Combined treatment with anti‐PSMA CAR NK‐92 cell and anti‐PD‐L1 monoclonal antibody enhances the antitumour efficacy against castration‐resistant prostate cancer

Abstract Background The chimeric antigen receptor NK‐92 (CAR NK‐92) cell targeting the prostate‐specific membrane antigen (PSMA) has shown antitumour effects in castration‐resistant prostate cancer (CRPC). However, the expression changes of programmed death ligand 1 (PD‐L1) and its mechanisms on CAR NK‐92 and CRPC cells and the effect of the anti‐PD‐L1 monoclonal antibody (mAb) on PD‐L1 expressed on CAR NK‐92 cells remain unknown. Methods Human dendritic cells and CD8+ T cells were acquired from blood samples of healthy donors and cocultured with C4‐2 cells. Changes in PD‐L1 expression were detected by flow cytometry. Differential gene expressions were investigated by RNA sequence analysis, while the regulation of PD‐L1 molecular signaling was explored using western blotting. In vitro cytotoxicity was evaluated using the Cell Counting Kit‐8 assay and the bioluminescent intensity (BLI) of green fluorescent protein‐labelled C4‐2 cells. CRPC growth in vivo was monitored using callipers and BLI in male NOD/SCID mice subcutaneously injected with C4‐2 cells and treated intravenously with anti‐PD‐L1/PD‐1 mAb, CAR NK‐92 or cocultured CD8+ T cells. Results Significantly upregulated expression of PD‐L1k was observed in cocultured C4‐2 and CAR NK‐92 cells. In addition, upregulation of PD‐L1 expression was dependent on interferon‐γ in C4‐2 cells, while it was dependent on direct cell‐to‐cell interaction via the NK group 2 member D/ phosphatidylinositol 3‐kinase/AKT pathway in CAR NK‐92 cells. The anti‐PD‐L1 mAb directly acted on PD‐L1 expressed on CAR NK‐92 cells and augmented the cytotoxicity of CAR NK‐92 cells against C4‐2 and CRPC cells from one patient in vitro. Anti‐PD‐L1 mAb significantly enhanced the antitumour effect of CAR NK‐92 cells against CRPC cells in vivo when compared to treatment with CAR NK‐92 cells or combined with anti‐PD‐1 mAb in the absence or presence of cocultured CD8+ T cells. Conclusion Combined treatment with CAR NK‐92 and anti‐PD‐L1 mAb improved the antitumour efficacy against CRPC, which is of extraordinary translational value in the clinical treatment of CRPC.

[1]  N. Xing,et al.  Allogeneic Expanded Human Peripheral NK Cells Control Prostate Cancer Growth in a Preclinical Mouse Model of Castration-Resistant Prostate Cancer , 2022, Journal of immunology research.

[2]  A. Horowitz,et al.  NK cells seize PD1 from leukaemia cells. , 2021, Nature reviews. Immunology.

[3]  Zhenhuan Guo,et al.  Sodium fluoride activates the extrinsic apoptosis via regulating NOX4/ROS-mediated p53/DR5 signaling pathway in lung cells both in vitro and in vivo. , 2021, Free radical biology & medicine.

[4]  T. Xiang,et al.  Digital RNA-seq transcriptome plus tissue anatomy analyses reveal the developmental mechanism of the calabash-shaped root in Tetrastigma hemsleyanum. , 2021, Tree physiology.

[5]  A. Jemal,et al.  Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries , 2021, CA: a cancer journal for clinicians.

[6]  Kongming Wu,et al.  Regulation of PD-L1 expression in the tumor microenvironment , 2021, Journal of Hematology & Oncology.

[7]  T. Song,et al.  SARS-CoV-2 induced intestinal responses with a biomimetic human gut-on-chip , 2020, Science Bulletin.

[8]  J. Qin,et al.  Biomimetic Human Disease Model of SARS‐CoV‐2‐Induced Lung Injury and Immune Responses on Organ Chip System , 2020, Advanced science.

[9]  Jianzhu Chen,et al.  CAR-NK cells: A promising cellular immunotherapy for cancer , 2020, EBioMedicine.

[10]  A. Sharpe,et al.  When killers become thieves: Trogocytosed PD-1 inhibits NK cells in cancer , 2020, bioRxiv.

[11]  T. Cathomen,et al.  PSMA-Directed CAR T Cells Combined with Low-Dose Docetaxel Treatment Induce Tumor Regression in a Prostate Cancer Xenograft Model , 2020, Molecular therapy oncolytics.

[12]  A. Rosato,et al.  Anti-PSMA CAR-Engineered NK-92 Cells: An Off-the-Shelf Cell Therapy for Prostate Cancer , 2020, Cells.

[13]  C. Klein,et al.  Dendritic cells dictate responses to PD-L1 blockade cancer immunotherapy , 2020, Science Translational Medicine.

[14]  Kwok-Kin Wong,et al.  PD-L1 engagement on T cells promotes self-tolerance and suppression of neighboring macrophages and effector T cells in cancer , 2020, Nature Immunology.

[15]  R. Garje,et al.  Review of Indications of FDA-Approved Immune Checkpoint Inhibitors per NCCN Guidelines with the Level of Evidence , 2020, Cancers.

[16]  G. Martinelli,et al.  CAR-T cell therapy: a potential new strategy against prostate cancer , 2019, Journal of Immunotherapy for Cancer.

[17]  R. Schreiber,et al.  Interferon γ and Its Important Roles in Promoting and Inhibiting Spontaneous and Therapeutic Cancer Immunity. , 2019, Cold Spring Harbor perspectives in biology.

[18]  M. Caligiuri,et al.  The mechanism of anti-PD-L1 antibody efficacy against PD-L1 negative tumors identifies NK cells expressing PD-L1 as a cytolytic effector. , 2019, Cancer discovery.

[19]  A. Copik,et al.  PD-L1 blockade enhances anti-tumor efficacy of NK cells , 2018, Oncoimmunology.

[20]  W. Wheat,et al.  Programmed Cell Death Ligand 1 (PD-L1) Signaling Regulates Macrophage Proliferation and Activation , 2018, Cancer Immunology Research.

[21]  T. Schumacher,et al.  Regulation and Function of the PD-L1 Checkpoint. , 2018, Immunity.

[22]  Mithat Gonen,et al.  Long‐Term Follow‐up of CD19 CAR Therapy in Acute Lymphoblastic Leukemia , 2018, The New England journal of medicine.

[23]  C. Rudin,et al.  Atezolizumab for the treatment of non-small cell lung cancer , 2017, Expert review of clinical pharmacology.

[24]  R. Madan,et al.  Immunotherapy of Prostate Cancer: Facts and Hopes , 2017, Clinical Cancer Research.

[25]  Mark S. Litwin,et al.  The Diagnosis and Treatment of Prostate Cancer: A Review , 2017, JAMA.

[26]  S. Khozin,et al.  U.S. Food and Drug Administration Approval Summary: Atezolizumab for Metastatic Non–Small Cell Lung Cancer , 2017, Clinical Cancer Research.

[27]  Koichi Araki,et al.  Rescue of exhausted CD8 T cells by PD-1–targeted therapies is CD28-dependent , 2016, Science.

[28]  E. Mardis,et al.  Temporally Distinct PD-L1 Expression by Tumor and Host Cells Contributes to Immune Escape , 2017, Cancer Immunology Research.

[29]  K. Harrington,et al.  Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. , 2016, The New England journal of medicine.

[30]  Romina E Araya,et al.  NK Cells Restrain Spontaneous Antitumor CD8+ T Cell Priming through PD-1/PD-L1 Interactions with Dendritic Cells , 2016, The Journal of Immunology.

[31]  N. Matsumura,et al.  Dual Faces of IFNγ in Cancer Progression: A Role of PD-L1 Induction in the Determination of Pro- and Antitumor Immunity , 2016, Clinical Cancer Research.

[32]  J. Taube,et al.  Control of PD-L1 Expression by Oncogenic Activation of the AKT-mTOR Pathway in Non-Small Cell Lung Cancer. , 2016, Cancer research.

[33]  J. Radford Nivolumab for recurrent squamous-cell carcinoma of the head and neck , 2016, BDJ.

[34]  M. Katsogiannou,et al.  The hallmarks of castration-resistant prostate cancers. , 2015, Cancer treatment reviews.

[35]  A. Rosato,et al.  PSMA-Specific CAR-Engineered T Cells Eradicate Disseminated Prostate Cancer in Preclinical Models , 2014, PloS one.

[36]  Shohei Koyama,et al.  Loss of Lkb1 and Pten leads to lung squamous cell carcinoma with elevated PD-L1 expression. , 2014, Cancer cell.

[37]  P. Sharma,et al.  PD-L1 Expression in Triple-Negative Breast Cancer , 2014, Cancer Immunology Research.

[38]  P. Sharma,et al.  PD-L 1 Expression in Triple-Negative Breast Cancer , 2014 .

[39]  Jason B. Williams,et al.  Up-Regulation of PD-L1, IDO, and Tregs in the Melanoma Tumor Microenvironment Is Driven by CD8+ T Cells , 2013, Science Translational Medicine.

[40]  Jun Hu,et al.  PTEN Loss Increases PD-L1 Protein Expression and Affects the Correlation between PD-L1 Expression and Clinical Parameters in Colorectal Cancer , 2013, PloS one.

[41]  Drew M. Pardoll,et al.  The blockade of immune checkpoints in cancer immunotherapy , 2012, Nature Reviews Cancer.

[42]  D. Weiner,et al.  HIV-Mediated Phosphatidylinositol 3-Kinase/Serine–Threonine Kinase Activation in APCs Leads to Programmed Death-1 Ligand Upregulation and Suppression of HIV-Specific CD8 T Cells , 2011, The Journal of Immunology.

[43]  A. Enk,et al.  ERK/p38 MAP‐kinases and PI3K are involved in the differential regulation of B7‐H1 expression in DC subsets , 2009, European journal of immunology.

[44]  J. Bluestone,et al.  Interactions between programmed death-1 and programmed death ligand-1 promote tolerance by blocking the T cell receptor-induced stop signal , 2009, Nature immunology.

[45]  P. Mischel,et al.  Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma , 2007, Nature Medicine.

[46]  Lieping Chen,et al.  Interferon regulatory factor‐1 is prerequisite to the constitutive expression and IFN‐γ‐induced upregulation of B7‐H1 (CD274) , 2006, FEBS letters.

[47]  L. Platanias Mechanisms of type-I- and type-II-interferon-mediated signalling , 2005, Nature Reviews Immunology.

[48]  Eric Vivier,et al.  Natural Killer Cell Signaling Pathways , 2004, Science.

[49]  M. Salgaller,et al.  Dendritic cells efficiently acquire and present antigen derived from lung cancer cells and induce antigen-specific T-cell responses , 2003, Cancer Immunology, Immunotherapy.

[50]  R. Barth,et al.  Equipotent Generation of Protective Antitumor Immunity by Various Methods of Dendritic Cell Loading With Whole Cell Tumor Antigens , 2001, Journal of immunotherapy.

[51]  A. Mackensen,et al.  Induction and clonal expansion of tumor‐specific cytotoxic T lymphocytes from renal cell carcinoma patients after stimulation with autologous dendritic cells loaded with tumor cells , 2001, International journal of cancer.

[52]  Nina Bhardwaj,et al.  Consequences of cell death: exposure to necrotic tumor cells , 2000 .

[53]  L. Falo,et al.  Physical interaction between dendritic cells and tumor cells results in an immunogen that induces protective and therapeutic tumor rejection. , 1998, Journal of immunology.

[54]  M. Albert,et al.  Dendritic cells acquire antigen from apoptotic cells and induce class I-restricted CTLs , 1998, Nature.