 4-1BB positive T cells when crosslinked by PD-L1 positive cells. Upon crosslinking, ATG-101 also activates PD1+TIM3+ exhausted T cells in vitro, suggesting a potential in reversing T-cell dysfunction and exhaustion (figure 1). ATG-101 shows potent anti-tumor activities in various animal models, including h4-1BB humanized mice bearing MC38 colon cancer, PD(L)1 blockade insensitive B16F10 melanoma and EL4 lymphoma, with no body weight loss observed. To evaluate ATG-101 efficacy in tumors progressing after anti-PD(L)1 treatment, mice bearing MC38 tumors were treated with anti-PDL1 initially to achieve tumor growth inhibition, and half of the mice switched to ATG-101 upon disease progression, the other mice continuing with anti-PD-L1 treatment. ATG-101 induced potent tumor growth inhibition and tumor regression in anti-PDL1-resistant tumors and prolonged survival. Flow cytometry and multiplex IHC staining of tumor samples from mice treated with ATG-101 or control suggest that ATG-101 increases the infiltration, proliferation and activation of CD8+ T cells (figure 2), the infiltration of natural killer T cells and the CD8+/Treg ratio in TILs. In a 4-week GLP toxicity study in cynomolgus monkey, up to 100mg/kg repeated doses of ATG-101 were well tolerated with no hepatotoxicity observed.Abstract 893 Figure 1ATG-101 conditionally activates exhausted T cells. (A) Mechanism of action of ATG-101 (B) Exhausted T cells were induced by CD3+T cells cultured with anti-CD3/CD28 beads for 6 days. The percentage of terminally exhausted T cells (PD-1+Tim-3+) and progenitor exhausted T cells (PD-1+Tim-3-) were increased on Day6 (C) With the presence of PD-L1 positive cells, ATG-101 induced the IL2 and INF-γ secretion by exhausted T cells.Abstract 893 Figure 2Potent in vivo efficacy of ATG-101. (A) Representative MC38 tumor growth curve for individual mice treated with PBS (black), 10mpk atezolizumab (Atezo) only (red) or mice initially treated with 10mpk atezolizumab and switched to 13mpk ATG-101 upon disease progression (red-green); the arrow indicates the day switching Atezo to ATG-101; (B) MC38 tumor growth curve for all individual mice treated with PBS (black, n=6), atezolizumab only (red, n=14), and atezolizumab initially before switching to ATG-101 upon disease progression (red-green, n=14) ; (C) Survival data of mouse shown in (B); (D) Representative images for multiplex IHC staining of tumor samples collected from mouse from (B). (E) Quantitative analysis of TILs shown in (D). Compared with PBS group or atezo-only group, ATG-101 significantly increased the infiltration of T cells in the tumor microenvironment. MHCII: APC Cells, CD4+ CD8-: Helper T Cells, CD4- CD8+: Cytotoxic T Cells, CD4- CD8- F4/80- PDL1+: Tumor Cells.ConclusionsATG-101 demonstrated significant anti-tumor activity in various tumor models including those progressing on anti PD(L)1 treatment. Good safety and PK/PD properties has been demonstrated in preclinical in vivo models. A phase I, multicenter, dose-escalating clinical trial evaluating ATG-101 in patients with solid tumors and hematologic malignancies is ongoing.Ethics ApprovalThe protocol and any amendment(s) or procedures involving the care and use of animals in this study were reviewed and approved by the Institutional Animal Care and Use Committee (IACUC) of CrownBio or Innostar prior to execution with an AUP number or IACUC approval number for each animal study. During the study, the care and use of animals were conducted in accordance with the regulations of the Association for Assessment and Accreditation of Laboratory Animal Care (AAALAC).All studies were conducted following an approved IACUC protocol. AUP NO.:2004-12-1465, 2004-12-1000; IACUC approval number: IACUC-2021-M-003


2018 ◽  
Vol 48 (1) ◽  
pp. 97-110 ◽  
Author(s):  
Abdulmannan Fadel ◽  
Andrew Plunkett ◽  
Weili Li ◽  
Yazan Ranneh ◽  
Vivian Elewosi Tessu Gyamfi ◽  
...  

Purpose The purpose of this study is to discuss recent research on arabinoxylans from rice bran and wheat byproducts and their immunomodulatory potentials. Also, a potential receptor for arabinoxylans is proposed in relation to arabinoxylans structure. Design/methodology/approach This review summarises recent publications on arabinoxylans from rice bran and wheat, classification of arabinoxylans, a brief background on their method of extraction and their immunomodulatory potentials as they induce pro-inflammatory response in vitro, in vivo and in humans. The mechanism of action in which arabinoxylans modulate the immune activity is yet to be discovered, However, the authors have proposed a potential receptor for arabinoxylans in relation to arabinoxylans structure and molecular weight. Findings The effects of arabinoxylans from rice bran and wheat on the immune response was found to cause a pro-inflammatory response in vitro, in vivo and in humans. Also, the immune response depends on arabinoxylans structure, the degree of branching and origin. Originality/value This review paper focuses on the effects of arabinoxylans from rice bran and wheat on immunomodulatory potentials in vitro, in vivo and in humans. A new mechanism of action has been proposed based on the literature and via linking between arabinoxylans and lipopolysaccharide structure, molecular weight and suggested proposed receptor, which might be activated via both of them.


Sign in / Sign up

Export Citation Format

Share Document