Treatment of solid tumors using bispecific anti-PDL-1/ICOS antibody

Author(s):  
Martin Perez-Santos ◽  
Maricruz Anaya-Ruiz ◽  
Gabriela Sanchez-Esgua ◽  
Luis Villafaña-Diaz ◽  
Diana Barron-Villaverde

PD-L1 and ICOS are immune control points in cancer and their presence in cancer tends to have a poor prognosis. WO2019122882 patent describes a bispecific antibody that targets PDL-1/ICOS with the potential application of cancer treatment. WO2019122882 patent describes a bispecific antibody with antitumor efficacy in CT26 model through of the depletion of TReg cells and improved ratio of CD8+ T cells: TReg in tumor microenvironment. The anti-PDL-1/ICOS antibody is new; however, only preclinical assays are shown using colon carcinoma model. So far, there are no reports of clinical trials to evaluate the safety, toxicity and efficacy, but it will be of great interest to analyze in the future if this antibody surpasses the action of the combinatorial therapy in cancer.

Author(s):  
Martin Perez-Santos

OX40 and 5T4 are molecules that play a role in T-cell expansion and cytoskeleton’s disruption in cancer, respectively. US2019161555 patent describes a bispecific antibody that targets OX40/5T4 with the potential application of cancer treatment. The method of analysis of the US201916155 patent consisted of claim’s analysis, as well as the chemical/biological information’s analysis of the bispecific antibody. The patent includes independent claims related to bispecific antibodies that bind to OX40/5T4, DNA encoding the antibodies, a vector that harbors the DNA, a host cell that contains the vector, a pharmaceutical composition containing a pharmaceutically effective amount of the antibodies, medical use of the antibodies, use of the antibodies in the treatment or prevention of neoplastic disorders and a method of treating neoplastic disorders. Bispecific antibodies that target OX40/5T4 can activate IL-2 secretion in CD4+ T cells.


Cancers ◽  
2021 ◽  
Vol 13 (8) ◽  
pp. 1850
Author(s):  
Justine Cinier ◽  
Margaux Hubert ◽  
Laurie Besson ◽  
Anthony Di Roio ◽  
Céline Rodriguez ◽  
...  

Regulatory T cells (Tregs) are present in a large majority of solid tumors and are mainly associated with a poor prognosis, as their major function is to inhibit the antitumor immune response contributing to immunosuppression. In this review, we will investigate the mechanisms involved in the recruitment, amplification and stability of Tregs in the tumor microenvironment (TME). We will also review the strategies currently developed to inhibit Tregs’ deleterious impact in the TME by either inhibiting their recruitment, blocking their expansion, favoring their plastic transformation into other CD4+ T-cell subsets, blocking their suppressive function or depleting them specifically in the TME to avoid severe deleterious effects associated with Treg neutralization/depletion in the periphery and normal tissues.


2019 ◽  
Vol 37 (15_suppl) ◽  
pp. e19028-e19028 ◽  
Author(s):  
Tong Chen ◽  
Yan Yuan ◽  
Liansheng Huang ◽  
Chengfei Pu ◽  
Tianling Ding ◽  
...  

e19028 Background: The chimeric antigen receptor (CAR) T cell treatment has been demonstrated as an effective therapy to relapse/refractory B cell malignancy. However, tumor microenvironment influences and affects CAR T treatment. For example, programmed death ligand 1/2 (PDL1/2) may inhibit the CAR T cells via interaction with up-regulated Programmed cell death protein 1 (PD1) during T cells activation, suppressing the tumor-killing capability of the CAR T cells. Thus, blockade of the PD1-PDL1/2 interaction may enhance the anti-tumor efficacy of CAR T therapy. Methods: We generated CAR T cells including an anti-CD19 second generation (2G) CAR molecule and a dominant negative PD1 molecule (Figure A). Compared with conventional CART cells, these “armored” CART cells show the enhanced capability of tumor killing after multiple-round tumor challenging and more “memory-like” phenotypes (Figure B). These results suggest dominant negative PD1 molecules may protect CART cells from exhaustion in the tumor microenvironment. Results: We report clinical trials of three refractory diffuse large B cell lymphomas (DLBCLs) patients that were successfully treated using the armored CAR T cells described above. All of these three patients failed to achieve response after multiple rounds of chemotherapy and radiotherapy. However, after infused with autologous CART cells at 5.23×10^6/kg and 1.97×10^6/kg, respectively, they showed significant tumor mass decrease and SUV max declines in PET/CT results and ongoing responses (e.g., from 34.48 to 3.89 at day 27, from 25.02 to 2.38 at day 31, respectively, see Figure C). Conclusions: These three clinical trials revealed the significant anti-bulky lymphoma response with respect to these armored CAR T cells and limited and tolerated cytokine release syndrome and central nervous system toxicity. Also, dominant negative PD1 molecules may augment CAR T cells persistence in patients after activation by lymphoma cells, thus enhancing the efficacy of CAR T cells in the treatment of hematomas. Finally, the techniques described herein are a platform technology and may be applied to other adoptive cellular immunotherapies such as TCR-T or TIL in the treatment of solid tumors. We are continuing to recruit more patients for the clinical trials. Clinical trial information: ChiCTR1900021295.


2016 ◽  
Vol 2016 ◽  
pp. 1-10 ◽  
Author(s):  
Xinfeng Chen ◽  
Mengjia Song ◽  
Bin Zhang ◽  
Yi Zhang

Reactive oxygen species (ROS) produced by cellular metabolism play an important role as signaling messengers in immune system. ROS elevated in the tumor microenvironment are associated with tumor-induced immunosuppression. T cell-based therapy has been recently approved to be effective for cancer treatment. However, T cells often become dysfunctional after reaching the tumor site. It has been reported that ROS participate extensively in T cells activation, apoptosis, and hyporesponsiveness. The sensitivity of T cells to ROS varies among different subsets. ROS can be regulated by cytokines, amino acid metabolism, and enzymatic activity. Immunosuppressive cells accumulate in the tumor microenvironment and induce apoptosis and functional suppression of T cells by producing ROS. Thus, modulating the level of ROS may be important to prolong survival of T cells and enhance their antitumor function. Combining T cell-based therapy with antioxidant treatment such as administration of ROS scavenger should be considered as a promising strategy in cancer treatment, aiming to improve antitumor T cells immunity.


2021 ◽  
Vol 12 ◽  
Author(s):  
Hua Zhu ◽  
Xinyao Hu ◽  
Lijuan Gu ◽  
Zhihong Jian ◽  
Liqin Li ◽  
...  

TUBA1C, a microtubule component, contributes to the development of several cancers. Our purpose was to study the expression of TUBA1C, its potential prognostic value, and its effects on the infiltration of immune cells of low-grade glioma (LGG). Through applying multiple bioinformatics analyses, we extracted and analyzed datasets from TCGA, TIMER, GTEx, GEPIA, and HPA to investigate the potential oncogenic mechanisms of TUBA1C, including the correlation between TUBA1C and prognosis, immune-checkpoints, tumor microenvironment (TME), and infiltration of immune cells in LGG. GO functional annotations and KEGG pathway analyses were further applied to investigate the potential action of TUBA1C in LGG. We revealed that the mRNA levels of TUBA1C were increased in LGG tumor tissues than in normal tissues. Additionally, TUBA1C was up-regulated in the grade III of LGG than in grade II. Moreover, we found that TUBA1C may be an independent prognostic factor of LGG, and high TUBA1C expression correlated to a poor prognosis of LGG. TUBA1C expression was positively associated with the infiltration of B cells, CD8 T+ cells, CD4+ T cells, macrophages, dendritic cells, and neutrophils. TUBA1C was also verified to be co-expressed with immune-related genes and immune-checkpoints. GO and KEGG pathway analyses indicated that TUBA1C may potentially regulate the pathogenesis of LGG through immune-related pathways, including chemokine pathway; JAK-STAT pathway; natural killer cell mediated cytotoxicity; T cell receptor pathway; leukocyte migration; negative regulation of immune system process; regulation of lymphocyte activation; T cell activation and other pathways. In conclusion, TUBA1C expression is increased in LGG and high TUAB1C expression is related to a poor prognosis. TUBA1C may influence tumor development by regulating the tumor-infiltrating cells in the TME. TUBA1C may be a potential target for immunotherapy.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A625-A625
Author(s):  
Christianne Groeneveldt ◽  
Priscilla Kinderman ◽  
Diana JM van den Wollenberg ◽  
Ruben L van den Oever ◽  
Jim Middelburg ◽  
...  

BackgroundThe use of T cell-engaging CD3-bispecific antibodies (CD3-bsAbs) is a promising immunotherapeutic strategy for cancer. Although this therapy has reached clinical practice for hematological malignancies, the absence of sufficient infiltrating T cells is a major barrier for efficacy in solid tumors.1 Oncolytic viruses are emerging as anti-cancer therapeutics, and accumulating evidence demonstrates their applicability to sensitize tumors for immune checkpoint immunotherapy.2 In this study, we exploited oncolytic reovirus as a strategy to enhance the efficacy of CD3-bsAbs in immune-silent, solid tumors.MethodsThe mutant p53 and K-ras induced murine pancreatic cancer model KPC3 resembles human pancreatic ductal adenocarcinomas with a desmoplastic tumor microenvironment, low T cell density, and resistance to immunotherapy. Immune-competent mice with established, subcutaneous KPC3 tumors were intratumorally injected with an optimized regimen of oncolytic reovirus (type 3 Dearing strain) and the reovirus-induced changes in the tumor microenvironment and lymphoid organs were analyzed over time by NanoString analysis, RT-qPCR and multicolor flow cytometry. The efficacy of combination with systemically injected CD3-bsAbs was evaluated in KPC3 and B16.F10 murine tumor models and the close-to-patient HER2+ BT474 breast cancer model with cell surface-expressed TRP1 and HER2 as target antigens, respectively. Primary outcome was tumor size, measured with caliper three times a week in a blinded-manner.ResultsReplication-competent reovirus induced an early IFN-signature, followed by a strong influx of CD8+ T cells (2.6-fold increase, p=0.0092). Viral replication declined after seven days and was associated with systemic activation of lymphocytes. Tumor-infiltrating T cells were mostly reovirus-specific and served as effector cells for the subsequently systemically administered CD3-bsAbs. The combination of reovirus and CD3-bsAbs induced regressions up to 70% in all mice with large, established KPC3, B16.F10, and BT474 tumors and significantly prolonged survival. Importantly, the employment of reovirus as a pre-conditioning regimen performed significantly better than the simultaneous or preceding administration of bsAbs. This combination treatment also induced regressions of non-injected distant lesions, suggesting that this therapy might be effective for metastatic disease.Abstract 590 Figure 1Reovirus sensitizes tumors for CD3-bsAb therapyReovirus-induced interferon signaling leads to increased T cell influx and subsequent effective CD3-bispecific antibody therapy in solid tumorsConclusionsOncolytic reovirus administration represents an effective strategy to induce a local IFN response and strong T cell influx, thereby sensitizing the tumor microenvironment for subsequent CD3-bsAb therapy (figure 1). Our data advocate for the inclusion of oncolytic viruses as a pre-conditioning strategy in T cell engaging antibody trials for solid tumors. Since both CD3-bispecific antibodies and oncolytic viruses are in advanced clinical development as monotherapies, efficient translation of this combination seems feasible.AcknowledgementsThis work was financially supported by the Dutch Cancer Society Bas Mulder Award 11056 (to NvM), a PhD fellowship from Leiden University Medical Center (to CG) and the Support Casper campaign by the Dutch foundation ‘Stichting Overleven met Alvleesklierkanker’ (supportcasper.nl) project numbers SOAK 17.04 and 19.03.Ethics ApprovalAll mouse studies were approved by the institutional Animal Welfare Body of Leiden University Medical Center and carried out under project licenses AVD1160020187004 or AVD116002015271, issued by the competent authority on animal experiments in the Netherland (named CCD).ReferencesBenonisson H, Altıntaş I, Sluijter M, Verploegen S, Labrijn AF, Schuurhuis DH, Houtkamp MA, Verbeek JS, Schuurman J and van Hall T. CD3-Bispecific antibody therapy turns solid tumors into inflammatory sites but does not install protective memory. Mol Cancer Ther 2019; 18(2):312–322.Groeneveldt, C, van Hall, T, van der Burg, SH, ten Dijke, P and van Montfoort, N. Immunotherapeutic potential of TGF-β inhibition and oncolytic viruses. Trends Immunol 2020; 41(5):406–420.


2022 ◽  
Vol 13 (1) ◽  
Author(s):  
Wen-jin Chen ◽  
Hao Cao ◽  
Jian-wei Cao ◽  
Li Zuo ◽  
Fa-jun Qu ◽  
...  

AbstractNon-clear renal cell carcinomas (nccRCCs) are less frequent in kidney cancer with histopathological heterogeneity. A better understanding of the tumor biology of nccRCC can provide more effective treatment paradigms for different subtypes. To reveal the heterogeneity of tumor microenvironment (TME) in nccRCC, we performed 10x sing-cell genomics on tumor and normal tissues from patients with papillary renal cell carcinoma (pRCC), chromophobe RCC (chrRCC), collecting duct carcinoma (CDRCC) and sarcomatoid RCC (sarRCC). 15 tissue samples were finally included. 34561 cells were identified as 16 major cell clusters with 34 cell subtypes. Our study presented the sing-cell landscape for four types of nccRCC, and demonstrated that CD8+ T cells exhaustion, tumor-associated macrophages (TAMs) and sarcomatoid process were the pivotal factors in immunosuppression of nccRCC tissues and were closely correlated with poor prognosis. Abnormal metabolic patterns were present in both cancer cells and tumor-infiltrating stromal cells, such as fibroblasts and endothelial cells. Combined with CIBERSORTx tool, the expression data of bulk RNA-seq from TCGA were labeled with cell types of our sing-cell data. Calculation of the relative abundance of cell types revealed that greater proportion of exhausted CD8+ T cells, TAMs and sarRCC derived cells were correlated with poor prognosis in the cohort of 274 nccRCC patients. To the best of our knowledge, this is the first study that provides a more comprehensive sight about the heterogeneity and tumor biology of nccRCC, which may potentially facilitate the development of more effective therapies for nccRCC.


2021 ◽  
Vol 22 (15) ◽  
pp. 8068
Author(s):  
Eleonora Timperi ◽  
Vincenzo Barnaba

CD39 is an enzyme which is responsible, together with CD73, for a cascade converting adenosine triphosphate into adenosine diphosphate and cyclic adenosine monophosphate, ultimately leading to the release of an immunosuppressive form of adenosine in the tumor microenvironment. Here, we first review the environmental and genetic factors shaping CD39 expression. Second, we report CD39 functions in the T cell compartment, highlighting its role in regulatory T cells, conventional CD4+ T cells and CD8+ T cells. Finally, we compile a list of studies, from preclinical models to clinical trials, which have made essential contributions to the discovery of novel combinatorial approaches in the treatment of cancer.


Sign in / Sign up

Export Citation Format

Share Document