scholarly journals SynNotch-CAR T cells overcome challenges of specificity, heterogeneity, and persistence in treating glioblastoma

2021 ◽  
Vol 13 (591) ◽  
pp. eabe7378 ◽  
Author(s):  
Joseph H. Choe ◽  
Payal B. Watchmaker ◽  
Milos S. Simic ◽  
Ryan D. Gilbert ◽  
Aileen W. Li ◽  
...  

Treatment of solid cancers with chimeric antigen receptor (CAR) T cells is plagued by the lack of ideal target antigens that are both absolutely tumor specific and homogeneously expressed. We show that multi-antigen prime-and-kill recognition circuits provide flexibility and precision to overcome these challenges in the context of glioblastoma. A synNotch receptor that recognizes a specific priming antigen, such as the heterogeneous but tumor-specific glioblastoma neoantigen epidermal growth factor receptor splice variant III (EGFRvIII) or the central nervous system (CNS) tissue-specific antigen myelin oligodendrocyte glycoprotein (MOG), can be used to locally induce expression of a CAR. This enables thorough but controlled tumor cell killing by targeting antigens that are homogeneous but not absolutely tumor specific. Moreover, synNotch-regulated CAR expression averts tonic signaling and exhaustion, maintaining a higher fraction of the T cells in a naïve/stem cell memory state. In immunodeficient mice bearing intracerebral patient-derived xenografts (PDXs) with heterogeneous expression of EGFRvIII, a single intravenous infusion of EGFRvIII synNotch-CAR T cells demonstrated higher antitumor efficacy and T cell durability than conventional constitutively expressed CAR T cells, without off-tumor killing. T cells transduced with a synNotch-CAR circuit primed by the CNS-specific antigen MOG also exhibited precise and potent control of intracerebral PDX without evidence of priming outside of the brain. In summary, by using circuits that integrate recognition of multiple imperfect but complementary antigens, we improve the specificity, completeness, and persistence of T cells directed against glioblastoma, providing a general recognition strategy applicable to other solid tumors.

2021 ◽  
Author(s):  
Joseph H Choe ◽  
Payal B Watchmaker ◽  
Milos S Simic ◽  
Ryan D Gilbert ◽  
Aileen W Li ◽  
...  

Treatment of solid cancers with chimeric antigen receptor (CAR) T cells is plagued by the lack of target antigens that are both tumor-specific and homogeneously expressed. We show that multiantigen prime-and-kill recognition circuits have the flexibility and precision to overcome these challenges in attacking glioblastoma. A synNotch receptor that recognizes a specific priming antigen, the heterogeneous glioblastoma neoantigen EGFRvIII or a brain tissue-specific antigen, is used to locally induce expression of a CAR, enabling thorough but controlled tumor killing by targeting of homogeneous antigens that are not absolutely tumor specific. Moreover, regulated CAR expression maintains a higher fraction of the T cells in the naïve-like state which is associated with higher durability in vivo. In summary, using circuits that integrate recognition of multiple imperfect but complementary antigens, we improve the specificity and persistence of T cells directed against glioblastoma, providing a general recognition strategy applicable to other solid tumors.


2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii111-ii112
Author(s):  
Payal Watchmaker ◽  
Joseph Choe ◽  
Milos Simic ◽  
Ryan Gilbert ◽  
Aileen li ◽  
...  

Abstract Treatment of solid cancers with chimeric antigen receptor (CAR) T-cells is challenging because of a lack of target antigens that are both tumor-specific and homogenously expressed. While epidermal growth factor receptor (EGFR)vIII represents a glioblastoma (GBM)-specific antigen, its expression is heterogeneous within the tumor resulting in tumor escape. In contrast, more homogenously expressed GBM-associated antigens (GAA), such as EphA2, are non-ideal because of expression in other normal organs, yielding potential cross-reactive toxicity. As a way to safely target GAAs in the tumor without attacking normal cells expressing the same GAAs outside of the brain, we adapted a novel synthetic Notch (synNotch) receptor system and established a “prime and kill” sequential two-receptor CAR circuit. A synNotch receptor recognizes a specific priming antigen; the heterogeneous GBM neoantigen EGFRvIII or a brain tissue-specific antigen to prime the local expression of a CAR that mediates cytotoxicity against a GAA (e.g. EphA2). In orthotopic GBM6 glioma model, a patient-derived xenograft (PDX) with heterogeneous expression of EGFRvIII, intravenous infusion of T-cells transduced with EGFRvIII synNotch→anti-IL-13Rα2/EphA2 tandem CAR circuit resulted in long-term (over 100 days) survival and eradication of the heterogeneous tumor in all of 12 mice in two independent experiments. In contrast, constitutive CARs targeting EGFRvIII or IL-13Rα2/EphA2 (as a tandem CAR) failed to exhibit long-term anti-tumor response. Moreover, T-cells transduced with synNotch-regulated CAR maintain a less differentiated state which is associated with higher durability compared with ones with constitutive CAR in vivo. T-cells transduced with a synNotch→CAR circuit primed by a brain-specific antigen, myelin oligodendrocyte glycoprotein (MOG), exhibited a precise and potent local control of intracranial PDX without evidence of priming in extracranial organs. These data support the utility of synNotch→CAR circuits in EGFRvIII-negative GBM cases. By integrating multiple imperfect but complementary antigens, we improve both the specificity and persistence of T-cells directed against GBM.


2021 ◽  
Vol 14 (1) ◽  
Author(s):  
Julien Edeline ◽  
Roch Houot ◽  
Aurélien Marabelle ◽  
Marion Alcantara

AbstractChimeric antigen receptor (CAR)-modified T cells and BiTEs are both immunotherapies which redirect T cell specificity against a tumor-specific antigen through the use of antibody fragments. They demonstrated remarkable efficacy in B cell hematologic malignancies, thus paving the way for their development in solid tumors. Nonetheless, the use of such new drugs to treat solid tumors is not straightforward. So far, the results from early phase clinical trials are not as impressive as expected but many improvements are under way. In this review we present an overview of the clinical development of CAR-T cells and BiTEs targeting the main antigens expressed by solid tumors. We emphasize the most frequent hurdles encountered by either CAR-T cells or BiTEs, or both, and summarize the strategies that have been proposed to overcome these obstacles.


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.


2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Zhifen Yang ◽  
Lingyu Li ◽  
Ahu Turkoz ◽  
Pohan Chen ◽  
Rona Harari-Steinfeld ◽  
...  

Abstract Background Adoptive transfer of chimeric antigen receptor (CAR)-engineered T cells combined with checkpoint inhibition may prevent T cell exhaustion and improve clinical outcomes. However, the approach is limited by cumulative costs and toxicities. Methods To overcome this drawback, we created a CAR-T (RB-340-1) that unites in one product the two modalities: a CRISPR interference-(CRISPRi) circuit prevents programmed cell death protein 1 (PD-1) expression upon antigen-encounter. RB-340-1 is engineered to express an anti-human epidermal growth factor receptor 2 (HER2) CAR single chain variable fragment (scFv), with CD28 and CD3ζ co-stimulatory domains linked to the tobacco etch virus (TEV) protease and a single guide RNA (sgRNA) targeting the PD-1 transcription start site (TSS). A second constructs includes linker for activation of T cells (LAT) fused to nuclease-deactivated spCas9 (dCas9)-Kruppel-associated box (KRAB) via a TEV-cleavable sequence (TCS). Upon antigen encounter, the LAT-dCas9-KRAB (LdCK) complex is cleaved by TEV allowing targeting of dCas9-KRAB to the PD-1 gene TSS. Results Here, we show that RB-340-1 consistently demonstrated higher production of homeostatic cytokines, enhanced expansion of CAR-T cells in vitro, prolonged in vivo persistence and more efficient suppression of HER2+ FaDu oropharyngeal cancer growth compared to the respective conventional CAR-T cell product. Conclusions As the first application of CRISPRi toward a clinically relevant product, RB-340-1 with the conditional, non-gene editing and reversible suppression promotes CAR-T cells resilience to checkpoint inhibition, and their persistence and effectiveness against HER2-expressing cancer xenografts.


2019 ◽  
Vol 2 (1) ◽  
pp. 01-11
Author(s):  
Muhammad AH Khoiruddin ◽  
Yusi Windya Febriyanti ◽  
Nafia Amalia

Uveal Melanoma (UM) is the primary intraocular tumor most commonly found in adults. The combination of therapy, Brachytherapy, surgery, Tranpupillary Thermal Therapy (TTT), Proton Beam Theraphy has not produced satisfactory results. The discovery of HER2 receptors expressed by UM cells can be used as a specific antigen target for the treatment of CAR T-celss. However, the effectiveness of CAR T-cell immunotherapy in tumors results in immunosuppressive T cells caused by an increase in Programmed cell Death Ligand-1 (PD-L1). This literature review demonstrates the success of HER2-specific CART T Cells as UM therapeutic efforts capable of eliminating tumor cells. In addition, CRSPR / Cas9 PD-1 / PD-L1-blocking modified HER2-specific CAR T cells can be a gene innovation in UM sufferers. Further clinical trials are needed to prove the effectiveness of CRISPR / Cas9 PD-1 / PD-L1-Blocking modified HER2-specific CAR T Cells in the treatment of UM patients.Keywords: Uveal melanoma, CAR T cell, reseptor HER


2021 ◽  
Author(s):  
Axel Hyrenius-Wittsten ◽  
Yang Su ◽  
Minhee Park ◽  
Julie M Garcia ◽  
Nathaniel Perry ◽  
...  

The lack of highly tumor-specific antigens limits the development of engineered T cell therapeutics because of life-threatening on-target/off-tumor toxicities. Here we identify ALPPL2 as a tumor-specific antigen expressed in a spectrum of solid tumors, including mesothelioma. ALPPL2 can act as a sole target for chimeric antigen receptor (CAR) therapy or be combined with tumor-associated antigens such as MCAM or mesothelin in synthetic Notch (synNotch) CAR combinatorial antigen circuits. SynNotch CAR T cells display superior tumor control when compared to CAR T cells to the same antigens by prevention of CAR-mediated tonic signaling allowing T cells to maintain a long-lived memory and non-exhausted phenotype. Collectively, we establish ALPPL2 as a clinically viable target for multiple solid tumors and demonstrate the multi-faceted therapeutic benefits of synNotch CAR T cells.


2021 ◽  
Vol 9 (9) ◽  
pp. e002737
Author(s):  
Justin T Huckaby ◽  
Elisa Landoni ◽  
Timothy M Jacobs ◽  
Barbara Savoldo ◽  
Gianpietro Dotti ◽  
...  

BackgroundChimeric antigen receptor (CAR) T cells have shown considerable promise as a personalized cellular immunotherapy against B cell malignancies. However, the complex and lengthy manufacturing processes involved in generating CAR T cell products ex vivo result in substantial production time delays and high costs. Furthermore, ex vivo expansion of T cells promotes cell differentiation that reduces their in vivo replicative capacity and longevity.MethodsHere, to overcome these limitations, CAR-T cells are engineered directly in vivo by administering a lentivirus expressing a mutant Sindbis envelope, coupled with a bispecific antibody binder that redirects the virus to CD3+ human T cells.ResultsThis redirected lentiviral system offers exceptional specificity and efficiency; a single dose of the virus delivered to immunodeficient mice engrafted with human peripheral blood mononuclear cells generates CD19-specific CAR-T cells that markedly control the growth of an aggressive pre-established xenograft B cell tumor.ConclusionsThese findings underscore in vivo engineering of CAR-T cells as a promising approach for personalized cancer immunotherapy.


2015 ◽  
Vol 33 (15) ◽  
pp. 1688-1696 ◽  
Author(s):  
Nabil Ahmed ◽  
Vita S. Brawley ◽  
Meenakshi Hegde ◽  
Catherine Robertson ◽  
Alexia Ghazi ◽  
...  

Purpose The outcome for patients with metastatic or recurrent sarcoma remains poor. Adoptive therapy with tumor-directed T cells is an attractive therapeutic option but has never been evaluated in sarcoma. Patients and Methods We conducted a phase I/II clinical study in which patients with recurrent/refractory human epidermal growth factor receptor 2 (HER2) –positive sarcoma received escalating doses (1 × 104/m2 to 1 × 108/m2) of T cells expressing an HER2-specific chimeric antigen receptor with a CD28.ζ signaling domain (HER2-CAR T cells). Results We enrolled 19 patients with HER2-positive tumors (16 osteosarcomas, one Ewing sarcoma, one primitive neuroectodermal tumor, and one desmoplastic small round cell tumor). HER2-CAR T-cell infusions were well tolerated with no dose-limiting toxicity. At dose level 3 (1 × 105/m2) and above, we detected HER2-CAR T cells 3 hours after infusion by quantitative polymerase chain reaction in 14 of 16 patients. HER2-CAR T cells persisted for at least 6 weeks in seven of the nine evaluable patients who received greater than 1 × 106/m2 HER2-CAR T cells (P = .005). HER2-CAR T cells were detected at tumor sites of two of two patients examined. Of 17 evaluable patients, four had stable disease for 12 weeks to 14 months. Three of these patients had their tumor removed, with one showing ≥ 90% necrosis. The median overall survival of all 19 infused patients was 10.3 months (range, 5.1 to 29.1 months). Conclusion This first evaluation of the safety and efficacy of HER2-CAR T cells in patients with cancer shows the cells can persist for 6 weeks without evident toxicities, setting the stage for studies that combine HER2-CAR T cells with other immunomodulatory approaches to enhance their expansion and persistence.


Blood ◽  
2020 ◽  
Vol 135 (9) ◽  
pp. 668-679 ◽  
Author(s):  
Natasha Vinanica ◽  
Arthur Yong ◽  
Desmond Wong ◽  
Yi Tian Png ◽  
See Voon Seow ◽  
...  

Abstract In adoptive T-cell immunotherapy of cancer, expansion and persistence of effector cells is a key determinant of response. We tested whether T lymphocytes could be rendered sensitive to erythropoietin (Epo) through ectopic expression of its wild-type receptor or a truncated form (EpoRm), which augments Epo signaling in erythrocyte progenitors. Both receptors could be expressed in human T lymphocytes; Epo ligation induced STAT5 phosphorylation, which was abrogated by nontoxic concentrations of the JAK1/2 inhibitor ruxolitinib. EpoRm had higher expression and triggered more potent stimulation than its wild-type counterpart, including superior T-cell survival and proliferation. Using a bicistronic vector, we expressed EpoRm together with an anti–CD19-41BB-CD3ζ chimeric antigen receptor (CAR), while maintaining the functions of each receptor. In the presence of Epo, EpoRm-CAR T cells had greater ex vivo expansion than CAR T cells and killed CD19+ leukemic cells more effectively in long-term cultures. In immunodeficient mice, physiologic levels of murine Epo were sufficient to preferentially expand EpoRm-CAR T cells, yielding a significantly higher antileukemic activity. Thus, outfitting adoptive T cells with EpoRm should yield greater effector-to-target ratios with a smaller number of infused cells; Epo or ruxolitinib administration could be used to adjust their levels postinfusion, maximizing antitumor activity and minimizing toxicity.


Sign in / Sign up

Export Citation Format

Share Document