Summary

Generation and Functional Verification of Hypoxia-Sensitive Chimeric Antigen Receptor-T Cells

Published: June 14, 2024
doi:

Summary

Here we present a protocol for the generation and functional verification of hypoxia-sensitive chimeric antigen receptor (CAR)-T cells. This protocol presents the lentivirus-based generation of hypoxia-sensitive CAR-T cells and their characterization, including the validation of hypoxia-dependent CAR expression and selective cytotoxicity.

Abstract

Extensive studies have proven the promise of chimeric antigen receptor T (CAR-T) cell therapy in treating hematological malignancies. However, treating solid tumors remains challenging, as exemplified by the safety concerns that arise when CAR-T cells attack normal cells expressing the target antigens. Researchers have explored various approaches to enhance the tumor selectivity of CAR-T cell therapy. One representative strategy along this line is the construction of hypoxia-sensitive CAR-T cells, which are designed by fusing an oxygen-dependent degradation domain to the CAR moiety and are strategized to attain high CAR expression only in a hypoxic environment-the tumor microenvironment (TME). This paper presents a protocol for the generation of such CAR-T cells and their functional characterization, including methods to analyze the changes in CAR expression and killing capacity in response to different oxygen levels established by a mobile incubator chamber. The constructed CAR-T cells are anticipated to demonstrate CAR expression and cytotoxicity in an oxygen-sensitive manner, thus supporting their capability to distinguish between hypoxic TME and normoxic normal tissues for selective activation.

Introduction

Chimeric antigen receptor T cell (CAR-T) therapy has represented a significant breakthrough in cancer treatment. Since the Food and Drug Administration (FDA) approved the first CAR-T therapy for treating advanced/resistant lymphoma and acute lymphoblastic leukemia in 20171,2,3, 10 CAR-T therapies targeting CD19 or B-cell maturation antigen (BCMA) have received approval globally4. However, despite extensive research, replicating the remarkable efficacy of CAR-T therapy in treating hematological malignancies remains challenging for its application to solid tumors5,6,7,8.

The immunosuppressive tumor microenvironment (TME) is a primary contributor to the poor efficacy of CAR-T in the solid tumor setting. TME impedes the activity and survival of CAR-T cells due to insufficient nutrients, hypoxia, an acidic pH, and the accumulation of metabolic waste9,10,11,12. Further hostility comes from infiltrating immunosuppressive cells such as regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), and tumor-associated macrophages (TAM), which, alongside tumor cells, secrete immunosuppressive cytokines that cause additional inhibition of CAR-T cells once they enter the tumor13,14.

Apart from the unsatisfactory therapeutic efficiency, safety issues are another Achilles' heel of CAR-T cells when dealing with solid tumors15,16. The safety concern arises from the fact that none of the tumor-specific antigens (TSA) identified so far are strictly restricted to tumor cells. In other words, the tumor-associated antigens (TAA) chosen as the target of CAR, although showing higher expression in tumor cells, are often also expressed by normal tissues17. On-target, off-tumor effects could therefore occur from the unexpected activation of CAR-T cells upon CAR efficiently recognizing normal tissues, leading to cytokine release syndrome (CRS), CAR-T-related encephalopathy syndrome (CRES)18, and other adverse outcomes19.

Many strategies have been explored to avoid such effects, including decreasing the affinity of CAR to allow CAR-T cells to distinguish tumor cells from normal cells based on the expression levels of the targeted TAA; equipping CAR-T cells with an off switch, such as a suicide gene or elimination marker to promote their elimination upon unexpected activation; partitioning the CD3ζ and co-stimulatory signals into two CAR moieties, whose simultaneous engagement is consequently required for effective activation of CAR-T cells; utilizing a synthetic Notch (synNotch)-based circuit that restricts the activity of CAR-T cells to targeted cells co-expressing two different TAAs; and engineering CAR-T cells to attain TME sensitivity by implementing a mechanism to tune CAR expression to changing environmental cues20,21,22,23,24,25,26.

A key consideration in the TME sensitivity option outlined above is the low oxygen level in the TME due to the rapid proliferation of tumor cells. The accommodation of tumor cells to hypoxia hinges on the activation of hypoxia-inducible factor-1 (HIF-1), a heterodimeric transcriptional factor consisting of an inducible subunit, HIF-1α, and a constitutively expressed subunit, HIF-1β27. Under normoxic conditions, the HIF-1α protein undergoes ubiquitination and rapid proteasomal degradation, dependent on its oxygen-dependent degradation domain (ODD)28. When the cellular supply of oxygen becomes limited, HIF-1 is stabilized and activates the transcription of its downstream target genes by binding to hypoxia-response elements (HREs)29. Given the nature of ODD and HRE as oxygen-sensitive elements, they have been explored to realize the conditional expression of CARs within the hypoxic TME30. Here, we present a protocol focusing on methods for phenotypic and functional characterization of hypoxia-sensitive CAR-T cells, preceded by a brief description of the CAR design and the preparation procedures of these cells. This protocol intends to provide a useful guideline for exploiting hypoxia-responsive CAR to generate CAR-T cells with restrained off-tumor toxicity.

Protocol

In this study, HER2-BBz-ODD, a hypoxia-sensitive CAR targeting HER2 (Gene ID: 2064) was compared with its regular counterpart, HER2-BBz. The schematics of the two CARs are illustrated in Figure 1A, which shows that HER2-BBz-ODD is derived from HER2-BBz by adding the ODD sequence to the C-terminal of CD3ξ. The construction of lentiviral vectors expressing the two CARs and the generation of the corresponding lentivirus by 293T cell transfection has been previously described<sup class="xre…

Representative Results

Fusing the ODD domain of HIF-1α to the CAR moiety represents a primary strategy for generating a hypoxia-sensitive CAR. The hypoxia-sensitive HER2-targeting CAR analyzed in this study, named HER2-BBz-ODD, was constructed using this strategy by integrating the ODD sequence into its conventional HER2-BBz (Figure 1A). In this study, we used lentiviral transduction to express HER2-BBz-ODD CAR or HER2-BBz CAR and subsequently examined their oxygen sensitivity in two cell types: human PBMCs a…

Discussion

Safety concerns are significant issues that must be addressed for any CAR-T cell therapy to advance to clinical use. Utilizing the unique properties of tumor cells or the TME has become a primary research direction focusing on the development of CAR-T cells that target tumor tissues selectively. Designing a hypoxia-sensitive CAR-T is an attractive strategy in this direction, with several approaches being explored, including the one presented in this study-fusing the CAR moiety with the naturally occurring hypoxia-sensing…

Disclosures

The authors have nothing to disclose.

Acknowledgements

This work was supported by grants from the National Key Research and Development Program of China (2016YFC1303402), the National Megaproject on Key Infectious Diseases (2017ZX10202102, 2017ZX10304402-002-007), and the General Program of Shanghai Municipal Health Commission (201740194).

Materials

1.5 mL Centrifuge tube QSP 509-GRD-Q Supernatants and cells cellection
Protocol Step 2,3,4
10% ExpressCast PAGE NCM biotech P2012 Immunoblotting
Protocol Step 3
10x PBS NCM biotech 20220812 Cell culture
Protocol Step 4
10 mL pipette Yueyibio YB-25H Pipetting
Protocol Step 1
10xTRIS-Glycine-SDS electrophoresis buffer Epizyme 3673020 Immunoblotting
Protocol Step 3
15 mL Centrifuge tube Thermo Scientific 339650 Supernatants and cells cellection
Protocol Step 1
25 cm2 EasYFlask Thermo Scientific 156367 Cell culture
Protocol Step 3,4
4x Protein SDS PAGE Loading Buffer Takara 9173 Immunoblotting
Protocol Step 3
6-well flat-bottom tissue culture plates Thermo Scientific 140675 T Cells culture
Protocol Step 1
96-well black flat-bottom tissue culture plates Greiner 655090 Cytotoxicity assay
Protocol Step 4
96-well ELISA plates Corning 3590 ELISA
Protocol Step 5
96-well plate shaker QILINBEIER MH-2 Shake
Protocol Step 4
96-well U-bottom tissue culture plates Thermo Scientific 268200 Supernatants cellection
Protocol Step 4,5
anti-FLAG antibody Sigma F1804-50UG Immunoblotting
Protocol Step 3
Carbinol Sinopharm 10010061 Immunoblotting
Protocol Step 3
Carbon dioxide incubator Thermo Scientific 360 Cell culture
Protocol Step 1,2,3,4
Cell counting plate Hausser scientific 1492 Cell counting
Protocol Step 1,3,4
CELLection Pan Mouse IgG Kit Thermo Scientific 11531D Mouse IgG magnetic beads
Protocol Step 1
Centrifuge Thermo Scientific 75002432 Cell culture
Protocol Step 1,3,4
Chemiluminescence gel imaging system BIO-RAD 12003154 Immunoblotting
Protocol Step 3
Cobalt chloride solution (0.5 M) bioleaper BR4000203 Hypoxic condition
Protocol Step 2,3,4
DMEM Corning 10-103-CV Cell culture
Protocol Step 4
Electronic balance Sartorius PRACTUM612-1CN weigh
Protocol Step 5
FBS BI 04-001-1ACS Cell culture
Protocol Step 3,4
GAPDH Mouse mAb ABclonal AC002 Immunoblotting
Protocol Step 3
Gel electrophoresis apparatus BIO-RAD 1645070 Immunoblotting
Protocol Step 3
GloMax Microplate Readers Promega GM3000 luciferase activity measurement
Protocol Step 4
Goat anti-Mouse IgG (H+L) Yeasen P1126151 Immunoblotting
Protocol Step 3
High speed microfreezing centrifuge eppendorf 5810 R Cell culture
Protocol Step 1
Human IFN-γ ELISA Set BD 555142 ELISA
Protocol Step 5
Items: Recombinant Human IFN-γ Lyophilized Standard, Detection Antibody Biotin Anti-Human IFN-γ , Capture Antibody Purified Anti-Human IFN-γ, Enzyme Reagent Streptavidin-horseradish peroxidase conjugate (SAv-HRP)
Human IL-2 ELISA Set BD 555190 ELISA
Protocol Step 5
Items: Recombinant Human IL-2 Lyophilized Standard, Detection Antibody Biotin Anti-Human IL-2 , Capture Antibody Purified Anti-Human IL-2, Enzyme Reagent Streptavidin-horseradish peroxidase conjugate (SAv-HRP)
IL-15 R&D systems P40933 T Cells culture
Protocol Step 1
IL-21 Novoprotein GMP-CC45 T Cells culture
Protocol Step 1
IL-7 R&D systems P13232 T Cells culture
Protocol Step 1
Inverted microscope Olympus CKX41 Cell culture
Protocol Step 1,3,4
Jurkat ATCC TIB-152 CAR-Jurkat construction
Protocol Step 3
LSRFortessa BD LSRFortessa Flow cytometry
Protocol Step 2
Luciferase Assay System Promega E1501 luciferase reporter assay
Protocol Step 4
Items: Passive lysis buffer, firefly luciferase substrate
Microplate reader BioTek HTX ELISA
Protocol Step 5
mobile CO2/O2/N2 Incubator Chamber China Innovation Instrument Co., Ltd. Smartor118 Hypoxic condition
Protocol Step 2, 3, 4
Mouse Anti-Hexa Histidine tag Sigma SAB2702218 Immunoblotting
Protocol Step 3
NcmBlot Rapid Transfer Buffer NCM biotech WB4600 Immunoblotting
NcmECL Ultra NCM biotech P10300 Immunoblotting
Protocol Step 3
Items: NcmECL Ultra Luminol/Enhancer Reagent (A) ,NcmECL Ultra Stabilized Peroxide Reagent (B) 
NovoNectin -coated 48-well flat plates Novoprotein GMP-CH38 CAR-T cells construction
Protocol Step 1
OPD (o-phenylenediamine dihydrochloride) tablet set Sigma P9187 Substrate Reagent
Protocol Step 5
Items: OPD tablet (silver foil),urea hydrogen peroxide tablet (gold foil)
PE-conjugated anti-DYKDDDDK Biolegend 637310 Flow cytometry
Protocol Step 2
Protamine sulfate Sigma P3369-1OG Lentivirus infection
Protocol Step 1
Protein Marker 10 Kda-250 KDa Epizyme WJ102 Immunoblotting
Protocol Step 3
 Purifed NA/LE Mouse Anti-Human CD3 BD 566685 T Cells culture
Protocol Step 1
Purified NA/LE Mouse Anti-Human CD28 BD 555725 T Cells culture
Protocol Step 1
PVDF membrane Millipore 168627 Immunoblotting
Protocol Step 3
RPMI 1640 Corning 10-040-CVRC Cell culture
Protocol Step 3
Skim milk powder Yeasen S9129060 Immunoblotting
Protocol Step 3
SKOV3-Luc ATCC HTB-77 Cytotoxicity assay
Protocol Step 4
Trypsin-EDTA NCM biotech C125C1 Cell culture
Protocol Step 4
Tween 20 Sinopharm 30189328 Immunoblotting
Protocol Step 3
Water bath keelrein NB014467 Heating
Protocol Step 1
X-VIVO 15  LONZA 04-418Q Serum-free lymphocyte culture medium
Protocol Step 1

References

  1. Boardman, A. P., Salles, G. CAR T-cell therapy in large B cell lymphoma. Hematol Oncol. 41 (S1), 112-118 (2023).
  2. Chen, Y. -. J., Abila, B., Mostafa Kamel, Y. CAR-T: What is next. Cancers. 15 (3), 663 (2023).
  3. Barsan, V., et al. Tisagenlecleucel utilisation and outcomes across refractory, first relapse and multiply relapsed B-cell acute lymphoblastic leukemia: A retrospective analysis of real-world patterns. eClinicalMedicine. 65, 102268 (2023).
  4. Liu, Y., et al. Oncolytic herpes simplex virus delivery of dual CAR targets of CD19 and BCMA as well as immunomodulators to enhance therapeutic efficacy in solid tumors combined with CAR-T cell therapy. Front Oncol. 12, 1037934 (2022).
  5. Liu, C., Qi, T., Milner, J. J., Lu, Y., Cao, Y. Speed and location both matter: Antigen stimulus dynamics controls CAR-T cell response. Front Immunol. 12 (748768), (2021).
  6. Li, N., et al. Improving the anti-solid tumor efficacy of CAR-T cells by inhibiting adenosine signaling pathway. Oncoimmunology. 9 (1), 1824643 (2020).
  7. Derenzo, C., Gottschalk, S. Genetic modification strategies to enhance CAR T cell persistence for patients with solid tumors. Front Immunol. 10, 218 (2019).
  8. Fu, R., et al. Delivery techniques for enhancing CAR T cell therapy against solid tumors. Advanced Functional Materials. 31 (44), 2009489 (2021).
  9. Johnson, A., Townsend, M., O’Neill, K. Tumor microenvironment immunosuppression: A roadblock to CAR T-cell advancement in solid tumors. Cells. 11 (22), 3626 (2022).
  10. Liu, Z., et al. Immunosuppression in tumor immune microenvironment and its optimization from CAR-T cell therapy. Theranostics. 12 (14), 6273-6290 (2022).
  11. Luo, Z., et al. Modulating tumor physical microenvironment for fueling CAR-T cell therapy. Adv Drug Deliv Rev. 185, 114301 (2022).
  12. Martinez, M., Moon, E. K. CAR T cells for solid tumors: New strategies for finding, infiltrating, and surviving in the tumor microenvironment. Front Immunol. 10, 128 (2019).
  13. Zhang, X., et al. The immunosuppressive microenvironment and immunotherapy in human glioblastoma. Front Immunol. 13, 1003651 (2022).
  14. Tie, Y., Tang, F., Wei, Y. Q., Wei, X. W. Immunosuppressive cells in cancer: Mechanisms and potential therapeutic targets. J Hematol Oncol. 15 (1), (2022).
  15. Wu, Y., et al. Engineering CAR T cells for enhanced efficacy and safety. APL Bioeng. 6 (1), 011502 (2022).
  16. Al-Haideri, M., et al. CAR-T cell combination therapy: The next revolution in cancer treatment. Cancer Cell Int. 22 (1), 365 (2022).
  17. Fuca, G., Reppel, L., Landoni, E., Savoldo, B., Dotti, G. Enhancing chimeric antigen receptor T-cell efficacy in solid tumors. Clin Cancer Res. 26 (11), 2444-2451 (2020).
  18. Neelapu, S. S., et al. Chimeric antigen receptor T-cell therapy – assessment and management of toxicities. Nat Rev Clin Oncol. 15 (1), 47-62 (2018).
  19. Mi, J., Ye, Q., Min, Y. Advances in nanotechnology development to overcome current roadblocks in CAR-T therapy for solid tumors. Front Immunol. 13, 849759 (2022).
  20. Sterner, R. C., Sterner, R. M. CAR-T cell therapy: Current limitations and potential strategies. Blood Cancer J. 11 (4), 69 (2021).
  21. Yu, S., Yi, M., Qin, S., Wu, K. Next generation chimeric antigen receptor T cells: Safety strategies to overcome toxicity. Mol Cancer. 18 (1), 125 (2019).
  22. Dana, H., et al. CAR-T cells: Early successes in blood cancer and challenges in solid tumors. Acta Pharm Sin B. 11 (5), 1129-1147 (2021).
  23. Van Der Schans, J. J., Van De Donk, N., Mutis, T. Dual targeting to overcome current challenges in multiple myeloma CAR T-cell treatment. Front Oncol. 10, 1362 (2020).
  24. Andrea, A. E., Chiron, A., Bessoles, S., Hacein-Bey-Abina, S. Engineering next-generation car-t cells for better toxicity management. Int J Mol Sci. 21 (22), 8620 (2020).
  25. Flugel, C. L., et al. Overcoming on-target, off-tumour toxicity of CAR T cell therapy for solid tumours. Nat Rev Clin Oncol. 20 (1), 49-62 (2023).
  26. Rababah, F., et al. Chimeric antigen receptor T cells therapy in solid tumors. Clin Transl Oncol. 25 (8), 2279-2296 (2023).
  27. Masoud, G. N., Li, W. Hif-1alpha pathway: Role, regulation and intervention for cancer therapy. Acta Pharm Sin B. 5 (5), 378-389 (2015).
  28. Semenza, G. L. Targeting hif-1 for cancer therapy. Nat Rev Cancer. 3 (10), 721-732 (2003).
  29. Harris, A. L. Hypoxia–a key regulatory factor in tumour growth. Nat Rev Cancer. 2 (1), 38-47 (2002).
  30. Juillerat, A., et al. An oxygen sensitive self-decision making engineered CAR T-cell. Sci Rep. 7, 39833 (2017).
  31. Liao, Q., et al. Engineering T cells with hypoxia-inducible chimeric antigen receptor (HiCAR) for selective tumor killing. Biomark Res. 8 (1), 56 (2020).
  32. Ivan, M., et al. Hifα targeted for vhl-mediated destruction by proline hydroxylation: Implications for o2 sensing. Science. 292 (5516), 464-468 (2001).
  33. Wang, G. L., Semenza, G. L. Purification and characterization of hypoxia-inducible factor 1. J Biol Chem. 270 (3), 1230-1237 (1995).
  34. D’alonzo, R. A., et al. In vivo noninvasive preclinical tumor hypoxia imaging methods: A review. Int J Radiat Biol. 97 (5), 593-631 (2021).
  35. He, H., et al. Conditioned car-t cells by hypoxia-inducible transcription amplification (hita) system significantly enhances systemic safety and retains antitumor efficacy. J Immunother Cancer. 9 (10), e002755 (2021).
  36. Barsoum, I. B., Smallwood, C. A., Siemens, D. R., Graham, C. H. A mechanism of hypoxia-mediated escape from adaptive immunity in cancer cells. Cancer Res. 74 (3), 665-674 (2014).
  37. Kroemer, G., Pouyssegur, J. Tumor cell metabolism: Cancer’s achilles’ heel. Cancer Cell. 13 (6), 472-482 (2008).
This article has been published
Video Coming Soon
Keep me updated:

.

Cite This Article
Xue, Y., Mao, Y., Liao, Q., Zhao, C., Zhang, X., Xu, J. Generation and Functional Verification of Hypoxia-Sensitive Chimeric Antigen Receptor-T Cells. J. Vis. Exp. (208), e66697, doi:10.3791/66697 (2024).

View Video