Autologous T cell responses to primary human colorectal cancer spheroids are enhanced by ectonucleotidase inhibition

Objective T cells are major effectors of the antitumoural immune response. Their activation by tumour-associated antigens can unleash their proliferation and cytotoxic functions, leading to tumour cell elimination. However, tumour-related immunosuppressive mechanisms including the overexpression of immune checkpoints like programmed cell death protein-1 (PD-1), are also engaged, promoting immune escape. Current immunotherapies targeting these pathways have demonstrated weak efficacy in colorectal cancer (CRC). It is thus crucial to find new targets for immunotherapy in this cancer type. Design In a prospective cohort of patients with CRC, we investigated the phenotype of tumour-related and non-tumour related intestinal T cells (n=44), particularly the adenosinergic pathway, correlating with clinical phenotype. An autologous coculture model was developed between patient-derived primary tumour spheroids and their autologous tumour-associated lymphocytes. We used this relevant model to assess the effects of CD39 blockade on the antitumour T cell response. Results We show the increased expression of CD39, and its co-expression with PD-1, on tumour infiltrating T cells compared with mucosal lymphocytes. CD39 expression was higher in the right colon and early-stage tumours, thus defining a subset of patients potentially responsive to CD39 blockade. Finally, we demonstrate in autologous conditions that CD39 blockade triggers T cell infiltration and tumour spheroid destruction in cocultures. Conclusion In CRC, CD39 is strongly expressed on tumour infiltrating lymphocytes and its inhibition represents a promising therapeutic strategy for treating patients.

[1]  K. Ouyang,et al.  A humanized monoclonal antibody targeting CD39 with novel mechanism for cancer treatment , 2021 .

[2]  G. Coukos,et al.  Probing the killing potency of tumor-infiltrating lymphocytes on microarrayed autologous tumoroids , 2021, bioRxiv.

[3]  P. Gibbs,et al.  Pembrolizumab in Microsatellite-Instability-High Advanced Colorectal Cancer. , 2020, The New England journal of medicine.

[4]  J. Powers,et al.  Targeting Metabolism of Extracellular Nucleotides via Inhibition of Ecto-Nucleotidases CD73 and CD39. , 2020, Journal of medicinal chemistry.

[5]  Matthias P. Lutolf,et al.  High-throughput automated organoid culture via stem-cell aggregation in microcavity arrays , 2020, Nature Biomedical Engineering.

[6]  Yue Zhang,et al.  Advances in immunotherapy for colorectal cancer: a review , 2020, Therapeutic advances in gastroenterology.

[7]  A. Broeks,et al.  Neoadjuvant immunotherapy leads to pathological responses in MMR-proficient and MMR-deficient early-stage colon cancers , 2020, Nature Medicine.

[8]  S. Gruber,et al.  The Prognostic Implications of Tumor Infiltrating Lymphocytes in Colorectal Cancer: A Systematic Review and Meta-Analysis , 2020, Scientific Reports.

[9]  D. Allard,et al.  On the mechanism of anti-CD39 immune checkpoint therapy , 2020, Journal for ImmunoTherapy of Cancer.

[10]  Bridget A. Baxter,et al.  Metabolite profile comparisons between ascending and descending colon tissue in healthy adults , 2020, World journal of gastroenterology.

[11]  Zaozao Wang,et al.  Phenotype molding of T cells in colorectal cancer by single‐cell analysis , 2020, International journal of cancer.

[12]  E. Cuppen,et al.  Patient-derived organoids can predict response to chemotherapy in metastatic colorectal cancer patients , 2019, Science Translational Medicine.

[13]  Song Liu,et al.  Tumor Infiltrating Lymphocytes and Macrophages Improve Survival in Microsatellite Unstable Colorectal Cancer , 2019, Scientific Reports.

[14]  Qin Li,et al.  Mismatch repair deficiency/microsatellite instability-high as a predictor for anti-PD-1/PD-L1 immunotherapy efficacy , 2019, Journal of Hematology & Oncology.

[15]  A. Roussel,et al.  Blocking Antibodies Targeting the CD39/CD73 Immunosuppressive Pathway Unleash Immune Responses in Combination Cancer Therapies. , 2019, Cell reports.

[16]  A. Toubert,et al.  Cocultures of human colorectal tumor spheroids with immune cells reveal the therapeutic potential of MICA/B and NKG2A targeting for cancer treatment , 2019, Journal of Immunotherapy for Cancer.

[17]  B. Gustavsson,et al.  CD39+ regulatory T cells accumulate in colon adenocarcinomas and display markers of increased suppressive function , 2018, Oncotarget.

[18]  M. Najafi,et al.  CD8+ cytotoxic T lymphocytes in cancer immunotherapy: A review , 2018, Journal of cellular physiology.

[19]  Hans Clevers,et al.  Generation of Tumor-Reactive T Cells by Co-culture of Peripheral Blood Lymphocytes and Tumor Organoids , 2018, Cell.

[20]  B. Fox,et al.  Co-expression of CD39 and CD103 identifies tumor-reactive CD8 T cells in human solid tumors , 2018, Nature Communications.

[21]  K. Jirström,et al.  Expression of programmed cell death protein 1 (PD-1) and its ligand PD-L1 in colorectal cancer: Relationship with sidedness and prognosis , 2018, Oncoimmunology.

[22]  M. Fehlings,et al.  Bystander CD8+ T cells are abundant and phenotypically distinct in human tumour infiltrates , 2018, Nature.

[23]  D. Sansom,et al.  CTLA-4: a moving target in immunotherapy. , 2018, Blood.

[24]  Yan Kang,et al.  What Else Can CD39 Tell Us? , 2017, Front. Immunol..

[25]  Xi-chen Zhang,et al.  Bovine Polymorphonuclear Neutrophils Cast Neutrophil Extracellular Traps against the Abortive Parasite Neospora caninum , 2017, Front. Immunol..

[26]  S. M. Toor,et al.  Intratumoral FoxP3+Helios+ Regulatory T Cells Upregulating Immunosuppressive Molecules Are Expanded in Human Colorectal Cancer , 2017, Front. Immunol..

[27]  S. Robson,et al.  The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets , 2017, Immunological reviews.

[28]  I. Pacella,et al.  Regulatory T cells with multiple suppressive and potentially pro-tumor activities accumulate in human colorectal cancer , 2016, Oncoimmunology.

[29]  L. Zitvogel,et al.  Targeting the tumor microenvironment: removing obstruction to anticancer immune responses and immunotherapy. , 2016, Annals of oncology : official journal of the European Society for Medical Oncology.

[30]  Z. Trajanoski,et al.  Integrative Analyses of Colorectal Cancer Show Immunoscore Is a Stronger Predictor of Patient Survival Than Microsatellite Instability. , 2016, Immunity.

[31]  Y. Ben-Neriah,et al.  Inflammatory networks underlying colorectal cancer , 2016, Nature Immunology.

[32]  B. Gustavsson,et al.  Regulatory T Cells from Colon Cancer Patients Inhibit Effector T-cell Migration through an Adenosine-Dependent Mechanism , 2016, Cancer Immunology Research.

[33]  B. Vogelstein,et al.  PD-1 blockade in tumors with mismatch repair deficiency. , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[34]  L. Zitvogel,et al.  Colorectal cancer: the first neoplasia found to be under immunosurveillance and the last one to respond to immunotherapy? , 2015, Oncoimmunology.

[35]  William W. Agace,et al.  Regional specialization within the intestinal immune system , 2014, Nature Reviews Immunology.

[36]  I. Frazer,et al.  The Kinematics of Cytotoxic Lymphocytes Influence Their Ability to Kill Target Cells , 2014, PloS one.

[37]  E. Tartour,et al.  Colorectal cancer and immunity: what we know and perspectives. , 2014, World journal of gastroenterology.

[38]  F. Marincola,et al.  Cancer classification using the Immunoscore: a worldwide task force , 2012, Journal of Translational Medicine.

[39]  David C. Smith,et al.  Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. , 2012, The New England journal of medicine.

[40]  Hans Clevers,et al.  Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett's epithelium. , 2011, Gastroenterology.

[41]  H. Friess,et al.  Impact of CD39 and purinergic signalling on the growth and metastasis of colorectal cancer , 2011, Purinergic Signalling.

[42]  L. Saltz,et al.  Phase II study of the anti-cytotoxic T-lymphocyte-associated antigen 4 monoclonal antibody, tremelimumab, in patients with refractory metastatic colorectal cancer. , 2010, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[43]  G. Fleuren,et al.  Tumor structure and extracellular matrix as a possible barrier for therapeutic approaches using immune cells or adenoviruses in colorectal cancer , 2001, Histochemistry and Cell Biology.