Spatiotemporal Omics-Refining the landscape of precision medicine
Jiajun Zhang, Jianhua Yin, Yang Heng, Ken Xie, Ao Chen, Ido Amit, Xiu-wu Bian, Xun Xu
Spatiotemporal Omics-Refining the landscape of precision medicine
Current streamline of precision medicine uses histomorphological and molecular information to indicate individual phenotypes and genotypes to achieve optimal outcome of treatment. The knowledge of detected mutations and alteration can hardly describe molecular interaction and biological process which can finally be manifested as a disease. With molecular diagnosis revising the modalities of disease, there is a trend in precision medicine to apply multiomic and multidimensional information to decode tumors, regarding heterogeneity, pathogenesis, prognosis, etc. Emerging state-of-art spatiotemporal omics provides a novel vision for in discovering clinicopathogenesis associated findings, some of which show a promising potential to be translated to facilitate clinical practice. Here, we summarize the available spatiotemporal omic technologies and algorithms, highlight the novel scientific findings and explore potential applications in the clinical scenario. Spatiotemporal omics present the ability to provide impetus to rewrite clinical pathology and to answer outstanding clinical questions. This review emphasizes the novel vision of spatiotemporal omics to refine the landscape of precision medicine in the clinic.
spatiotemporal omics / precision medicine / pathomechanism of disease / spatial algorithms / technologies of spatial omics
[1] |
RobertC. A decade of immune-checkpoint inhibitors in cancer therapy. Nat Commun 2020;11:3801.
CrossRef
Google scholar
|
[2] |
KumarV, AbbasAK, AsterJC. Robbins and Cotran Pathologic Basis of Disease. 9th edn. Philadelphia, PA: Elsevier/Saunders, 2015.
|
[3] |
Dagogo-JackI, ShawAT, ShawAT. Tumour heterogeneity and resistance to cancer therapies. Nat Rev Clin Oncol 2018;15:81–94.
CrossRef
Google scholar
|
[4] |
GartnerLP. Textbook of Histology. 5th edn. Philadelphia, PA: Elsevier, 2021.
|
[5] |
ZhangL, ChenD, SongD, et al. Clinical and translational values of spatial transcriptomics. Signal Transduct Targeted Ther 2022;7:111.
CrossRef
Google scholar
|
[6] |
LewisSM, Asselin-Labat ML, NguyenQ, et al. Spatial omics and multiplexed imaging to explore cancer biology. Nat Methods 2021;18:997–1012.
CrossRef
Google scholar
|
[7] |
WangM, HuQ, LvT, et al. High-resolution 3D spatiotemporal transcriptomic maps of developing Drosophila embryos and larvae. Dev Cell 2022;57:1271–83.e4.
CrossRef
Google scholar
|
[8] |
ChenJ, SuoS, TamPP, et al. Spatial transcriptomic analysis of cryosectioned tissue samples with Geo-seq. Nat Protoc 2017;12:566–80.
CrossRef
Google scholar
|
[9] |
MoffittJR, HaoJ, WangG, et al. High-throughput single-cell gene-expression profiling with multiplexed error-robust fluorescence in situ hybridization. Proc Natl Acad Sci USA 2016;113:11046–51.
CrossRef
Google scholar
|
[10] |
SalmenF, StahlPL, MollbrinkA, et al. Barcoded solid-phase RNA capture for Spatial Transcriptomics profiling in mammalian tissue sections. Nat Protoc 2018;13:2501–34.
CrossRef
Google scholar
|
[11] |
KeR, Mignardi M, PacureanuA, et al. In situ sequencing for RNA analysis in preserved tissue and cells. Nat Methods 2013;10:857–60.
CrossRef
Google scholar
|
[12] |
LiaoJ, LuX, ShaoX, et al. Uncovering an organ’s molecular architecture at single-cell resolution by spatially resolved transcriptomics. Trends Biotechnol 2021;39:43–58.
CrossRef
Google scholar
|
[13] |
MoffittJR, Lundberg E, HeynH. The emerging landscape of spatial profiling technologies. Nat Rev Genet 2022;23:741–59.
CrossRef
Google scholar
|
[14] |
HaoY, HaoS, Andersen-NissenE, et al. Integrated analysis of multimodal single-cell data. Cell 2021;184:3573–87.e29.
CrossRef
Google scholar
|
[15] |
BlackS, Phillips D, HickeyJW, et al. CODEX multiplexed tissue imaging with DNA-conjugated antibodies. Nat Protoc 2021;16:3802–35.
CrossRef
Google scholar
|
[16] |
JhaveriN, Nikulina N, ZongH, et al. Abstract 3877: deep ultrahigh- plex spatial phenotyping of human cancer tissues. Cancer Res 2022;82:3877.
CrossRef
Google scholar
|
[17] |
ChengY, Burrack RM, LiQ. Spatially resolved and highly complexed protein and RNA detection by combining CODEX with RNAscope in situ hybridization. bioRxiv 2022.02.10.479971, 2022.
CrossRef
Google scholar
|
[18] |
HeS, BhattR, BrownC, et al. High-plex multiomic analysis in FFPE at subcellular level by spatial molecular imaging. bioRxiv 2021.11.03.467020, 2022.
CrossRef
Google scholar
|
[19] |
GiesenC, WangHA, SchapiroD, et al. Highly multiplexed imaging of tumor tissues with subcellular resolution by mass cytometry. Nat Methods 2014;11:417–22.
CrossRef
Google scholar
|
[20] |
KerenL, BosseM, MarquezD, et al. A structured tumor-immune microenvironment in triple negative breast cancer revealed by multiplexed ion beam imaging. Cell 2018;174:1373–87.e19.
CrossRef
Google scholar
|
[21] |
SchulzD, Zanotelli VRT, FischerJR, et al. Simultaneous multiplexed imaging of mRNA and proteins with subcellular resolution in breast cancer tissue samples by mass cytometry. Cell Syst 2018;6:25–36.e5.
CrossRef
Google scholar
|
[22] |
LewisSM, Asselin-Labat ML. Spatial omics and multiplexed imaging to explore cancer biology. Nat Methods 2021;18:997–1012.
CrossRef
Google scholar
|
[23] |
ChenA, LiaoS, ChengM, et al. Spatiotemporal transcriptomic atlas of mouse organogenesis using DNA nanoball-patterned arrays. Cell 2022;185:1777–92.e21.
|
[24] |
VickovicS, Lotstedt B, KlughammerJ, et al. SM-Omics is an automated platform for high-throughput spatial multi-omics. Nat Commun 2022;13:795.
CrossRef
Google scholar
|
[25] |
Ben-ChetritN, NiuX, SwettAD, et al. Integrated protein and transcriptome high-throughput spatial profiling. bioRxiv 2022.03.15.484516, 2022.
CrossRef
Google scholar
|
[26] |
ChewJ, Uytingco C, AnaparthyN, et al. Tumor microenvironment characterization using a spatial multiomic assay to simultaneously profile protein and gene expression in FFPE tumors. FASEB J 2022;36.
CrossRef
Google scholar
|
[27] |
LiuY, YangM, DengY, et al. High-spatial-resolution multi-omics sequencing via deterministic barcoding in tissue. Cell 2020;183:1665–81.e18.
CrossRef
Google scholar
|
[28] |
LiuY, DiStasio M, SuG, et al. Spatial-CITE-seq: spatially resolved high-plex protein and whole transcriptome co-mapping. bioRxiv 2022.04.01.486788, 2022.
CrossRef
Google scholar
|
[29] |
DengY, Bartosovic M, MaS, et al. Spatial profiling of chromatin accessibility in mouse and human tissues. Nature 2022;609:375–83.
CrossRef
Google scholar
|
[30] |
DengY, Bartosovic M, KukanjaP, et al. Spatial-CUT&Tag: spatially resolved chromatin modification profiling at the cellular level. Science 2022;375:681–6.
CrossRef
Google scholar
|
[31] |
NichterwitzS, ChenG, Aguila BenitezJ, et al. Laser capture microscopy coupled with Smart-seq2 for precise spatial transcriptomic profiling. Nat Commun 2016;7:12139.
CrossRef
Google scholar
|
[32] |
EllisP, MooreL, SandersMA, et al. Reliable detection of somatic mutations in solid tissues by laser-capture microdissection and low-input DNA sequencing. Nat Protoc 2021;16:841–71.
CrossRef
Google scholar
|
[33] |
LiottaLA, Pappalardo PA, CarpinoA, et al. Laser Capture Proteomics: spatial tissue molecular profiling from the bench to personalized medicine. Expert Rev Proteomics 2021;18:845–61.
CrossRef
Google scholar
|
[34] |
ThorntonCA, Mulqueen RM, TorkenczyKA, et al. Spatially mapped single-cell chromatin accessibility. Nat Commun 2021;12:1274.
CrossRef
Google scholar
|
[35] |
MerrittCR, OngGT, ChurchSE, et al. Multiplex digital spatial profiling of proteins and RNA in fixed tissue. Nat Biotechnol 2020;38:586–99.
CrossRef
Google scholar
|
[36] |
Jerby-ArnonL, NeftelC, ShoreME, et al. Opposing immune and genetic mechanisms shape oncogenic programs in synovial sarcoma. Nat Med 2021;27:289–300.
CrossRef
Google scholar
|
[37] |
DesaiN, NeyazA, SzabolcsA, et al. Temporal and spatial heterogeneity of host response to SARS-CoV-2 pulmonary infection. Nat Commun 2020;11:6319.
CrossRef
Google scholar
|
[38] |
ChoCS, XiJ, SiY, et al. Microscopic examination of spatial transcriptome using Seq-Scope. Cell 2021;184:3559–72.e22.
CrossRef
Google scholar
|
[39] |
FuX, SunL, ChenJY, et al. Continuous polony gels for tissue mapping with high resolution and RNA capture efficiency. bioRxiv 2021.03.17.435795, 2021.
CrossRef
Google scholar
|
[40] |
LiuC, LiR, LiY, et al. Spatiotemporal mapping of gene expression landscapes and developmental trajectories during zebrafish embryogenesis. Dev Cell 2022;57:1284–1298.e5.
CrossRef
Google scholar
|
[41] |
MantriM, Scuderi GJ, Abedini-NassabR, et al. Spatiotemporal single- cell RNA sequencing of developing chicken hearts identifies interplay between cellular differentiation and morphogenesis. Nat Commun 2021;12:1771.
CrossRef
Google scholar
|
[42] |
Ben-MosheS, VegT, MancoR, et al. The spatiotemporal program of zonal liver regeneration following acute injury. Cell Stem Cell 2022;29:973–89.e10.
CrossRef
Google scholar
|
[43] |
ZhaoE, StoneMR, RenX, et al. Spatial transcriptomics at subspot resolution with BayesSpace. Nat Biotechnol 2021;39:1375–84.
CrossRef
Google scholar
|
[44] |
FuH, XuH, ChongK, et al. Unsupervised spatially embedded deep representation of spatial transcriptomics. bioRxiv 2021.06.15.448542, 2021.
CrossRef
Google scholar
|
[45] |
HuJ, LiX, ColemanK, et al. SpaGCN: integrating gene expression, spatial location and histology to identify spatial domains and spatially variable genes by graph convolutional network. Nat Methods 2021;18:1342–51.
CrossRef
Google scholar
|
[46] |
CableDM, MurrayE, ZouLS, et al. Robust decomposition of cell type mixtures in spatial transcriptomics. Nat Biotechnol 2022;40:517–26.
CrossRef
Google scholar
|
[47] |
HirzT, MeiS, SarkarH, et al. Integrated single-cell and spatial transcriptomic analyses unravel the heterogeneity of the prostate tumor microenvironment. 2022.
CrossRef
Google scholar
|
[48] |
KleshchevnikovV, Shmatko A, DannE, et al. Cell2location maps fine-grained cell types in spatial transcriptomics. Nat Biotechnol 2022;40:661–71.
CrossRef
Google scholar
|
[49] |
LopezR, LiB, Keren-ShaulH, et al. Multi-resolution deconvolution of spatial transcriptomics data reveals continuous patterns of inflammation. bioRxiv 2021.05.10.443517, 2021.
CrossRef
Google scholar
|
[50] |
BiancalaniT, ScaliaG, BuffoniL, et al. Deep learning and alignment of spatially resolved single-cell transcriptomes with Tangram. Nat Methods 2021;18:1352–62.
CrossRef
Google scholar
|
[51] |
La MannoG, Siletti K, FurlanA, et al. Molecular architecture of the developing mouse brain. Nature 2021;596:92–6.
CrossRef
Google scholar
|
[52] |
WeiR, HeS, BaiS, et al. Spatial charting of single-cell transcriptomes in tissues. Nat Biotechnol 2022;40:1190–9.
CrossRef
Google scholar
|
[53] |
BaeS, NaKJ, KohJ, et al. CellDART: cell type inference by domain adaptation of single-cell and spatial transcriptomic data. Nucleic Acids Res 2022;50:e57.
CrossRef
Google scholar
|
[54] |
ParkJ-B, ChoiJ-Y, ChoiH, et al. Spatial transcriptomics-based identification of molecular markers for nanomedicine distribution in tumor tissue. bioRxiv 2022.03.02.482584, 2022.
CrossRef
Google scholar
|
[55] |
XuY, McCordRP, McCordRP. CoSTA: unsupervised convolutional neural network learning for spatial transcriptomics analysis. BMC Bioinf 2021;22:397.
CrossRef
Google scholar
|
[56] |
DeTomasoD, YosefN, YosefN. Hotspot identifies informative gene modules across modalities of single-cell genomics. Cell Syst 2021;12:446–56.e9.
CrossRef
Google scholar
|
[57] |
YangD, JonesMG, NaranjoS, et al. Lineage recording reveals the phylodynamics, plasticity and paths of tumor evolution. 2021.
CrossRef
Google scholar
|
[58] |
ChowdhuryS, Ferri-Borgogno S, YangP, et al. Learning directed acyclic graphs for ligands and receptors based on spatially resolved transcriptomic analysis of ovarian cancer. 2021.
CrossRef
Google scholar
|
[59] |
EfremovaM, Vento-Tormo M, TeichmannSA, et al. CellPhoneDB: inferring cell-cell communication from combined expression of multi-subunit ligand-receptor complexes. Nat Protoc 2020;15:1484–506.
CrossRef
Google scholar
|
[60] |
PallaG, Spitzer H, KleinM, et al. Squidpy: a scalable framework for spatial omics analysis. Nat Methods 2022;19:171–8.
CrossRef
Google scholar
|
[61] |
TureiD, Valdeolivas A, GulL, et al. Integrated intra- and intercellular signaling knowledge for multicellular omics analysis. Mol Syst Biol 2021;17:e9923.
CrossRef
Google scholar
|
[62] |
ZhouL, WenR, YuG, et al. Spatial transcriptomic revealed intratumor heterogeneity and cancer stem cell enrichment in colorectal cancer metastasis. 2022.
CrossRef
Google scholar
|
[63] |
ChanJM, Quintanal-Villalonga A, GaoVR, et al. Signatures of plasticity, metastasis, and immunosuppression in an atlas of human small cell lung cancer. Cancer Cell 2021;39:1479–1496.e18.
CrossRef
Google scholar
|
[64] |
TrapnellC, Cacchiarelli D, GrimsbyJ, et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat Biotechnol 2014;32:381–6.
CrossRef
Google scholar
|
[65] |
WolfFA, HameyFK, PlassM, et al. PAGA: graph abstraction reconciles clustering with trajectory inference through a topology preserving map of single cells. Genome Biol 2019;20:59.
CrossRef
Google scholar
|
[66] |
StreetK, RissoD, FletcherRB, et al. Slingshot: cell lineage and pseudotime inference for single-cell transcriptomics. BMC Genom 2018;19:477.
CrossRef
Google scholar
|
[67] |
La MannoG, Soldatov R, ZeiselA, et al. RNA velocity of single cells. Nature 2018;560:494–8.
CrossRef
Google scholar
|
[68] |
BergenV, LangeM, PeidliS, et al. Generalizing RNA velocity to transient cell states through dynamical modeling. Nat Biotechnol 2020;38:1408–14.
CrossRef
Google scholar
|
[69] |
QiuX, ZhangY, Martin-RufinoJD, et al. Mapping transcriptomic vector fields of single cells. Cell 2022;185:690–711.e45.
CrossRef
Google scholar
|
[70] |
QuinnJJ, JonesMG, OkimotoRA, et al. Single-cell lineages reveal the rates, routes, and drivers of metastasis in cancer xenografts. Science 2021;371:eabc1944.
CrossRef
Google scholar
|
[71] |
Figueres-OnateM, Sanchez-Gonzalez R, Lopez-MascaraqueL. Deciphering neural heterogeneity through cell lineage tracing. Cell Mol Life Sci 2021;78:1971–82.
CrossRef
Google scholar
|
[72] |
MatsushitaY, OnoW, OnoN. Synergy of single-cell sequencing analyses and in vivo lineage-tracing approaches: a new opportunity for stem cell biology. Biocell 2022;46:1157–62.
CrossRef
Google scholar
|
[73] |
ChenC, LiaoY, PengG. Connecting past and present: single-cell lineage tracing. Protein Cell 2022;13:790–807.
CrossRef
Google scholar
|
[74] |
Sautès-FridmanC, Petitprez F, CalderaroJ, et al. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat Rev Cancer 2019;19:307–25.
CrossRef
Google scholar
|
[75] |
Dieu-NosjeanMC, GocJ, GiraldoNA, et al. Tertiary lymphoid structures in cancer and beyond. Trends Immunol 2014;35:571–80.
CrossRef
Google scholar
|
[76] |
PitzalisC, JonesGW, BombardieriM, et al. Ectopic lymphoid-like structures in infection, cancer and autoimmunity. Nat Rev Immunol 2014;14:447–62.
CrossRef
Google scholar
|
[77] |
AnderssonA, Larsson L, StenbeckL, et al. Spatial deconvolution of HER2-positive breast cancer delineates tumor-associated cell type interactions. Nat Commun 2021;12:6012.
CrossRef
Google scholar
|
[78] |
MeylanM, Petitprez F, BechtE, et al. Tertiary lymphoid structures generate and propagate anti-tumor antibody-producing plasma cells in renal cell cancer. Immunity 2022;55:527–541.e5.
CrossRef
Google scholar
|
[79] |
WuR, GuoW, QiuX, et al. Comprehensive analysis of spatial architecture in primary liver cancer. Sci Adv 2021;7:eabg3750.
CrossRef
Google scholar
|
[80] |
WuL, YanJ, BaiY, et al. Spatially-resolved transcriptomics analyses of invasive fronts in solid tumors. bioRxiv 2021.10.21.465135, 2021.
CrossRef
Google scholar
|
[81] |
JiAL, RubinAJ, ThraneK, et al. Multimodal analysis of composition and spatial architecture in human squamous cell carcinoma. Cell 2020;182:497–514.e22.
CrossRef
Google scholar
|
[82] |
OmbratoL, NolanE, KurelacI, et al. Metastatic-niche labelling reveals parenchymal cells with stem features. Nature 2019;572:603–8.
CrossRef
Google scholar
|
[83] |
BarkleyD, Moncada R, PourM, et al. Cancer cell states recur across tumor types and form specific interactions with the tumor microenvironment. Nat Genet 2022;54:1192–201.
CrossRef
Google scholar
|
[84] |
DanenbergE, Bardwell H, ZanotelliVRT, et al. Breast tumor microenvironment structures are associated with genomic features and clinical outcome. Nat Genet 2022;54:660–9.
CrossRef
Google scholar
|
[85] |
Cui ZhouD, Jayasinghe RG, ChenS, et al. Spatially restricted drivers and transitional cell populations cooperate with the microenvironment in untreated and chemo-resistant pancreatic cancer. Nat Genet 2022;54:1390–405.
CrossRef
Google scholar
|
[86] |
ChenWT, LuA, CraessaertsK, et al. Spatial transcriptomics and in situ sequencing to study Alzheimer’s disease. Cell 2020;182:976–91.e19.
CrossRef
Google scholar
|
[87] |
RendeiroAF, Ravichandran H, BramY, et al. The spatial landscape of lung pathology during COVID-19 progression. Nature 2021;593:564–9.
CrossRef
Google scholar
|
[88] |
KuppeC, Ramirez Flores RO, LiZ, et al. Spatial multi-omic map of human myocardial infarction. Nature 2022;608:766–77.
CrossRef
Google scholar
|
[89] |
ChungBK, OgaardJ, ReimsHM, et al. Spatial transcriptomics identifies enriched gene expression and cell types in human liver fibrosis. Hepatol Commun 2022;6:2538–50.
CrossRef
Google scholar
|
[90] |
PengL, JinX, LiBY, et al. Integrating single-cell RNA sequencing with spatial transcriptomics reveals immune landscape for interstitial cystitis. Signal Transduct Target Ther 2022;7:161.
CrossRef
Google scholar
|
[91] |
VickovicS, Eraslan G, SalménF, et al. High-definition spatial transcriptomics for in situ tissue profiling. Nat Methods 2019;16:987–90.
CrossRef
Google scholar
|
[92] |
AkbarM, MacDonald L, CroweLAN, et al. Single cell and spatial transcriptomics in human tendon disease indicate dysregulated immune homeostasis. Ann Rheum Dis 2021;80:1494–7.
CrossRef
Google scholar
|
[93] |
RaoA, Barkley D, FrancaGS, et al. Exploring tissue architecture using spatial transcriptomics. Nature 2021;596:211–20.
CrossRef
Google scholar
|
[94] |
BourzacK. Diagnosis: early warning system. Nature 2014;513:S4–6.
CrossRef
Google scholar
|
[95] |
BaronM, TagoreM, HunterMV, et al. The stress-like cancer cell state is a consistent component of tumorigenesis. Cell Syst 2020;11:536–546.e7.
CrossRef
Google scholar
|
[96] |
WangJ, HuangTY, HouY, et al. Epigenomic landscape and 3D genome structure in pediatric high-grade glioma. Sci Adv 2021;7:eabg4126.
CrossRef
Google scholar
|
[97] |
ManiatisS, Äijö T, VickovicS, et al. Spatiotemporal dynamics of molecular pathology in amyotrophic lateral sclerosis. Science 2019;364:89–93.
CrossRef
Google scholar
|
[98] |
YoosufN, Navarro JF, SalménF, et al. Identification and transfer of spatial transcriptomics signatures for cancer diagnosis. Breast Cancer Res 2020;22:1–10.
CrossRef
Google scholar
|
[99] |
YersalO, Barutca S, BarutcaS. Biological subtypes of breast cancer: prognostic and therapeutic implications. World J Clin Oncol 2014;5:412–24.
CrossRef
Google scholar
|
[100] |
MosesL, Pachter L. Museum of spatial transcriptomics. Nat Methods 2022;19:534–46.
CrossRef
Google scholar
|
[101] |
MaynardKR, JaffeAE, MartinowichK. Spatial transcriptomics: putting genome-wide expression on the map. Neuropsychopharmacology 2020;45:232–3.
CrossRef
Google scholar
|
[102] |
LiuS, Kurzrock R, KurzrockR. Toxicity of targeted therapy: implications for response and impact of genetic polymorphisms. Cancer Treat Rev 2014;40:883–91.
CrossRef
Google scholar
|
[103] |
HaslamA, KimMS, PrasadV. Updated estimates of eligibility for and response to genome-targeted oncology drugs among US cancer patients, 2006-2020. Ann Oncol 2021;32:926–32.
CrossRef
Google scholar
|
[104] |
ZhouX, YaoZ, YangH, et al. Are immune-related adverse events associated with the efficacy of immune checkpoint inhibitors in patients with cancer? A systematic review and meta-analysis. BMC Med 2020;18:87.
CrossRef
Google scholar
|
[105] |
DoroshowDB, Sanmamed MF, HastingsK, et al. Immunotherapy in non–small cell lung cancer: facts and hopes. Clin Cancer Res 2019;25:4592–602.
CrossRef
Google scholar
|
[106] |
SchoenfeldAJ, Hellmann MD, HellmannMD. Acquired resistance to immune checkpoint inhibitors. Cancer Cell 2020;37:443–55.
CrossRef
Google scholar
|
[107] |
QiJ, SunH, ZhangY, et al. Single-cell and spatial analysis reveal interaction of FAP(+) fibroblasts and SPP1(+) macrophages in colorectal cancer. Nat Commun 2022;13:1742.
CrossRef
Google scholar
|
[108] |
ZugazagoitiaJ, GuptaS, LiuY, et al. Biomarkers associated with beneficial PD-1 checkpoint blockade in non–small cell lung cancer (NSCLC) identified using high-Plex digital spatial profiling. Clin Cancer Res 2020;26:4360–8.
CrossRef
Google scholar
|
[109] |
ColwellJ. Is PD-L1 expression a biomarker of response? Cancer Discov 2015;5:1232.
CrossRef
Google scholar
|
[110] |
FaresJ, FaresMY, KhachfeHH, et al. Molecular principles of metastasis: a hallmark of cancer revisited. Signal Transduct Target Ther 2020;5:28.
CrossRef
Google scholar
|
[111] |
ZlobecI, BergerMD, LugliA. Tumour budding and its clinical implications in gastrointestinal cancers. Br J Cancer 2020;123:700–8.
CrossRef
Google scholar
|
[112] |
SchurchCM, BhateSS, BarlowGL, et al. Coordinated cellular neighborhoods orchestrate antitumoral immunity at the colorectal cancer invasive front. Cell 2020;182:1341–59.e19.
CrossRef
Google scholar
|
[113] |
JamiesonNB, Legrini A, LeslieH, et al. High-plex proteomic prognostic marker discovery for patients with pancreatic cancer adenocarcinoma using digital spatial profiling. J Clin Oncol 2022;40:611.
CrossRef
Google scholar
|
[114] |
WangY, Kartasalo K, WeitzP, et al. Predicting molecular phenotypes from histopathology images: a transcriptome-wide expression–morphology analysis in breast cancer. Cancer Res 2021;81:5115–26.
CrossRef
Google scholar
|
[115] |
LopesMT. 2017 World Health Organization classification of tumors of the pituitary gland: a summary. Acta Neuropathol 2017;134:521–35.
CrossRef
Google scholar
|
[116] |
Rodriguez-HernandezI, Maiques O, KohlhammerL, et al. WNT11-FZD7-DAAM1 signalling supports tumour initiating abilities and melanoma amoeboid invasion. Nat Commun 2020;11:5315.
CrossRef
Google scholar
|
[117] |
TomeME, Johnson DB, RimszaLM, et al. A redox signature score identifies diffuse large B-cell lymphoma patients with a poor prognosis. Blood 2005;106:3594–601.
CrossRef
Google scholar
|
[118] |
AlmetAA, CangZ, JinS, et al. The landscape of cell–cell communication through single-cell transcriptomics. Curr Opin Syst Biol 2021;26:12–23.
CrossRef
Google scholar
|
/
〈 | 〉 |