Autologous Dendritic Cells in Combination With Chemotherapy Restore Responsiveness of T Cells in Breast Cancer Patients: A Single-Arm Phase I/II Trial

David A Bernal-Estévez, Mauren A Ortíz Barbosa, Paola Ortíz-Montero, Claudia Cifuentes, Ramiro Sánchez, Carlos A Parra-López, David A Bernal-Estévez, Mauren A Ortíz Barbosa, Paola Ortíz-Montero, Claudia Cifuentes, Ramiro Sánchez, Carlos A Parra-López

Abstract

Introduction: Animal studies and preclinical studies in cancer patients suggest that the induction of immunogenic cell death (ICD) by neoadjuvant chemotherapy with doxorubicin and cyclophosphamide (NAC-AC) recovers the functional performance of the immune system. This could favor immunotherapy schemes such as the administration of antigen-free autologous dendritic cells (DCs) in combination with NAC-AC to profit as cryptic vaccine immunogenicity of treated tumors.

Objective: To explore the safety and immunogenicity of autologous antigen-free DCs administered to breast cancer patients (BCPs) in combination with NAC-AC.

Materials and methods: A phase I/II cohort clinical trial was performed with 20 BCPs treated with NAC-AC [nine who received DCs and 11 who did not (control group)]. The occurrence of adverse effects and the functional performance of lymphocytes from BCPs before and after four cycles of NAC-AC receiving DCs or not were assessed using flow cytometry and compared with that from healthy donors (HDs). Flow cytometry analysis using manual and automated algorithms led us to examine functional performance and frequency of different lymphocyte compartments in response to a stimulus in vitro. This study was registered at clinicaltrials.gov (NCT03450044).

Results: No grade II or higher adverse effects were observed associated with the transfer of DCs to patients during NAC-AC. Interestingly, in response to the in vitro stimulation, deficient phosphorylation of Zap70 and AKT proteins observed before chemotherapy in most patients' CD4 T cells significantly recovered after NAC-AC only in patients who received DCs.

Conclusions: The transfer of autologous DCs in combination with NAC-AC in BCPs is a safe procedure. That, in BCPs, the administration of DCs in combination with NAC-AC favors the recovery of the functional capacity of T cells suggests that this combination may potentiate the adjuvant effect of ICD induced by NAC-AC on T cells and, hence, potentiate the immunogenicity of tumors as cryptic vaccines.

Keywords: breast cancer; clinical trial; dendritic cells; immunotherapy; neoadjuvant chemotherapy.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Bernal-Estévez, Ortíz Barbosa, Ortíz-Montero, Cifuentes, Sánchez and Parra-López.

Figures

Figure 1
Figure 1
Vaccination scheme and dendritic cell (DC) characterization. (A) Patient interventions, before NAC, patients have gone to an apheresis collection to obtain peripheral blood mononuclear cells (PBMCs) and therefore induce monocyte-derived DCs (cryopreserved until use). After four doses of chemotherapy, a new blood sample was taken to compare between pre- and post-chemotherapy. (B) Representative histograms of immature DCs (red), alpha DCs (blue), and standard DCs (yellow), comparing the expression levels of CD86, CD209, CCR7, CD80, CD83, and HLA-DR. (C) Clinical trial scheme per patient in weeks, week 0 is referred to the first dose of chemotherapy, the process starts with the evaluation criteria for patients (w-2), apheresis (vaccinated group) or blood sample (control group) was taken in w-1. Between doses of chemotherapy, we transferred two doses of DCs for a total of six doses. One week after the fourth dose of A/C, we collect the second blood sample.
Figure 2
Figure 2
Restoration of T-cell early activation profile after neoadjuvant chemotherapy with A/C (NAC) + dendritic cell (DC) therapy. (A) Representative contour plots of CD69 vs. CD3 before and after in vitro stimulation of peripheral blood mononuclear cells (PBMCs) obtained from healthy donors (HDs) and patients before (PRE) and post-chemotherapy alone [non-vaccinated (NV)] and vaccinated patients (VAC), numbers represent percentage of cells in each quadrant. (B) Mean expression of surface CD3 (normalized compared to control samples and represented over a 100%) in HDs (red bar), before chemotherapy (PRE, black bar), non-vaccinated (NV, yellow bar), and vaccinated (VAC, blue bar) patients. (C) Quantification of intracellular CD3 (normalized to control samples and represented over a 100%) in the four groups described in panel (B). (D) percentage of CD69+CD3low cells in the four groups of samples before stimulation (empty bars) and after in vitro stimulation (tinted bars). Non-parametric Mann–Whitney test between populations or groups. *p < 0.05, **p < 0.01.
Figure 3
Figure 3
NAC plus dendritic cell (DC) therapy restores T-cell function reflected by the increasing phosphorylation of ZAP70, AKT, and mammalian target of rapamycin (mTOR). Relative expression of p-ZAP70, p-AKT, and p-mTOR in CD4+ and CD8+ T cells, central memory T cells (TCM), and terminal effector T cells (TEF). The expression was normalized based on the median fluorescent intensity (MFI) of each molecule in non-stimulated peripheral blood mononuclear cells (PBMCs) over stimulated cells (represented over 100%) in the four groups, patients before chemotherapy (PRE, black bars), post-chemotherapy alone [non-vaccinated (NV), yellow bars], vaccinated patients (VAC, blue bars), and healthy donors (HDs, red bars). Non-parametric Mann–Whitney test between populations or groups. *p

Figure 4

Identification of T-cell populations associated…

Figure 4

Identification of T-cell populations associated with dendritic cell (DC) transfer by FlowSOM. FlowSOM…

Figure 4
Identification of T-cell populations associated with dendritic cell (DC) transfer by FlowSOM. FlowSOM analysis of two different staining panels, inhibitory (AD) and T-cell receptor (TCR) activation (EH). tSNE plots of concatenated samples with the relative location of the eight populations determined by FlowSOM with their respective heat map of each marker (A, E). Spanning tree of the FlowSOM populations with their respective pie chart (size of each circle is proportional to cell frequency) (B, F). Distribution of stimulated (red) and control (blue) samples in tSNE plot of each patient group (C, G). Relative cell frequency in each population for the individual groups between stimulated (red bars) vs. control (blue bars) (D, H). Direct comparison of the relative cell frequency among groups for the FlowSOM analysis in inhibitory panel (I, control; J, stimulated) and in the TCR panel (K, control; L, stimulated). Non-parametric Mann–Whitney test between populations or groups. *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001.

Figure 5

Specific population of T cells…

Figure 5

Specific population of T cells are favored by combination therapy NAC + dendritic…

Figure 5
Specific population of T cells are favored by combination therapy NAC + dendritic cells (DCs). CITRUS analysis of peripheral blood mononuclear cell (PBMC) samples obtained from healthy donors (HDs), before chemotherapy (PRE), non-vaccinated (NV), and vaccinated (VAC) patients based on the T-cell receptor (TCR) signaling FC panel. (A) Cluster distribution (hierarchy) and expression level of the corresponding marker from low (blue) to high expression (yellow) for each marker, arrow points at cluster 385357. (B) Histograms depicting the phenotype of cluster 385357 (red histograms) relative to background expression (blue histograms) for each marker (CD3, CD8, CD45RO, CD62L, CD25, and CD69). (C) Relative abundance (Log10) of cluster 385357 in the four groups, patients before chemotherapy (PRE, black bars), post-chemotherapy alone [non-vaccinated (NV), yellow bars], vaccinated patients (VAC, blue bars), HDs (red bars). (D) Cluster hierarchy and expression levels of TCR signaling panel in ex vivo samples, arrows point to clusters 178097 and 178119. (E) Histograms of clusters 178097 and 178119 (red histograms) compared to background expression (blue histograms) in each cluster. (F) Relative expression (Log10) of clusters 178097 and 178119. Non-parametric Mann–Whitney test between populations or groups. *p < 0.05, **p < 0.01.

Figure 6

CDR3 sequences found in tumor-infiltrating…

Figure 6

CDR3 sequences found in tumor-infiltrating lymphocytes correlates with expansion of T cells in…

Figure 6
CDR3 sequences found in tumor-infiltrating lymphocytes correlates with expansion of T cells in a patient after doxorubicin and cyclophosphamide (NAC-AC) plus dendritic cell (DC) treatment. (A) Similarity analysis of the CDR3 sequence by Morisita index 0–1 (no similarity to complete similarity, respectively) represented in a heat map. (B) Venn diagram showing the number of overlapping rearrangements present in the four sequenced samples [tumor, lymph node, and peripheral blood mononuclear cells (PBMCs) before and after NAC+DC vaccination]. (C) Scatter dot plot comparing the frequency of unique sequences with significant differences (increased or decreased) between samples (orange and blue dots), excluded sequences [white dots, below threshold (orange dashed line)] and nonsignificant differences [gray dots, near frequency equality (black dashed line)]. (D) Paired comparison of the frequency of shared rearrangements among the three samples (n = 182), PRE (black dots), VAC (blue dots), and tumor (red dots). Top CDR3 sequences were denoted alongside the tumor CDR3 rearrangement. Parametric one-way ANOVA test with Turkey’s multiple comparison test among the three samples, ****p < 0.0001.
Figure 4
Figure 4
Identification of T-cell populations associated with dendritic cell (DC) transfer by FlowSOM. FlowSOM analysis of two different staining panels, inhibitory (AD) and T-cell receptor (TCR) activation (EH). tSNE plots of concatenated samples with the relative location of the eight populations determined by FlowSOM with their respective heat map of each marker (A, E). Spanning tree of the FlowSOM populations with their respective pie chart (size of each circle is proportional to cell frequency) (B, F). Distribution of stimulated (red) and control (blue) samples in tSNE plot of each patient group (C, G). Relative cell frequency in each population for the individual groups between stimulated (red bars) vs. control (blue bars) (D, H). Direct comparison of the relative cell frequency among groups for the FlowSOM analysis in inhibitory panel (I, control; J, stimulated) and in the TCR panel (K, control; L, stimulated). Non-parametric Mann–Whitney test between populations or groups. *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001.
Figure 5
Figure 5
Specific population of T cells are favored by combination therapy NAC + dendritic cells (DCs). CITRUS analysis of peripheral blood mononuclear cell (PBMC) samples obtained from healthy donors (HDs), before chemotherapy (PRE), non-vaccinated (NV), and vaccinated (VAC) patients based on the T-cell receptor (TCR) signaling FC panel. (A) Cluster distribution (hierarchy) and expression level of the corresponding marker from low (blue) to high expression (yellow) for each marker, arrow points at cluster 385357. (B) Histograms depicting the phenotype of cluster 385357 (red histograms) relative to background expression (blue histograms) for each marker (CD3, CD8, CD45RO, CD62L, CD25, and CD69). (C) Relative abundance (Log10) of cluster 385357 in the four groups, patients before chemotherapy (PRE, black bars), post-chemotherapy alone [non-vaccinated (NV), yellow bars], vaccinated patients (VAC, blue bars), HDs (red bars). (D) Cluster hierarchy and expression levels of TCR signaling panel in ex vivo samples, arrows point to clusters 178097 and 178119. (E) Histograms of clusters 178097 and 178119 (red histograms) compared to background expression (blue histograms) in each cluster. (F) Relative expression (Log10) of clusters 178097 and 178119. Non-parametric Mann–Whitney test between populations or groups. *p < 0.05, **p < 0.01.
Figure 6
Figure 6
CDR3 sequences found in tumor-infiltrating lymphocytes correlates with expansion of T cells in a patient after doxorubicin and cyclophosphamide (NAC-AC) plus dendritic cell (DC) treatment. (A) Similarity analysis of the CDR3 sequence by Morisita index 0–1 (no similarity to complete similarity, respectively) represented in a heat map. (B) Venn diagram showing the number of overlapping rearrangements present in the four sequenced samples [tumor, lymph node, and peripheral blood mononuclear cells (PBMCs) before and after NAC+DC vaccination]. (C) Scatter dot plot comparing the frequency of unique sequences with significant differences (increased or decreased) between samples (orange and blue dots), excluded sequences [white dots, below threshold (orange dashed line)] and nonsignificant differences [gray dots, near frequency equality (black dashed line)]. (D) Paired comparison of the frequency of shared rearrangements among the three samples (n = 182), PRE (black dots), VAC (blue dots), and tumor (red dots). Top CDR3 sequences were denoted alongside the tumor CDR3 rearrangement. Parametric one-way ANOVA test with Turkey’s multiple comparison test among the three samples, ****p < 0.0001.

References

    1. Bertucci F, Gonçalves A. Immunotherapy in Breast Cancer: The Emerging Role of PD-1 and PD-L1. Curr Oncol Rep (2017) 19(10):64. 10.1007/s11912-017-0627-0
    1. Ge Y, Xi H, Ju S, Zhang X. Blockade of PD-1/PD-L1 Immune Checkpoint During DC Vaccination Induces Potent Protective Immunity Against Breast Cancer in Hu-SCID Mice. Cancer Lett (2013) 336(2):253–9. 10.1016/j.canlet.2013.03.010
    1. Koski GK, Koldovsky U, Xu S, Mick R, Sharma A, Fitzpatrick E, et al. . A Novel Dendritic Cell-Based Immunization Approach for the Induction of Durable Th1-Polarized Anti-HER-2/neu Responses in Women With Early Breast Cancer. J Immunother (2012) 35(1):54–65. 10.1097/CJI.0b013e318235f512
    1. Liu L, Wang Y, Miao L, Liu Q, Musetti S, Li J, et al. . Combination Immunotherapy of MUC1 mRNA Nano-Vaccine and CTLA-4 Blockade Effectively Inhibits Growth of Triple Negative Breast Cancer. Mol Ther (2018) 26(1):45–55. 10.1016/j.ymthe.2017.10.020
    1. Obeid M, Panaretakis T, Tesniere A, Joza N, Tufi R, Apetoh L, et al. . Leveraging the Immune System During Chemotherapy: Moving Calreticulin to the Cell Surface Converts Apoptotic Death From “Silent” to Immunogenic. Cancer Res (2007) 67(17):7941–4. 10.1158/0008-5472.CAN-07-1622
    1. Aoto K, Mimura K, Okayama H, Saito M, Chida S, Noda M, et al. . Immunogenic Tumor Cell Death Induced by Chemotherapy in Patients With Breast Cancer and Esophageal Squamous Cell Carcinoma. Oncol Rep (2018) 39(1):151–9. 10.3892/or.2017.6097
    1. Rapoport BL, Anderson R. Realizing the Clinical Potential of Immunogenic Cell Death in Cancer Chemotherapy and Radiotherapy. Int J Mol Sci (2019) 20(4):949. 10.3390/ijms20040959
    1. Galluzzi L, Buqué A, Kepp O, Zitvogel L, Kroemer G. Immunogenic Cell Death in Cancer and Infectious Disease. Nat Rev Immunol (2017) 17(2):97–111. 10.1038/nri.2016.107
    1. Bernal-Estevez D, Sanchez R, Tejada RE, Parra-Lopez C. Chemotherapy and Radiation Therapy Elicits Tumor Specific T Cell Responses in a Breast Cancer Patient. BMC Cancer (2016) 16:591. 10.1186/s12885-016-2625-2
    1. Bernal-Estevez DA, Garcia O, Sanchez R, Parra-Lopez CA. Monitoring the Responsiveness of T and Antigen Presenting Cell Compartments in Breast Cancer Patients is Useful to Predict Clinical Tumor Response to Neoadjuvant Chemotherapy. BMC Cancer (2018) 18(1):77. 10.1186/s12885-017-3982-1
    1. Pinzon-Charry A, Ho CS, Maxwell T, McGuckin MA, Schmidt C, Furnival C, et al. . Numerical and Functional Defects of Blood Dendritic Cells in Early- and Late-Stage Breast Cancer. Br J Cancer (2007) 97(9):1251–9. 10.1038/sj.bjc.6604018
    1. Prado-Garcia H, Romero-Garcia S. The Role of Exhaustion in Tumor-Induced T-Cell Dysfunction in Cancer. Cancer Immunology Springer (2020). p:117–32. 10.1007/978-3-030-30845-2_8
    1. Anguille S, Smits EL, Lion E, van Tendeloo VF, Berneman ZN. Clinical Use of Dendritic Cells for Cancer Therapy. Lancet Oncol (2014) 15(7):e257–67. 10.1016/S1470-2045(13)70585-0
    1. Mailliard RB, Wankowicz-Kalinska A, Cai Q, Wesa A, Hilkens CM, Kapsenberg ML, et al. . Alpha-Type-1 Polarized Dendritic Cells: A Novel Immunization Tool With Optimized CTL-Inducing Activity. Cancer Res (2004) 64(17):5934–7. 10.1158/0008-5472.CAN-04-1261
    1. Czerniecki BJ, Koski GK, Koldovsky U, Xu S, Cohen PA, Mick R, et al. . Targeting HER-2/neu in Early Breast Cancer Development Using Dendritic Cells With Staged Interleukin-12 Burst Secretion. Cancer Res (2007) 67(4):1842–52. 10.1158/0008-5472.CAN-06-4038
    1. Hansen M, Hjorto GM, Donia M, Met O, Larsen NB, Andersen MH, et al. . Comparison of Clinical Grade Type 1 Polarized and Standard Matured Dendritic Cells for Cancer Immunotherapy. Vaccine (2013) 31(4):639–46. 10.1016/j.vaccine.2012.11.053
    1. Wegner J, Hackenberg S, Scholz CJ, Chuvpilo S, Tyrsin D, Matskevich AA, et al. . High-Density Preculture of PBMCs Restores Defective Sensitivity of Circulating CD8 T Cells to Virus- and Tumor-Derived Antigens. Blood (2015) 126(2):185–94. 10.1182/blood-2015-01-622704
    1. Van Gassen S, Callebaut B, Van Helden MJ, Lambrecht BN, Demeester P, Dhaene T, et al. . FlowSOM: Using Self-Organizing Maps for Visualization and Interpretation of Cytometry Data. Cytometry A (2015) 87(7):636–45. 10.1002/cyto.a.22625
    1. Bruggner RV, Bodenmiller B, Dill DL, Tibshirani RJ, Nolan GP. Automated Identification of Stratifying Signatures in Cellular Subpopulations. Proc Natl Acad Sci U S A (2014) 111(26):E2770–7. 10.1073/pnas.1408792111
    1. Team RC. R: A Language and Environment for Statistical Computing. Vienna, Austria: R Foundation for Statistical Computing. (2019).
    1. Ladoire S, Enot D, Andre F, Zitvogel L, Kroemer G. Immunogenic Cell Death-Related Biomarkers: Impact on the Survival of Breast Cancer Patients After Adjuvant Chemotherapy. Oncoimmunology (2016) 5(2):e1082706. 10.1080/2162402X.2015.1082706
    1. Levine JH, Simonds EF, Bendall SC, Davis KL, Amir E-A, Tadmor MD, et al. . Data-Driven Phenotypic Dissection of AML Reveals Progenitor-Like Cells That Correlate With Prognosis. Cell (2015) 162(1):184–97. 10.1016/j.cell.2015.05.047
    1. Lowenfeld L, Mick R, Datta J, Xu S, Fitzpatrick E, Fisher CS, et al. . Dendritic Cell Vaccination Enhances Immune Responses and Induces Regression of HER2(pos) DCIS Independent of Route: Results of Randomized Selection Design Trial. Clin Cancer Res (2017) 23(12):2961–71. 10.1158/1078-0432.CCR-16-1924
    1. Constantino J, Gomes C, Falcao A, Neves BM, Cruz MT. Dendritic Cell-Based Immunotherapy: A Basic Review and Recent Advances. Immunol Res (2017) 65(4):798–810. 10.1007/s12026-017-8931-1
    1. Garg AD, Vara Perez M, Schaaf M, Agostinis P, Zitvogel L, Kroemer G, et al. . Trial Watch: Dendritic Cell-Based Anticancer Immunotherapy. Oncoimmunology (2017) 6(7):e1328341. 10.1080/2162402X.2017.1328341
    1. Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic Cell Death in Cancer Therapy. Annu Rev Immunol (2013) 31:51–72. 10.1146/annurev-immunol-032712-100008
    1. Ho O, Green WR. Alternative Translational Products and Cryptic T Cell Epitopes: Expecting the Unexpected. J Immunol (2006) 177(12):8283–9. 10.4049/jimmunol.177.12.8283
    1. Gnjatic S, Jäger E, Chen W, Altorki NK, Matsuo M, Lee S-Y, et al. . CD8+ T Cell Responses Against a Dominant Cryptic HLA-A2 Epitope After NY-ESO-1 Peptide Immunization of Cancer Patients. Proc Natl Acad Sci (2002) 99(18):11813–8. 10.1073/pnas.142417699
    1. Ma Y, Kepp O, Ghiringhelli F, Apetoh L, Aymeric L, Locher C, et al. . Chemotherapy and Radiotherapy: Cryptic Anticancer Vaccines. Semin Immunol. Elsevier; (2010) 22(3):113–24. 10.1016/j.smim.2010.03.001
    1. Bernal-Estévez DA, Tovar Murillo DR, Parra-López CA. Functional and Phenotypic Analysis of Two-Day Monocyte-Derived Dendritic Cells Suitable for Immunotherapy Purposes. SOJ Immunol (2016) 4(2):1–16. 10.15226/2372-0948/4/2/00153
    1. Dauer M, Schad K, Herten J, Junkmann J, Bauer C, Kiefl R, et al. . FastDC Derived From Human Monocytes Within 48 H Effectively Prime Tumor Antigen-Specific Cytotoxic T Cells. J Immunol Methods (2005) 302(1-2):145–55. 10.1016/j.jim.2005.05.010
    1. Testi R, D’Ambrosio D, De Maria R, Santoni A. The CD69 Receptor: A Multipurpose Cell-Surface Trigger for Hematopoietic Cells. Immunol Today (1994) 15(10):479–83. 10.1016/0167-5699(94)90193-7
    1. Cibrian D, Sanchez-Madrid F. CD69: From Activation Marker to Metabolic Gatekeeper. Eur J Immunol (2017) 47(6):946–53. 10.1002/eji.201646837
    1. Ji C, Yang B, Yang YL, He SH, Miao DS, He L, et al. . Exogenous Cell-Permeable C6 Ceramide Sensitizes Multiple Cancer Cell Lines to Doxorubicin-Induced Apoptosis by Promoting AMPK Activation and Mtorc1 Inhibition. Oncogene (2010) 29(50):6557–68. 10.1038/onc.2010.379
    1. Mineharu Y, Kamran N, Lowenstein PR, Castro MG. Blockade of mTOR Signaling via Rapamycin Combined With Immunotherapy Augments Antiglioma Cytotoxic and Memory T-Cell Functions. Mol Cancer Ther (2014) 13(12):3024–36. 10.1158/1535-7163.MCT-14-0400
    1. Norian LA, Rodriguez PC, O’Mara LA, Zabaleta J, Ohoa AC, Cella M, et al. . Tumor-Infiltrating Regulatory Dendritic Cells Inhibit CD8+ T Cell Function Via L-Arginine Metabolism. Cancer Res (2009) 69(7):3086–94. 10.1158/0008-5472.CAN-08-2826
    1. Baselga J, Campone M, Piccart M, Burris HA, III, Rugo HS, Sahmoud T, et al. . Everolimus in Postmenopausal Hormone-Receptor–Positive Advanced Breast Cancer. New Engl J Med (2012) 366(6):520–9. 10.1056/NEJMoa1109653
    1. Wolff AC, Lazar AA, Bondarenko I, Garin AM, Brincat S, Chow L, et al. . Randomized Phase III Placebo-Controlled Trial of Letrozole Plus Oral Temsirolimus as First-Line Endocrine Therapy in Postmenopausal Women With Locally Advanced or Metastatic Breast Cancer. J Clin Oncol (2013) 31(2):195. 10.1200/JCO.2011.38.3331
    1. Paplomata E, O’Regan R. The PI3K/AKT/mTOR Pathway in Breast Cancer: Targets, Trials and Biomarkers. Ther Adv Med Oncol (2014) 6(4):154–66. 10.1177/1758834014530023
    1. Newell EW, Sigal N, Nair N, Kidd BA, Greenberg HB, Davis MM. Combinatorial Tetramer Staining and Mass Cytometry Analysis Facilitate T-Cell Epitope Mapping and Characterization. Nat Biotechnol (2013) 31(7):623–9. 10.1038/nbt.2593
    1. Murphy RF. Automated Identification of Subpopulations in Flow Cytometric List Mode Data Using Cluster Analysis. Cytometry (1985) 6(4):302–9. 10.1002/cyto.990060405
    1. Qiu P, Simonds EF, Bendall SC, Gibbs KD, Jr, Bruggner RV, Linderman MD, et al. . Extracting a Cellular Hierarchy From High-Dimensional Cytometry Data With SPADE. Nat Biotechnol (2011) 29(10):886–91. 10.1038/nbt.1991
    1. Qian Y, Wei C, Eun-Hyung Lee F, Campbell J, Halliley J, Lee JA, et al. . Elucidation of Seventeen Human Peripheral Blood B-Cell Subsets and Quantification of the Tetanus Response Using a Density-Based Method for the Automated Identification of Cell Populations in Multidimensional Flow Cytometry Data. Cytometry B Clin Cytom (2010) 78(Suppl 1):S69–82. 10.1002/cyto.b.20554
    1. Amir el AD, Davis KL, Tadmor MD, Simonds EF, Levine JH, Bendall SC, et al. . viSNE Enables Visualization of High Dimensional Single-Cell Data and Reveals Phenotypic Heterogeneity of Leukemia. Nat Biotechnol (2013) 31(6):545–52. 10.1038/nbt.2594
    1. Shekhar K, Brodin P, Davis MM, Chakraborty AK. Automatic Classification of Cellular Expression by Nonlinear Stochastic Embedding (ACCENSE). Proc Natl Acad Sci U S A (2014) 111(1):202–7. 10.1073/pnas.1321405111
    1. Kidd BA, Wroblewska A, Boland MR, Agudo J, Merad M, Tatonetti NP, et al. . Mapping the Effects of Drugs on the Immune System. Nat Biotechnol (2016) 34(1):47–54. 10.1038/nbt.3367
    1. Gattinoni L, Lugli E, Ji Y, Pos Z, Paulos CM, Quigley MF, et al. . A Human Memory T Cell Subset With Stem Cell-Like Properties. Nat Med (2011) 17(10):1290–7. 10.1038/nm.2446
    1. Poran A, Scherer J, Bushway ME, Besada R, Balogh KN, Wanamaker A, et al. . Combined TCR Repertoire Profiles and Blood Cell Phenotypes Predict Melanoma Patient Response to Personalized Neoantigen Therapy Plus Anti-PD-1. Cell Rep Med (2020) 1(8):100141. 10.1016/j.xcrm.2020.100141

Source: PubMed

3
S'abonner