Activation of Adenosine Phosphate Signaling Promotes Antitumor Immunity in Tumor Microenvironment and Facilitate Immunotherapy

Yantao Xu , Ying Wang , Zixi Jiang , Yi He , Guowei Zhou , Benliang Wei , Jiachen Liu , Xiang Chen

MEDCOMM - Oncology ›› 2025, Vol. 4 ›› Issue (2) : e70022

PDF
MEDCOMM - Oncology ›› 2025, Vol. 4 ›› Issue (2) : e70022 DOI: 10.1002/mog2.70022
ORIGINAL ARTICLE

Activation of Adenosine Phosphate Signaling Promotes Antitumor Immunity in Tumor Microenvironment and Facilitate Immunotherapy

Author information +
History +
PDF

Abstract

Adenosine 5′-triphosphate (ATP) plays a crucial role in intracellular energetic metabolism and functions as a signal transducer in shaping the tumor microenvironment (TME). However, the understanding of the biological functions of adenosine phosphate signaling and its clinical relevance remains limited. Here, we deciphered the multi-omics dysregulation of 15 purinergic P2 receptors (P2Rs) and their clinical relevance. We revealed the presence of 5 ATP signaling subtypes in melanoma, with two distinct functional metaprograms—one metabolic and the other inflammatory. We developed an adenosine phosphate signaling model (APsig) that showed promising prognostic value in melanoma, as well as predictive efficacy of immunotherapy across 1068 tumor samples in 9 independent public cohorts. High APsig was associated with longer overall survival (OS) and improved response to tumor immunotherapy. Additionally, through single-cell and spatial transcriptomic analysis, we explored how APsig promotes antitumor immunity by activating myeloid lineage cells for antigen presentation. Our comprehensive characterization of P2R-mediated adenosine phosphate signaling at both bulk/single-cell and spatial transcriptomic levels highlights its potential as a promising target for developing novel anticancer agents, particularly in combination with immune checkpoint inhibitors.

Keywords

acral melanoma / adenosine / adenosine 5′-triphosphate (ATP) / immunotherapy / purinergic / skin cutaneous melanoma

Cite this article

Download citation ▾
Yantao Xu, Ying Wang, Zixi Jiang, Yi He, Guowei Zhou, Benliang Wei, Jiachen Liu, Xiang Chen. Activation of Adenosine Phosphate Signaling Promotes Antitumor Immunity in Tumor Microenvironment and Facilitate Immunotherapy. MEDCOMM - Oncology, 2025, 4(2): e70022 DOI:10.1002/mog2.70022

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

M. Arnold, D. Singh, M. Laversanne, et al., “Global Burden of Cutaneous Melanoma in 2020 and Projections to 2040,” JAMA Dermatology 158, no. 5 (2022): 495–503.

[2]

L. Lin, Z. Li, L. Yan, Y. Liu, H. Yang, and H. Li, “Global, Regional, and National Cancer Incidence and Death for 29 Cancer Groups in 2019 and Trends Analysis of the Global Cancer Burden, 1990-2019,” Journal of Hematology & Oncology 14, no. 1 (2021): 197.

[3]

C. Karimkhani, A. C. Green, T. Nijsten, et al., “The Global Burden of Melanoma: Results From the Global Burden of Disease Study 2015,” British Journal of Dermatology 177, no. 1 (2017): 134–140.

[4]

P. Aggarwal, P. Knabel, and A. B. Fleischer, “United States Burden of Melanoma and Non-Melanoma Skin Cancer From 1990 to 2019,” Journal of the American Academy of Dermatology 85, no. 2 (2021): 388–395.

[5]

T. Dyba, G. Randi, F. Bray, et al., “The European Cancer Burden in 2020: Incidence and Mortality Estimates for 40 Countries and 25 Major Cancers,” European Journal of Cancer 157 (2021): 308–347.

[6]

Y. Wu, Y. Wang, L. Wang, P. Yin, Y. Lin, and M. Zhou, “Burden of Melanoma in China, 1990-2017: Findings From the 2017 Global Burden of Disease Study,” International Journal of Cancer 147, no. 3 (2020): 692–701.

[7]

F. Islami, E. M. Ward, H. Sung, et al., “Annual Report to the Nation on the Status of Cancer, Part 1: National Cancer Statistics,” Journal of the National Cancer Institute 113, no. 12 (2021): 1648–1669.

[8]

W. Guo, H. Wang, and C. Li, “Signal Pathways of Melanoma and Targeted Therapy,” Signal Transduction and Targeted Therapy 6, no. 1 (2021): 424.

[9]

C. Twyman-Saint Victor, A. J. Rech, A. Maity, et al., “Radiation and Dual Checkpoint Blockade Activate Non-Redundant Immune Mechanisms in Cancer,” Nature 520, no. 7547 (2015): 373–377.

[10]

M. Yarchoan, A. Hopkins, and E. M. Jaffee, “Tumor Mutational Burden and Response Rate to PD-1 Inhibition,” New England Journal of Medicine 377, no. 25 (2017): 2500–2501.

[11]

S. A. Weiss, J. D. Wolchok, and M. Sznol, “Immunotherapy of Melanoma: Facts and Hopes,” Clinical Cancer Research 25, no. 17 (2019): 5191–5201.

[12]

X. Chen and E. Song, “The Theory of Tumor Ecosystem,” Cancer Communications 42, no. 7 (2022): 587–608.

[13]

K. Thrane, H. Eriksson, J. Maaskola, J. Hansson, and J. Lundeberg, “Spatially Resolved Transcriptomics Enables Dissection of Genetic Heterogeneity in Stage III Cutaneous Malignant Melanoma,” Cancer Research 78, no. 20 (2018): 5970–5979.

[14]

A. Thyagarajan, K. Y. Tsai, and R. P. Sahu, “MicroRNA Heterogeneity in Melanoma Progression,” Seminars in Cancer Biology 59 (2019): 208–220.

[15]

M. Marzagalli, N. D. Ebelt, and E. R. Manuel, “Unraveling the Crosstalk Between Melanoma and Immune Cells in the Tumor Microenvironment,” Seminars in Cancer Biology 59 (2019): 236–250.

[16]

C. Abildgaard and P. Guldberg, “Molecular Drivers of Cellular Metabolic Reprogramming in Melanoma,” Trends in Molecular Medicine 21, no. 3 (2015): 164–171.

[17]

A. Tasdogan, B. Faubert, V. Ramesh, et al., “Metabolic Heterogeneity Confers Differences in Melanoma Metastatic Potential,” Nature 577, no. 7788 (2020): 115–120.

[18]

Y. Imura, Y. Morizawa, R. Komatsu, et al., “Microglia Release ATP by Exocytosis,” GLIA 61, no. 8 (2013): 1320–1330.

[19]

I. Martins, Y. Wang, M. Michaud, et al., “Molecular Mechanisms of ATP Secretion During Immunogenic Cell Death,” Cell Death & Differentiation 21, no. 1 (2014): 79–91.

[20]

Q. Cao, K. Zhao, X. Z. Zhong, et al., “SLC17A9 Protein Functions as a Lysosomal ATP Transporter and Regulates Cell Viability,” Journal of Biological Chemistry 289, no. 33 (2014): 23189–23199.

[21]

C. E. Stout, J. L. Costantin, C. C. G. Naus, and A. C. Charles, “Intercellular Calcium Signaling in Astrocytes via ATP Release Through Connexin Hemichannels,” Journal of Biological Chemistry 277, no. 12 (2002): 10482–10488.

[22]

J. Kang, N. Kang, D. Lovatt, et al., “Connexin 43 Hemichannels Are Permeable to ATP,” Journal of Neuroscience 28, no. 18 (2008): 4702–4711.

[23]

F. B. Chekeni, M. R. Elliott, J. K. Sandilos, et al., “Pannexin 1 Channels Mediate ‘Find-Me’ Signal Release and Membrane Permeability During Apoptosis,” Nature 467, no. 7317 (2010): 863–867.

[24]

C. B. Medina, P. Mehrotra, S. Arandjelovic, et al., “Metabolites Released From Apoptotic Cells Act as Tissue Messengers,” Nature 580, no. 7801 (2020): 130–135.

[25]

M. Michaud, I. Martins, A. Q. Sukkurwala, et al., “Autophagy-Dependent Anticancer Immune Responses Induced by Chemotherapeutic Agents in Mice,” Science 334, no. 6062 (2011): 1573–1577.

[26]

K. Tatsuno, T. Yamazaki, D. Hanlon, et al., “Extracorporeal Photochemotherapy Induces Bona Fide Immunogenic Cell Death,” Cell Death & Disease 10, no. 8 (2019): 578.

[27]

M. E. Rodriguez-Ruiz, I. Vitale, K. J. Harrington, I. Melero, and L. Galluzzi, “Immunological Impact of Cell Death Signaling Driven by Radiation on the Tumor Microenvironment,” Nature Immunology 21, no. 2 (2020): 120–134.

[28]

O. Kepp, L. Bezu, T. Yamazaki, et al., “ATP and Cancer Immunosurveillance,” EMBO Journal 40, no. 13 (2021): e108130.

[29]

A. M. Chiarella, Y. K. Ryu, G. A. Manji, and A. K. Rustgi, “Extracellular ATP and Adenosine in Cancer Pathogenesis and Treatment,” Trends in Cancer 7, no. 8 (2021): 731–750.

[30]

Y. Ai, H. Wang, L. Liu, et al., “Purine and Purinergic Receptors in Health and Disease,” MedComm 4, no. 5 (2023): e359.

[31]

M. R. Elliott, F. B. Chekeni, P. C. Trampont, et al., “Nucleotides Released by Apoptotic Cells Act as a Find-Me Signal to Promote Phagocytic Clearance,” Nature 461, no. 7261 (2009): 282–286.

[32]

D. G. Perregaux, P. McNiff, R. Laliberte, M. Conklyn, and C. A. Gabel, “ATP Acts as an Agonist to Promote Stimulus-Induced Secretion of IL-1β and IL-18 in Human Blood,” Journal of Immunology 165, no. 8 (2000): 4615–4623.

[33]

D. Schumacher, B. Strilic, K. K. Sivaraj, N. Wettschureck, and S. Offermanns, “Platelet-Derived Nucleotides Promote Tumor-Cell Transendothelial Migration and Metastasis via P2Y2 Receptor,” Cancer Cell 24, no. 1 (2013): 130–137.

[34]

G. Chen, W. Xie, J. Nah, et al., “3,4-Dimethoxychalcone Induces Autophagy Through Activation of the Transcription Factors TFE3 and TFEB,” EMBO Molecular Medicine 11, no. 11 (2019): e10469.

[35]

Y. Wang, W. Xie, J. Humeau, et al., “Autophagy Induction by Thiostrepton Improves the Efficacy of Immunogenic Chemotherapy,” Journal for Immunotherapy of Cancer 8, no. 1 (2020): e000462.

[36]

D. Hanahan and R. A. Weinberg, “Hallmarks of Cancer: The Next Generation,” Cell 144, no. 5 (2011): 646–674.

[37]

M. K. van der Kooij, F. M. Speetjens, S. H. van der Burg, and E. Kapiteijn, “Uveal Versus Cutaneous Melanoma; Same Origin, Very Distinct Tumor Types,” Cancers 11, no. 6 (2019): 845.

[38]

E. P. Hoefsmit, E. A. Rozeman, T. M. Van, et al., “Comprehensive Analysis of Cutaneous and Uveal Melanoma Liver Metastases,” Journal for Immunotherapy of Cancer 8, no. 2 (2020): e001501.

[39]

C. Amalinei, A. Grigoraș, L. Lozneanu, I. D. Căruntu, S. E. Giușcă, and R. A. Balan, “The Interplay Between Tumour Microenvironment Components in Malignant Melanoma,” Medicina 58, no. 3 (2022): 365.

[40]

B. K. Armstrong and C. D. Holman, “Malignant Melanoma of the Skin,” Bulletin of the World Health Organization 65, no. 2 (1987): 245–252.

[41]

A. Bagaev, N. Kotlov, K. Nomie, et al., “Conserved Pan-Cancer Microenvironment Subtypes Predict Response to Immunotherapy,” Cancer Cell 39, no. 6 (2021): 845–865.e7.

[42]

J. Galon and D. Bruni, “Approaches to Treat Immune Hot, Altered and Cold Tumours With Combination Immunotherapies,” Nature Reviews Drug Discovery 18, no. 3 (2019): 197–218.

[43]

Q. Duan, H. Zhang, J. Zheng, and L. Zhang, “Turning Cold Into Hot: Firing Up the Tumor Microenvironment,” Trends in Cancer 6, no. 7 (2020): 605–618.

[44]

H. Jung, H. S. Kim, J. Y. Kim, et al., “DNA Methylation Loss Promotes Immune Evasion of Tumours With High Mutation and Copy Number Load,” Nature Communications 10, no. 1 (2019): 4278.

[45]

T. N. Gide, C. Quek, A. M. Menzies, et al., “Distinct Immune Cell Populations Define Response to Anti-PD-1 Monotherapy and Anti-PD-1/Anti-CTLA-4 Combined Therapy,” Cancer Cell 35, no. 2 (2019): 238–255.e6.

[46]

Y. He, Y. Dong, Y. Chen, et al., “Multi-Omics Characterization and Therapeutic Liability of Ferroptosis in Melanoma,” Signal Transduction and Targeted Therapy 7, no. 1 (2022): 268.

[47]

W. Yang, J. Soares, P. Greninger, et al., “Genomics of Drug Sensitivity in Cancer (GDSC): A Resource for Therapeutic Biomarker Discovery in Cancer Cells,” Nucleic Acids Research 41, no. Database issue (2013): 955–961.

[48]

O. Rozenblatt-Rosen, A. Regev, P. Oberdoerffer, et al., “The Human Tumor Atlas Network: Charting Tumor Transitions Across Space and Time at Single-Cell Resolution,” Cell 181, no. 2 (2020): 236–249.

[49]

X. Ren, L. Zhang, Y. Zhang, Z. Li, N. Siemers, and Z. Zhang, “Insights Gained From Single-Cell Analysis of Immune Cells in the Tumor Microenvironment,” Annual Review of Immunology 39 (2021): 583–609.

[50]

J. Qian, S. Olbrecht, B. Boeckx, et al., “A Pan-Cancer Blueprint of the Heterogeneous Tumor Microenvironment Revealed by Single-Cell Profiling,” Cell Research 30, no. 9 (2020): 745–762.

[51]

L. Zheng, S. Qin, W. Si, et al., “Pan-Cancer Single-Cell Landscape of Tumor-Infiltrating T Cells,” Science 374, no. 6574 (2021): abe6474.

[52]

E. Azizi, A. J. Carr, G. Plitas, et al., “Single-Cell Map of Diverse Immune Phenotypes in the Breast Tumor Microenvironment,” Cell 174, no. 5 (2018): 1293–1308.e36.

[53]

L. Jerby-Arnon, P. Shah, M. S. Cuoco, et al., “A Cancer Cell Program Promotes T Cell Exclusion and Resistance to Checkpoint Blockade,” Cell 175, no. 4 (2018): 984–997.e24.

[54]

C. C. Darmawan, G. Jo, S. E. Montenegro, et al., “Early Detection of Acral Melanoma: A Review of Clinical, Dermoscopic, Histopathologic, and Molecular Characteristics,” Journal of the American Academy of Dermatology 81, no. 3 (2019): 805–812.

[55]

I. Yeh, E. Jorgenson, L. Shen, et al., “Targeted Genomic Profiling of Acral Melanoma,” Journal of the National Cancer Institute 111, no. 10 (2019): 1068–1077.

[56]

J. Li, I. Smalley, Z. Chen, et al., “Single-Cell Characterization of the Cellular Landscape of Acral Melanoma Identifies Novel Targets for Immunotherapy,” Clinical Cancer Research 28, no. 10 (2022): 2131–2146.

[57]

M. Kanehisa, “KEGG: Kyoto Encyclopedia of Genes and Genomes,” Nucleic Acids Research 28, no. 1 (2000): 27–30.

[58]

A. Rao, D. Barkley, G. S. França, and I. Yanai, “Exploring Tissue Architecture Using Spatial Transcriptomics,” Nature 596, no. 7871 (2021): 211–220.

[59]

E. Zhao, M. R. Stone, X. Ren, et al., “Spatial Transcriptomics at Subspot Resolution With BayesSpace,” Nature Biotechnology 39, no. 11 (2021): 1375–1384.

[60]

S. Jin, C. F. Guerrero-Juarez, L. Zhang, et al., “Inference and Analysis of Cell-Cell Communication Using CellChat,” Nature Communications 12, no. 1 (2021): 1088.

[61]

J. Neefjes, M. L. M. Jongsma, P. Paul, and O. Bakke, “Towards a Systems Understanding of MHC Class I and MHC Class II Antigen Presentation,” Nature Reviews Immunology 11, no. 12 (2011): 823–836.

[62]

E. M. Muntjewerff, L. D. Meesters, and G. van den Bogaart, “Antigen Cross-Presentation by Macrophages,” Frontiers in Immunology 11 (2020): 1276.

[63]

J. Qi, H. Sun, Y. Zhang, et al., “Single-Cell and Spatial Analysis Reveal Interaction of FAP(+) Fibroblasts and SPP1(+) Macrophages in Colorectal Cancer,” Nature Communications 13, no. 1 (2022): 1742.

[64]

R. D. Leone and J. D. Powell, “Metabolism of Immune Cells in Cancer,” Nature Reviews Cancer 20, no. 9 (2020): 516–531.

[65]

G. Bergers and S. M. Fendt, “The Metabolism of Cancer Cells During Metastasis,” Nature Reviews Cancer 21, no. 3 (2021): 162–180.

[66]

L. Zheng, X. Zhang, F. Yang, et al., “Regulation of the P2X7R by microRNA-216b in Human Breast Cancer,” Biochemical and Biophysical Research Communications 452, no. 1 (2014): 197–204.

[67]

A. Zech, C. K. Ayata, F. Pankratz, et al., “MicroRNA-155 Modulates P2R Signaling and Th2 Priming of Dendritic Cells During Allergic Airway Inflammation in Mice,” Allergy 70, no. 9 (2015): 1121–1129.

[68]

W. Fu, T. McCormick, X. Qi, et al., “Activation of P2X(7)-Mediated Apoptosis Inhibits DMBA/TPA-Induced Formation of Skin Papillomas and Cancer in Mice,” BMC Cancer 9 (2009): 114.

[69]

B. Jelassi, A. Chantôme, F. Alcaraz-Pérez, et al., “P2X(7) Receptor Activation Enhances SK3 Channels- and Cystein Cathepsin-Dependent Cancer Cells Invasiveness,” Oncogene 30, no. 18 (2011): 2108–2122.

[70]

K. Yang, A. Halima, and T. A. Chan, “Antigen Presentation in Cancer—Mechanisms and Clinical Implications for Immunotherapy,” Nature Reviews Clinical Oncology 20, no. 9 (2023): 604–623.

[71]

S. Jhunjhunwala, C. Hammer, and L. Delamarre, “Antigen Presentation in Cancer: Insights Into Tumour Immunogenicity and Immune Evasion,” Nature Reviews Cancer 21, no. 5 (2021): 298–312.

[72]

A. R. Sánchez-Paulete, A. Teijeira, F. J. Cueto, et al., “Antigen Cross-Presentation and T-Cell Cross-Priming in Cancer Immunology and Immunotherapy,” supplement, Annals of Oncology 28, no. S2 (2017): xii74.

[73]

X. Gong and R. Karchin, “Pan-Cancer HLA Gene-Mediated Tumor Immunogenicity and Immune Evasion,” Molecular Cancer Research 20, no. 8 (2022): 1272–1283.

[74]

J. M. Zaretsky, A. Garcia-Diaz, D. S. Shin, et al., “Mutations Associated With Acquired Resistance to PD-1 Blockade in Melanoma,” New England Journal of Medicine 375, no. 9 (2016): 819–829.

[75]

E. Kotsiliti, “MAPK Inhibition in BRAF(V600E) CRC,” Nature Reviews Gastroenterology & Hepatology 20, no. 4 (2023): 201.

[76]

L. Fong, A. Hotson, J. D. Powderly, et al., “Adenosine 2A Receptor Blockade as an Immunotherapy for Treatment-Refractory Renal Cell Cancer,” Cancer Discovery 10, no. 1 (2020): 40–53.

[77]

B. Sidders, P. Zhang, K. Goodwin, et al., “Adenosine Signaling Is Prognostic for Cancer Outcome and Has Predictive Utility for Immunotherapeutic Response,” Clinical Cancer Research 26, no. 9 (2020): 2176–2187.

[78]

C. Ma, Q. Fu, L. P. Diggs, et al., “Platelets Control Liver Tumor Growth Through P2Y12-Dependent CD40L Release in NAFLD,” Cancer Cell 40, no. 9 (2022): 986–998.e5.

[79]

D. W. Laird and S. Penuela, “Pannexin Biology and Emerging Linkages to Cancer,” Trends in Cancer 7, no. 12 (2021): 1119–1131.

[80]

F. Di Virgilio, A. C. Sarti, S. Falzoni, E. De Marchi, and E. Adinolfi, “Extracellular ATP and P2 Purinergic Signalling in the Tumour Microenvironment,” Nature Reviews Cancer 18, no. 10 (2018): 601–618.

[81]

B. Allard, D. Allard, L. Buisseret, and J. Stagg, “The Adenosine Pathway in Immuno-Oncology,” Nature Reviews Clinical Oncology 17, no. 10 (2020): 611–629.

[82]

A. K. Moesta, X. Y. Li, and M. J. Smyth, “Targeting CD39 in Cancer,” Nature Reviews Immunology 20, no. 12 (2020): 739–755.

[83]

R. Cabrita, M. Lauss, A. Sanna, et al., “Tertiary Lymphoid Structures Improve Immunotherapy and Survival in Melanoma,” Nature 577, no. 7791 (2020): 561–565.

[84]

K. Jayawardana, S. J. Schramm, L. Haydu, et al., “Determination of Prognosis in Metastatic Melanoma Through Integration of Clinico-Pathologic, Mutation, mRNA, microRNA, and Protein Information,” International Journal of Cancer 136, no. 4 (2015): 863–874.

[85]

G. Jönsson, C. Busch, S. Knappskog, et al., “Gene Expression Profiling-Based Identification of Molecular Subtypes in Stage IV Melanomas With Different Clinical Outcome,” Clinical Cancer Research 16, no. 13 (2010): 3356–3367.

[86]

B. Badal, A. Solovyov, S. Di Cecilia, et al., “Transcriptional Dissection of Melanoma Identifies a High-Risk Subtype Underlying TP53 Family Genes and Epigenome Deregulation,” JCI Insight 2, no. 9 (2017): e92102.

[87]

N. Riaz, J. J. Havel, V. Makarov, et al., “Tumor and Microenvironment Evolution During Immunotherapy With Nivolumab,” Cell 171, no. 4 (2017): 934–949.e16.

[88]

I. Tirosh, B. Izar, S. M. Prakadan, et al., “Dissecting the Multicellular Ecosystem of Metastatic Melanoma by Single-Cell RNA-seq,” Science 352, no. 6282 (2016): 189–196.

[89]

R. Gaujoux and C. Seoighe, “A Flexible R Package for Nonnegative Matrix Factorization,” BMC Bioinformatics 11 (2010): 367.

[90]

J. P. Brunet, P. Tamayo, T. R. Golub, and J. P. Mesirov, “Metagenes and Molecular Pattern Discovery Using Matrix Factorization,” Proceedings of the National Academy of Sciences 101, no. 12 (2004): 4164–4169.

[91]

S. Anders and W. Huber, “Differential Expression Analysis for Sequence Count Data,” Genome Biology 11, no. 10 (2010): R106.

[92]

S. Hänzelmann, R. Castelo, and J. Guinney, “GSVA: Gene Set Variation Analysis for Microarray and RNA-seq Data,” BMC Bioinformatics 14 (2013): 7.

[93]

A. Liberzon, C. Birger, H. Thorvaldsdóttir, M. Ghandi, J. P. Mesirov, and P. Tamayo, “The Molecular Signatures Database Hallmark Gene Set Collection,” Cell systems 1, no. 6 (2015): 417–425.

[94]

T. Wu, E. Hu, S. Xu, et al., “clusterProfiler 4.0: A Universal Enrichment Tool for Interpreting Omics Data,” Innovation 2, no. 3 (2021): 100141.

[95]

A. M. Newman, C. L. Liu, M. R. Green, et al., “Robust Enumeration of Cell Subsets From Tissue Expression Profiles,” Nature Methods 12, no. 5 (2015): 453–457.

[96]

Y. Hao, S. Hao, E. Andersen-Nissen, et al., “Integrated Analysis of Multimodal Single-Cell Data,” Cell 184, no. 13 (2021): 3573–3587.e29.

[97]

R. Browaeys, W. Saelens, and Y. Saeys, “NicheNet: Modeling Intercellular Communication by Linking Ligands to Target Genes,” Nature Methods 17, no. 2 (2020): 159–162.

RIGHTS & PERMISSIONS

2025 The Author(s). MedComm – Oncology published by John Wiley & Sons Australia, Ltd on behalf of Sichuan International Medical Exchange & Promotion Association (SCIMEA).

AI Summary AI Mindmap
PDF

14

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/