Tumour organoids and assembloids: Patient-derived cancer avatars for immunotherapy

Jie Mei , Xingjian Liu , Hui-Xiang Tian , Yixuan Chen , Yang Cao , Jun Zeng , Yung-Chiang Liu , Yaping Chen , Yang Gao , Ji-Ye Yin , Peng-Yuan Wang

Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (4) : e1656

PDF
Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (4) : e1656 DOI: 10.1002/ctm2.1656
REVIEW

Tumour organoids and assembloids: Patient-derived cancer avatars for immunotherapy

Author information +
History +
PDF

Abstract

• The current PDTO models have not yet constructed key cellular and noncellular components.

• PDTOs should be expandable and editable.

• PDTOs are promising preclinical models for immunotherapy unless mature PDTOs can be established.

• PDTO biobanks with consensual standards are urgently

Keywords

organoid / assembloid / cell therapy / immunotherapy / PDTO biobank

Cite this article

Download citation ▾
Jie Mei, Xingjian Liu, Hui-Xiang Tian, Yixuan Chen, Yang Cao, Jun Zeng, Yung-Chiang Liu, Yaping Chen, Yang Gao, Ji-Ye Yin, Peng-Yuan Wang. Tumour organoids and assembloids: Patient-derived cancer avatars for immunotherapy. Clinical and Translational Medicine, 2024, 14(4): e1656 DOI:10.1002/ctm2.1656

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

TuvesonD, Clevers H. Cancer modeling meets human organoid technology. Science. 2019;364(6444):952-955.

[2]

AboulkheyrEH, Montazeri L, ArefAR, et al. Personalized cancer medicine: an organoid approach. Trends Biotechnol. 2018;36(4):358-371.

[3]

Bartusik-AebisherD, Chrzanowski G, BoberZ, et al. An analytical study of trastuzumab-dendrimer-fluorine drug delivery system in breast cancer therapy in vitro. Biomed Pharmacother. 2021;133:111053.

[4]

DuttonJS, HinmanSS, KimR, et al. Primary cell-derived intestinal models: recapitulating physiology. Trends Biotechnol. 2019;37(7):744-760.

[5]

FagetJ, ContatC, ZanggerN, et al. RANKL signaling sustains primary tumor growth in genetically engineered mouse models of lung adenocarcinoma. J Thorac Oncol. 2018;13(3):387-398.

[6]

AnnunziatoS, LutzC, HennemanL, et al. In situ CRISPR-Cas9 base editing for the development of genetically engineered mouse models of breast cancer. Embo J. 2020;39(5):e102169.

[7]

ZhanT, Rindtorff N, BetgeJ, et al. CRISPR/Cas9 for cancer research and therapy. Semin Cancer Biol. 2019;55:106-119.

[8]

ChoKW, KimSJ, KimJ, et al. Large scale and integrated platform for digital mass culture of anchorage dependent cells. Nat Commun. 2019;10(1):4824.

[9]

LiuR, MengX, YuX, et al. From 2D to 3D co-culture systems: a review of co-culture models to study the neural cells interaction. Int J Mol Sci. 2022;23(21):13116.

[10]

WuM, YuE, YeK, et al. Evaluation of the effect of fibroblasts on melanoma metastasis using a biomimetic co-culture model. ACS Biomater Sci Eng. 2023;9(5):2347-2361.

[11]

Gomez-RomanN, Stevenson K, GilmourL, et al. A novel 3D human glioblastoma cell culture system for modeling drug and radiation responses. Neuro Oncol. 2017;19(2):229-241.

[12]

SchmittN, JannJC, AltrockE, et al. Preclinical evaluation of eltrombopag in a PDX model of myelodysplastic syndromes. Leukemia. 2022;36(1):236-247.

[13]

Method of the Year 2017: organoids. Nat Methods. 2018;15(1).

[14]

LancasterMA, Knoblich JA. Organogenesis in a dish: modeling development and disease using organoid technologies. Science. 2014;345(6194):1247125.

[15]

SachsN, de Ligt J, KopperO, et al. A living biobank of breast cancer organoids captures disease heterogeneity. Cell. 2018;172(1-2):373-386.

[16]

SatoT, VriesRG, SnippertHJ, et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459(7244):262-265.

[17]

CleversH. Modeling development and disease with organoids. Cell. 2016;165(7):1586-1597.

[18]

LeeKK, McCauley HA, BrodaTR, et al. Human stomach-on-a-chip with luminal flow and peristaltic-like motility. Lab Chip. 2018;18(20):3079-3085.

[19]

RichardsZ, McCrayT, MarsiliJ, et al. Prostate stroma increases the viability and maintains the branching phenotype of human prostate organoids. Iscience. 2019;12:304-317.

[20]

Cruz-AcuñaR, Quirós M, FarkasAE, et al. Synthetic hydrogels for human intestinal organoid generation and colonic wound repair. Nat Cell Biol. 2017;19(11):1326-1335.

[21]

PriorN, InacioP, HuchM. Liver organoids: from basic research to therapeutic applications. Gut. 2019;68(12):2228-2237.

[22]

TakasatoM, ErPX, ChiuHS, et al. Kidney organoids from human iPS cells contain multiple lineages and model human nephrogenesis. Nature. 2015;526(7574):564-568.

[23]

SalahudeenAA, ChoiSS, RustagiA, et al. Progenitor identification and SARS-CoV-2 infection in human distal lung organoids. Nature. 2020;588(7839):670-675.

[24]

ParkDS, KozakiT, TiwariSK, et al. iPS-cell-derived microglia promote brain organoid maturation via cholesterol transfer. Nature. 2023;623(7986):397-405.

[25]

NorrieJL, Nityanandam A, LaiK, et al. Retinoblastoma from human stem cell-derived retinal organoids. Nat Commun. 2021;12(1):4535.

[26]

MunSJ, RyuJS, LeeMO, et al. Generation of expandable human pluripotent stem cell-derived hepatocyte-like liver organoids. J Hepatol. 2019;71(5):970-985.

[27]

CorròC, Novellasdemunt L, LiV. A brief history of organoids. Am J Physiol Cell Physiol. 2020;319(1):C151-C165.

[28]

QuJ, Kalyani FS, LiuL, et al. Tumor organoids: synergistic applications, current challenges, and future prospects in cancer therapy. Cancer Commun (Lond). 2021;41(12):1331-1353.

[29]

SatoT, StangeDE, FerranteM, et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett's epithelium. Gastroenterology. 2011;141(5):1762-1772.

[30]

DonzelliS, CioceM, SacconiA, et al. A PIK3CA-mutant breast cancer metastatic patient-derived organoid approach to evaluate alpelisib treatment for multiple secondary lesions. Mol Cancer. 2022;21(1):152.

[31]

LeeSH, HuW, MatulayJT, et al. Tumor evolution and drug response in patient-derived organoid models of bladder cancer. Cell. 2018;173(2):515-528.

[32]

LoYH, KolahiKS, DuY, et al. A CRISPR/Cas9-engineered ARID1A-deficient human gastric cancer organoid model reveals essential and nonessential modes of oncogenic transformation. Cancer Discov. 2021;11(6):1562-1581.

[33]

LiX, Francies HE, SecrierM, et al. Organoid cultures recapitulate esophageal adenocarcinoma heterogeneity providing a model for clonality studies and precision therapeutics. Nat Commun. 2018;9(1):2983.

[34]

KimM, MunH, SungCO, et al. Patient-derived lung cancer organoids as in vitro cancer models for therapeutic screening. Nat Commun. 2019;10(1):3991.

[35]

LiZ, XuH, YuL, et al. Patient-derived renal cell carcinoma organoids for personalized cancer therapy. Clin Transl Med. 2022;12(7):e970.

[36]

WangHM, ZhangCY, PengKC, et al. Using patient-derived organoids to predict locally advanced or metastatic lung cancer tumor response: a real-world study. Cell Rep Med. 2023;4(2):100911.

[37]

KimE, ChoiS, KangB, et al. Creation of bladder assembloids mimicking tissue regeneration and cancer. Nature. 2020;588(7839):664-669.

[38]

PașcaSP, Arlotta P, BateupHS, et al. A nomenclature consensus for nervous system organoids and assembloids. Nature. 2022;609(7929):907-910.

[39]

AndersenJ, RevahO, MiuraY, et al. Generation of functional human 3D cortico-motor assembloids. Cell. 2020;183(7):1913-1929.

[40]

LinM, HartlK, HeubergerJ, et al. Establishment of gastrointestinal assembloids to study the interplay between epithelial crypts and their mesenchymal niche. Nat Commun. 2023;14(1):3025.

[41]

WangL, Sievert D, ClarkAE, et al. A human three-dimensional neural-perivascular ‘assembloid’ promotes astrocytic development and enables modeling of SARS-CoV-2 neuropathology. Nat Med. 2021;27(9):1600-1606.

[42]

AiZ, NiuB, YinY, et al. Dissecting peri-implantation development using cultured human embryos and embryo-like assembloids. Cell Res. 2023;33(9):661-678.

[43]

LvQ, WangY, XiongZ, et al. Microvascularized tumor assembloids model for drug delivery evaluation colorectal cancer-derived peritoneal metastasis. Acta Biomater. 2023;168:346-360.

[44]

ZhuY, ZhangX, SunL, et al. Engineering human brain assembloids by microfluidics. Adv Mater. 2023;35(14):e2210083.

[45]

QuF, BroughSC, MichnoW, et al. Crosstalk between small-cell lung cancer cells and astrocytes mimics brain development to promote brain metastasis. Nat Cell Biol. 2023;25(10):1506-1519.

[46]

RothJG, BrunelLG, HuangMS, et al. Spatially controlled construction of assembloids using bioprinting. Nat Commun. 2023;14(1):4346.

[47]

DongH, LiZ, BianS, et al. Culture of patient-derived multicellular clusters in suspended hydrogel capsules for pre-clinical personalized drug screening. Bioact Mater. 2022;18:164-177.

[48]

DaytonTL, AlcalaN, MoonenL, et al. Druggable growth dependencies and tumor evolution analysis in patient-derived organoids of neuroendocrine neoplasms from multiple body sites. Cancer Cell. 2023;41(12):2083-2099.

[49]

GuillenKP, FujitaM, ButterfieldAJ, et al. A human breast cancer-derived xenograft and organoid platform for drug discovery and precision oncology. Nat Cancer. 2022;3(2):232-250.

[50]

JungYH, ChoiDH, ParkK, et al. Drug screening by uniform patient derived colorectal cancer hydro-organoids. Biomaterials. 2021;276:121004.

[51]

MosqueraMJ, KimS, BarejaR, et al. Extracellular matrix in synthetic hydrogel-based prostate cancer organoids regulate therapeutic response to EZH2 and DRD2 inhibitors. Adv Mater. 2022;34(2):e2100096.

[52]

MahadevanKK, McAndrews KM, LeBleuVS, et al. KRAS(G12D) inhibition reprograms the microenvironment of early and advanced pancreatic cancer to promote FAS-mediated killing by CD8(+) T cells. Cancer Cell. 2023;41(9):1606-1620.

[53]

ChakrabartiJ, KohV, SoJ, et al. A preclinical human-derived autologous gastric cancer organoid/immune cell co-culture model to predict the efficacy of targeted therapies. J Vis Exp. 2021(173):e61443.

[54]

RevahO, GoreF, KelleyKW, et al. Maturation and circuit integration of transplanted human cortical organoids. Nature. 2022;610(7931):319-326.

[55]

HwangboH, ChaeS, KimW, et al. Tumor-on-a-chip models combined with mini-tissues or organoids for engineering tumor tissues. Theranostics. 2024;14(1):33-55.

[56]

LiuYC, ChenP, ChangR, et al. Artificial tumor matrices and bioengineered tools for tumoroid generation. Biofabrication. 2024;16(2):022004.

[57]

ShirureVS, BiY, CurtisMB, et al. Tumor-on-a-chip platform to investigate progression and drug sensitivity in cell lines and patient-derived organoids. Lab Chip. 2018;18(23):3687-3702.

[58]

HomanKA, GuptaN, KrollKT, et al. Flow-enhanced vascularization and maturation of kidney organoids in vitro. Nat Methods. 2019;16(3):255-262.

[59]

HoferM, LutolfMP. Engineering organoids. Nat Rev Mater. 2021;6(5):402-420.

[60]

JinBJ, Battula S, ZachosN, et al. Microfluidics platform for measurement of volume changes in immobilized intestinal enteroids. Biomicrofluidics. 2014;8(2):24106.

[61]

SchusterB, JunkinM, KashafSS, et al. Automated microfluidic platform for dynamic and combinatorial drug screening of tumor organoids. Nat Commun. 2020;11(1):5271.

[62]

YaqubN, WayneG, BirchallM, et al. Recent advances in human respiratory epithelium models for drug discovery. Biotechnol Adv. 2022;54:107832.

[63]

IzadifarZ, Sontheimer-Phelps A, LubambaBA, et al. Modeling mucus physiology and pathophysiology in human organs-on-chips. Adv Drug Deliv Rev. 2022;191:114542.

[64]

NealJT, LiX, ZhuJ, et al. Organoid modeling of the tumor immune microenvironment. Cell. 2018;175(7):1972-1988.

[65]

KirschbaumA, GeisseNC, SisterTJ, et al. Effect of certain folic acid antagonists on transplanted myeloid and lymphoid leukemias of the F strain of mice. Cancer Res. 1950;10(12):762-768.

[66]

BhattS, PiosoMS, OlesinskiEA, et al. Reduced mitochondrial apoptotic priming drives resistance to BH3 mimetics in acute myeloid leukemia. Cancer Cell. 2020;38(6):872-890.

[67]

KarkampounaS, La Manna F, BenjakA, et al. Patient-derived xenografts and organoids model therapy response in prostate cancer. Nat Commun. 2021;12(1):1117.

[68]

BoseS, Clevers H, ShenX. Promises and challenges of organoid-guided precision medicine. Medicine (N Y). 2021;2(9):1011-1026.

[69]

AparicioS, Hidalgo M, KungAL. Examining the utility of patient-derived xenograft mouse models. Nat Rev Cancer. 2015;15(5):311-316.

[70]

HidalgoM, AmantF, BiankinAV, et al. Patient-derived xenograft models: an emerging platform for translational cancer research. Cancer Discov. 2014;4(9):998-1013.

[71]

MoS, TangP, LuoW, et al. Patient-derived organoids from colorectal cancer with paired liver metastasis reveal tumor heterogeneity and predict response to chemotherapy. Adv Sci (Weinh). 2022;9(31):e2204097.

[72]

WeeberF, OoftSN, DijkstraKK, et al. Tumor organoids as a pre-clinical cancer model for drug discovery. Cell Chem Biol. 2017;24(9):1092-1100.

[73]

ChenP, ZhangX, DingR, et al. Patient-derived organoids can guide personalized-therapies for patients with advanced breast cancer. Adv Sci (Weinh). 2021;8(22):e2101176.

[74]

DekkersJF, Wiegerinck CL, de JongeHR, et al. A functional CFTR assay using primary cystic fibrosis intestinal organoids. Nat Med. 2013;19(7):939-945.

[75]

DekkersJF, Berkers G, KruisselbrinkE, et al. Characterizing responses to CFTR-modulating drugs using rectal organoids derived from subjects with cystic fibrosis. Sci Transl Med. 2016;8(344):344ra84.

[76]

DriehuisE, Kretzschmar K, CleversH. Establishment of patient-derived cancer organoids for drug-screening applications. Nat Protoc. 2020;15(10):3380-3409.

[77]

RenX, HuangM, WengW, et al. Personalized drug screening in patient-derived organoids of biliary tract cancer and its clinical application. Cell Rep Med. 2023;4(11):101277.

[78]

WadmanM. FDA no longer needs to require animal tests before human drug trials. Science Scienceinsider; 2023.

[79]

YuYY, ZhuYJ, XiaoZZ, et al. The pivotal application of patient-derived organoid biobanks for personalized treatment of gastrointestinal cancers. Biomark Res. 2022;10(1):73.

[80]

ToshimitsuK, TakanoA, FujiiM, et al. Organoid screening reveals epigenetic vulnerabilities in human colorectal cancer. Nat Chem Biol. 2022;18(6):605-614.

[81]

VlachogiannisG, Hedayat S, VatsiouA, et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science. 2018;359(6378):920-926.

[82]

HerpersB, EppinkB, JamesMI, et al. Functional patient-derived organoid screenings identify MCLA-158 as a therapeutic EGFR × LGR5 bispecific antibody with efficacy in epithelial tumors. Nat Cancer. 2022;3(4):418-436.

[83]

PowleyIR, PatelM, MilesG, et al. Patient-derived explants (PDEs) as a powerful preclinical platform for anti-cancer drug and biomarker discovery. Br J Cancer. 2020;122(6):735-744.

[84]

AlzeebG, MetgesJP, CorcosL, et al. Three-dimensional culture systems in gastric cancer research. Cancers (Basel). 2020;12(10):2800.

[85]

Le CompteM, Cardenas DLHE, PeetersS, et al. Multiparametric tumor organoid drug screening using widefield live-cell imaging for bulk and single-organoid analysis. J Vis Exp. 2022(190).

[86]

LeeHL, ChiouJF, WangPY, et al. Ex vivo expansion and drug sensitivity profiling of circulating tumor cells from patients with small cell lung cancer. Cancers (Basel). 2020;12(11):3394.

[87]

WuYH, HungYP, ChiuNC, et al. Correlation between drug sensitivity profiles of circulating tumour cell-derived organoids and clinical treatment response in patients with pancreatic ductal adenocarcinoma. Eur J Cancer. 2022;166:208-218.

[88]

LinD, ShenL, LuoM, et al. Circulating tumor cells: biology and clinical significance. Signal Transduct Target Ther. 2021;6(1):404.

[89]

LiuJF, Kindelberger D, DoyleC, et al. Predictive value of circulating tumor cells (CTCs) in newly-diagnosed and recurrent ovarian cancer patients. Gynecol Oncol. 2013;131(2):352-356.

[90]

ZhangL, Ridgway LD, WetzelMD, et al. The identification and characterization of breast cancer CTCs competent for brain metastasis. Sci Transl Med. 2013;5(180):180ra48.

[91]

CeganM, Kolostova K, MatkowskiR, et al. In vitro culturing of viable circulating tumor cells of urinary bladder cancer. Int J Clin Exp Pathol. 2014;7(10):7164-7171.

[92]

KolostovaK, BroulM, SchramlJ, et al. Circulating tumor cells in localized prostate cancer: isolation, cultivation in vitro and relationship to T-stage and Gleason score. Anticancer Res. 2014;34(7):3641-3646.

[93]

ZhangZ, Shiratsuchi H, LinJ, et al. Expansion of CTCs from early stage lung cancer patients using a microfluidic co-culture model. Oncotarget. 2014;5(23):12383-12397.

[94]

GaoD, VelaI, SbonerA, et al. Organoid cultures derived from patients with advanced prostate cancer. Cell. 2014;159(1):176-187.

[95]

MoutL, van Dessel LF, KraanJ, et al. Generating human prostate cancer organoids from leukapheresis enriched circulating tumour cells. Eur J Cancer. 2021;150:179-189.

[96]

De AngelisML, Francescangeli F, NicolazzoC, et al. An organoid model of colorectal circulating tumor cells with stem cell features, hybrid EMT state and distinctive therapy response profile. J Exp Clin Cancer Res. 2022;41(1):86.

[97]

Rivera-BáezL, Lohse I, LinE, et al. Expansion of circulating tumor cells from patients with locally advanced pancreatic cancer enable patient derived xenografts and functional studies for personalized medicine. Cancers (Basel). 2020;12(4):1011.

[98]

YangC, XiaBR, JinWL, et al. Circulating tumor cells in precision oncology: clinical applications in liquid biopsy and 3D organoid model. Cancer Cell Int. 2019;19:341.

[99]

FaugerouxV, Pailler E, OulhenM, et al. Genetic characterization of a unique neuroendocrine transdifferentiation prostate circulating tumor cell-derived eXplant model. Nat Commun. 2020;11(1):1884.

[100]

TayounT, Faugeroux V, OulhenM, et al. Targeting genome integrity dysfunctions impedes metastatic potency in non-small cell lung cancer circulating tumor cell-derived explants. JCI Insight. 2022;7(11):e155804.

[101]

EnkhbatM, LiuY, KimJ, et al. Expansion of rare cancer cells into tumoroids for therapeutic regimen and cancer therapy. Adv Ther (Weinh). 2021;4(7):2100017.

[102]

LinKC, TingLL, ChangCL, et al. Ex vivo expanded circulating tumor cells for clinical anti-cancer drug prediction in patients with head and neck cancer. Cancers (Basel). 2021;13(23):6076.

[103]

WangPY, PingleH, KoeglerP, et al. Self-assembled binary colloidal crystal monolayers as cell culture substrates. J Mater Chem B. 2015;3(12):2545-2552.

[104]

HaratiJ, LiuK, ShahsavaraniH, et al. Defined physicochemical cues steering direct neuronal reprogramming on colloidal self-assembled patterns (cSAPs). ACS Nano. 2022;17(2):1054-1067.

[105]

EnkhbatM, ZhongB, ChangR, et al. Harnessing focal adhesions to accelerate p53 accumulation and anoikis of A549 cells using colloidal self-assembled patterns (cSAPs). ACS Appl Bio Mater. 2022;5(1):322-333.

[106]

LiuY, Burnouf T, ChangC, et al. Treatment response prediction with circulating tumor cell-derived organoids for soft tissue sarcoma. J Clin Oncol. 2023;41(16_suppl):e23521.

[107]

LiuY, ChenY, ChenS, et al. Abstract 6723: application of in vitro drug screening of circulating tumor cells in pediatric glioma therapy. Cancer Res. 2023;83(7_Supplement):6723.

[108]

WuYH, ChaoHS, ChiangCL, et al. Personalized cancer avatars for patients with thymic malignancies: a pilot study with circulating tumor cell-derived organoids. Thorac Cancer. 2023;14(25):2591-2600.

[109]

OkamotoT, Natsume Y, DoiM, et al. Integration of human inspection and artificial intelligence-based morphological typing of patient-derived organoids reveals interpatient heterogeneity of colorectal cancer. Cancer Sci. 2022;113(8):2693-2703.

[110]

HuangXZ, PangMJ, LiJY, et al. Single-cell sequencing of ascites fluid illustrates heterogeneity and therapy-induced evolution during gastric cancer peritoneal metastasis. Nat Commun. 2023;14(1):822.

[111]

IovannaJ. Implementing biological markers as a tool to guide clinical care of patients with pancreatic cancer. Transl Oncol. 2021;14(1):100965.

[112]

ZhouX, QuM, TebonP, et al. Screening cancer immunotherapy: when engineering approaches meet artificial intelligence. Adv Sci (Weinh). 2020;7(19):2001447.

[113]

GajewskiTF, Schreiber H, FuYX. Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol. 2013;14(10):1014-1022.

[114]

WuQ, YouL, NepovimovaE, et al. Hypoxia-inducible factors: master regulators of hypoxic tumor immune escape. J Hematol Oncol. 2022;15(1):77.

[115]

ZhengH, WuL, XiaoQ, et al. Epigenetically suppressed tumor cell intrinsic STING promotes tumor immune escape. Biomed Pharmacother. 2023;157:114033.

[116]

CerezoM, Guemiri R, DruillennecS, et al. Translational control of tumor immune escape via the eIF4F-STAT1-PD-L1 axis in melanoma. Nat Med. 2018;24(12):1877-1886.

[117]

Casanova-AcebesM, Dalla E, LeaderAM, et al. Tissue-resident macrophages provide a pro-tumorigenic niche to early NSCLC cells. Nature. 2021;595(7868):578-584.

[118]

ShiQ, ShenQ, LiuY, et al. Increased glucose metabolism in TAMs fuels O-GlcNAcylation of lysosomal Cathepsin B to promote cancer metastasis and chemoresistance. Cancer Cell. 2022;40(10):1207-1222.

[119]

WangR, SongS, QinJ, et al. Evolution of immune and stromal cell states and ecotypes during gastric adenocarcinoma progression. Cancer Cell. 2023;41(8):1407-1426.

[120]

LiH, YangP, WangJ, et al. HLF regulates ferroptosis, development and chemoresistance of triple-negative breast cancer by activating tumor cell-macrophage crosstalk. J Hematol Oncol. 2022;15(1):2.

[121]

OliveiraG, WuCJ. Dynamics and specificities of T cells in cancer immunotherapy. Nat Rev Cancer. 2023;23(5):295-316.

[122]

TodoT, ItoH, InoY, et al. Intratumoral oncolytic herpes virus G47∆ for residual or recurrent glioblastoma: a phase 2 trial. Nat Med. 2022;28(8):1630-1639.

[123]

ParkS, OckCY, KimH, et al. Artificial intelligence-powered spatial analysis of tumor-infiltrating lymphocytes as complementary biomarker for immune checkpoint inhibition in non-small-cell lung cancer. J Clin Oncol. 2022;40(17):1916-1928.

[124]

BoulchM, CazauxM, Loe-MieY, et al. A cross-talk between CAR T cell subsets and the tumor microenvironment is essential for sustained cytotoxic activity. Sci Immunol. 2021;6(57):eabd4344.

[125]

LiC, ZhouF, WangJ, et al. Novel CD19-specific γ/δ TCR-T cells in relapsed or refractory diffuse large B-cell lymphoma. J Hematol Oncol. 2023;16(1):5.

[126]

SarnaikAA, HamidO, KhushalaniNI, et al. Lifileucel, a tumor-infiltrating lymphocyte therapy, in metastatic melanoma. J Clin Oncol. 2021;39(24):2656-2666.

[127]

NabetBY, QiuY, ShabasonJE, et al. Exosome RNA unshielding couples stromal activation to pattern recognition receptor signaling in cancer. Cell. 2017;170(2):352-366.

[128]

BagchiS, YuanR, EnglemanEG. Immune checkpoint inhibitors for the treatment of cancer: clinical impact and mechanisms of response and resistance. Annu Rev Pathol. 2021;16:223-249.

[129]

WangY, LiuS, YangZ, et al. Anti-PD-1/L1 lead-in before MAPK inhibitor combination maximizes antitumor immunity and efficacy. Cancer Cell. 2021;39(10):1375-1387.

[130]

ZappasodiR, Serganova I, CohenIJ, et al. CTLA-4 blockade drives loss of T(reg) stability in glycolysis-low tumours. Nature. 2021;591(7851):652-658.

[131]

CuriglianoG, Gelderblom H, MachN, et al. Phase I/Ib clinical trial of sabatolimab, an anti-TIM-3 antibody, alone and in combination with spartalizumab, an anti-PD-1 antibody, in advanced solid tumors. Clin Cancer Res. 2021;27(13):3620-3629.

[132]

YangR, SunL, LiCF, et al. Galectin-9 interacts with PD-1 and TIM-3 to regulate T cell death and is a target for cancer immunotherapy. Nat Commun. 2021;12(1):832.

[133]

YuY, ZengD, OuQ, et al. Association of survival and immune-related biomarkers with immunotherapy in patients with non-small cell lung cancer: a meta-analysis and individual patient-level analysis. JAMA Netw Open. 2019;2(7):e196879.

[134]

KraehenbuehlL, WengCH, EghbaliS, et al. Enhancing immunotherapy in cancer by targeting emerging immunomodulatory pathways. Nat Rev Clin Oncol. 2022;19(1):37-50.

[135]

MoradG, Helmink BA, SharmaP, et al. Hallmarks of response, resistance, and toxicity to immune checkpoint blockade. Cell. 2021;184(21):5309-5337.

[136]

ChennamadhavuniA, Abushahin L, JinN, et al. Risk factors and biomarkers for immune-related adverse events: a practical guide to identifying high-risk patients and rechallenging immune checkpoint inhibitors. Front Immunol. 2022;13:779691.

[137]

MokT, WuYL, KudabaI, et al. Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): a randomised, open-label, controlled, phase 3 trial. Lancet. 2019;393(10183):1819-1830.

[138]

HeratiRS, KnorrDA, VellaLA, et al. PD-1 directed immunotherapy alters Tfh and humoral immune responses to seasonal influenza vaccine. Nat Immunol. 2022;23(8):1183-1192.

[139]

MaX, GuoZ, WeiX, et al. Spatial distribution and predictive significance of dendritic cells and macrophages in esophageal cancer treated with combined chemoradiotherapy and PD-1 blockade. Front Immunol. 2021;12:786429.

[140]

DatarI, Sanmamed MF, WangJ, et al. Expression analysis and significance of PD-1, LAG-3, and TIM-3 in human non-small cell lung cancer using spatially resolved and multiparametric single-cell analysis. Clin Cancer Res. 2019;25(15):4663-4673.

[141]

TaubeJM, RomanK, EngleEL, et al. Multi-institutional TSA-amplified Multiplexed Immunofluorescence Reproducibility Evaluation (MITRE) Study. J Immunother Cancer. 2021;9(7):e002197.

[142]

JacobF, MingGL, SongH. Generation and biobanking of patient-derived glioblastoma organoids and their application in CAR T cell testing. Nat Protoc. 2020;15(12):4000-4033.

[143]

JacobF, Salinas RD, ZhangDY, et al. A patient-derived glioblastoma organoid model and biobank recapitulates inter-and intra-tumoral heterogeneity. Cell. 2020;180(1):188-204.

[144]

ChanJD, LaiJ, SlaneyCY, et al. Cellular networks controlling T cell persistence in adoptive cell therapy. Nat Rev Immunol. 2021;21(12):769-784.

[145]

WangZ, CaoYJ. Adoptive cell therapy targeting neoantigens: a frontier for cancer research. Front Immunol. 2020;11:176.

[146]

PaijensST, Vledder A, de BruynM, et al. Tumor-infiltrating lymphocytes in the immunotherapy era. Cell Mol Immunol. 2021;18(4):842-859.

[147]

RohaanMW, BorchTH, van den BergJH, et al. Tumor-infiltrating lymphocyte therapy or ipilimumab in advanced melanoma. N Engl J Med. 2022;387(23):2113-2125.

[148]

WangS, SunJ, ChenK, et al. Perspectives of tumor-infiltrating lymphocyte treatment in solid tumors. BMC Med. 2021;19(1):140.

[149]

MonbergTJ, BorchTH, SvaneIM, et al. TIL therapy: facts and hopes. Clin Cancer Res. 2023;29(17):3275-3283.

[150]

NorbergSM, Hinrichs CS. Engineered T cell therapy for viral and non-viral epithelial cancers. Cancer Cell. 2023;41(1):58-69.

[151]

HendryS, Salgado R, GevaertT, et al. Assessing tumor-infiltrating lymphocytes in solid tumors: a practical review for pathologists and proposal for a standardized method from the International Immuno-Oncology Biomarkers Working Group: Part 2: TILs in Melanoma, Gastrointestinal Tract Carcinomas, Non-Small Cell Lung Carcinoma and Mesothelioma, Endometrial and Ovarian Carcinomas, Squamous Cell Carcinoma of the Head and Neck, Genitourinary Carcinomas, and Primary Brain Tumors. Adv Anat Pathol. 2017;24(6):311-335.

[152]

KazemiMH, SadriM, NajafiA, et al. Tumor-infiltrating lymphocytes for treatment of solid tumors: it takes two to tango?Front Immunol. 2022;13:1018962.

[153]

BrummelK, Eerkens AL, de BruynM, et al. Tumour-infiltrating lymphocytes: from prognosis to treatment selection. Br J Cancer. 2023;128(3):451-458.

[154]

TengF, MengX, KongL, et al. Tumor-infiltrating lymphocytes, forkhead box P3, programmed death ligand-1, and cytotoxic T lymphocyte-associated antigen-4 expressions before and after neoadjuvant chemoradiation in rectal cancer. Transl Res. 2015;166(6):721-732.

[155]

OuL, LiuS, WangH, et al. Patient-derived melanoma organoid models facilitate the assessment of immunotherapies. Ebiomedicine. 2023;92:104614.

[156]

DijkstraKK, Cattaneo CM, WeeberF, et al. Generation of tumor-reactive T cells by co-culture of peripheral blood lymphocytes and tumor organoids. Cell. 2018;174(6):1586-1598.

[157]

HeJ, XiongX, YangH, et al. Defined tumor antigen-specific T cells potentiate personalized TCR-T cell therapy and prediction of immunotherapy response. Cell Res. 2022;32(6):530-542.

[158]

ZhengN, FangJ, XueG, et al. Induction of tumor cell autosis by myxoma virus-infected CAR-T and TCR-T cells to overcome primary and acquired resistance. Cancer Cell. 2022;40(9):973-985.

[159]

BauluE, GardetC, ChuvinN, et al. TCR-engineered T cell therapy in solid tumors: state of the art and perspectives. Sci Adv. 2023;9(7):eadf3700.

[160]

WuJ, ZouZ, LiuY, et al. CRISPR/Cas9-induced structural variations expand in T lymphocytes in vivo. Nucleic Acids Res. 2022;50(19):11128-11137.

[161]

SilvaDN, Chrobok M, RovestiG, et al. Process development for adoptive cell therapy in academia: a pipeline for clinical-scale manufacturing of multiple TCR-T cell products. Front Immunol. 2022;13:896242.

[162]

HammerlD, RiederD, MartensJ, et al. Adoptive T cell therapy: new avenues leading to safe targets and powerful allies. Trends Immunol. 2018;39(11):921-936.

[163]

de RooijM, RemstD, van der SteenDM, et al. A library of cancer testis specific T cell receptors for T cell receptor gene therapy. Mol Ther Oncolytics. 2023;28:1-14.

[164]

XieX, LiX, SongW. Tumor organoid biobank-new platform for medical research. Sci Rep. 2023;13(1):1819.

[165]

NarayanV, Barber-Rotenberg JS, JungIY, et al. PSMA-targeting TGFβ-insensitive armored CAR T cells in metastatic castration-resistant prostate cancer: a phase 1 trial. Nat Med. 2022;28(4):724-734.

[166]

SchubertML, Schmitt M, WangL, et al. Side-effect management of chimeric antigen receptor (CAR) T-cell therapy. Ann Oncol. 2021;32(1):34-48.

[167]

SternerRC, Sterner RM. CAR-T cell therapy: current limitations and potential strategies. Blood Cancer J. 2021;11(4):69.

[168]

PanK, Farrukh H, ChittepuV, et al. CAR race to cancer immunotherapy: from CAR T, CAR NK to CAR macrophage therapy. J Exp Clin Cancer Res. 2022;41(1):119.

[169]

JuneCH, O'Connor RS, KawalekarOU, et al. CAR T cell immunotherapy for human cancer. Science. 2018;359(6382):1361-1365.

[170]

JayaramanJ, Mellody MP, HouAJ, et al. CAR-T design: elements and their synergistic function. Ebiomedicine. 2020;58:102931.

[171]

MailankodyS, MatousJV, ChhabraS, et al. Allogeneic BCMA-targeting CAR T cells in relapsed/refractory multiple myeloma: phase 1 UNIVERSAL trial interim results. Nat Med. 2023;29(2):422-429.

[172]

LinH, ChengJ, MuW, et al. Advances in universal CAR-T cell therapy. Front Immunol. 2021;12:744823.

[173]

GilletteAA, PhamDL, SkalaMC. Touch-free optical technologies to streamline the production of T cell therapies. Curr Opin Biomed Eng. 2023;25:100434.

[174]

HuY, ZhouY, ZhangM, et al. Genetically modified CD7-targeting allogeneic CAR-T cell therapy with enhanced efficacy for relapsed/refractory CD7-positive hematological malignancies: a phase I clinical study. Cell Res. 2022;32(11):995-1007.

[175]

JingR, ScarfoI, NajiaMA, et al. EZH1 repression generates mature iPSC-derived CAR T cells with enhanced antitumor activity. Cell Stem Cell. 2022;29(8):1181-1196.

[176]

PanR, RyanJ, PanD, et al. Augmenting NK cell-based immunotherapy by targeting mitochondrial apoptosis. Cell. 2022;185(9):1521-1538.

[177]

KabelitzD, WeschD, HeW. Perspectives of gammadelta T cells in tumor immunology. Cancer Res. 2007;67(1):5-8.

[178]

YunYS, Hargrove ME, TingCC. In vivo antitumor activity of anti-CD3-induced activated killer cells. Cancer Res. 1989;49(17):4770-4774.

[179]

FayyazF, Yazdanpanah N, RezaeiN. Cytokine-induced killer cells mediated pathways in the treatment of colorectal cancer. Cell Commun Signal. 2022;20(1):41.

[180]

SongH, LiuS, ZhaoZ, et al. Increased cycles of DC/CIK immunotherapy decreases frequency of Tregs in patients with resected NSCLC. Int Immunopharmacol. 2017;52:197-202.

[181]

MuléJJ, ShuS, SchwarzSL, et al. Adoptive immunotherapy of established pulmonary metastases with LAK cells and recombinant interleukin-2. Science. 1984;225(4669):1487-1489.

[182]

WuY, LiJ, JabbarzadehKP, et al. Natural killer cells as a double-edged sword in cancer immunotherapy: a comprehensive review from cytokine therapy to adoptive cell immunotherapy. Pharmacol Res. 2020;155:104691.

[183]

WolfNK, Kissiov DU, RauletDH. Roles of natural killer cells in immunity to cancer, and applications to immunotherapy. Nat Rev Immunol. 2023;23(2):90-105.

[184]

XieG, DongH, LiangY, et al. CAR-NK cells: a promising cellular immunotherapy for cancer. Ebiomedicine. 2020;59:102975.

[185]

FDA clears IND application for CAR natural killer cell therapy to treat solid tumors. Healio; 2021.

[186]

HsuHJ, TungCP, YuCM, et al. Eradicating mesothelin-positive human gastric and pancreatic tumors in xenograft models with optimized anti-mesothelin antibody-drug conjugates from synthetic antibody libraries. Sci Rep. 2021;11(1):15430.

[187]

HwangHJ, OhMS, LeeDW, et al. Multiplex quantitative analysis of stroma-mediated cancer cell invasion, matrix remodeling, and drug response in a 3D co-culture model of pancreatic tumor spheroids and stellate cells. J Exp Clin Cancer Res. 2019;38(1):258.

[188]

LeBlancVG, TrinhDL, AslanpourS, et al. Single-cell landscapes of primary glioblastomas and matched explants and cell lines show variable retention of inter-and intratumor heterogeneity. Cancer Cell. 2022;40(4):379-392.

[189]

CourauT, Bonnereau J, ChicoteauJ, et al. Cocultures of human colorectal tumor spheroids with immune cells reveal the therapeutic potential of MICA/B and NKG2A targeting for cancer treatment. J Immunother Cancer. 2019;7(1):74.

[190]

CollinsA, MilesGJ, WoodJ, et al. Patient-derived explants, xenografts and organoids: 3-dimensional patient-relevant pre-clinical models in endometrial cancer. Gynecol Oncol. 2020;156(1):251-259.

[191]

van RenterghemA, van de Haar J, VoestEE. Functional precision oncology using patient-derived assays: bridging genotype and phenotype. Nat Rev Clin Oncol. 2023;20(5):305-317.

[192]

ShafiAA, Schiewer MJ, de LeeuwR, et al. Patient-derived models reveal impact of the tumor microenvironment on therapeutic response. Eur Urol Oncol. 2018;1(4):325-337.

[193]

BellomoG, RainerC, QuarantaV, et al. Chemotherapy-induced infiltration of neutrophils promotes pancreatic cancer metastasis via Gas6/AXL signalling axis. Gut. 2022;71(11):2284-2299.

[194]

TieuT, IraniS, BremertKL, et al. Patient-derived prostate cancer explants: a clinically relevant model to assess siRNA-based nanomedicines. Adv Healthc Mater. 2021;10(6):e2001594.

[195]

WuKZ, AdineC, MitriashkinA, et al. Making in vitro tumor models whole again. Adv Healthc Mater. 2023;12(14):e2202279.

[196]

LeeE, LeeEA, KongE, et al. An agonistic anti-Tie2 antibody suppresses the normal-to-tumor vascular transition in the glioblastoma invasion zone. Exp Mol Med. 2023;55(2):470-484.

[197]

ParikhAY, MasiR, GasmiB, et al. Using patient-derived tumor organoids from common epithelial cancers to analyze personalized T-cell responses to neoantigens. Cancer Immunol Immunother. 2023;72(10):3149-3162.

[198]

GrönholmM, Feodoroff M, AntignaniG, et al. Patient-derived organoids for precision cancer immunotherapy. Cancer Res. 2021;81(12):3149-3155.

[199]

GellerLT, Barzily-Rokni M, DaninoT, et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science. 2017;357(6356):1156-1160.

[200]

GurusamyD, CleverD, EilR, et al. Novel “elements” of immune suppression within the tumor microenvironment. Cancer Immunol Res. 2017;5(6):426-433.

[201]

TangQ, YangS, HeG, et al. Tumor-derived exosomes in the cancer immune microenvironment and cancer immunotherapy. Cancer Lett. 2022;548:215823.

[202]

PodazaE, KuoHH, NguyenJ, et al. Next generation patient derived tumor organoids. Transl Res. 2022;250:84-97.

[203]

LeSavageBL, SuharRA, BroguiereN, et al. Next-generation cancer organoids. Nat Mater. 2022;21(2):143-159.

[204]

NejmanD, Livyatan I, FuksG, et al. The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Science. 2020;368(6494):973-980.

[205]

HuangY, DaiY, LiM, et al. Exposure to cadmium induces neuroinflammation and impairs ciliogenesis in hESC-derived 3D cerebral organoids. Sci Total Environ. 2021;797:149043.

[206]

KimDK, JeongJ, LeeDS, et al. PD-L1-directed PlGF/VEGF blockade synergizes with chemotherapy by targeting CD141(+) cancer-associated fibroblasts in pancreatic cancer. Nat Commun. 2022;13(1):6292.

[207]

QiaoL, HuS, HuangK, et al. Tumor cell-derived exosomes home to their cells of origin and can be used as Trojan horses to deliver cancer drugs. Theranostics. 2020;10(8):3474-3487.

[208]

KimJW, NamSA, YiJ, et al. Kidney decellularized extracellular matrix enhanced the vascularization and maturation of human kidney organoids. Adv Sci (Weinh). 2022;9(15):e2103526.

[209]

SmoleA, BentonA, PoussinMA, et al. Expression of inducible factors reprograms CAR-T cells for enhanced function and safety. Cancer Cell. 2022;40(12):1470-1487.

[210]

FoySP, JacobyK, BotaDA, et al. Non-viral precision T cell receptor replacement for personalized cell therapy. Nature. 2023;615(7953):687-696.

[211]

RoperJ, Tammela T, CetinbasNM, et al. In vivo genome editing and organoid transplantation models of colorectal cancer and metastasis. Nat Biotechnol. 2017;35(6):569-576.

[212]

ChenB, Scurrah CR, McKinleyET, et al. Differential pre-malignant programs and microenvironment chart distinct paths to malignancy in human colorectal polyps. Cell. 2021;184(26):6262-6280.

[213]

MarseeA, RoosF, VerstegenM, et al. Building consensus on definition and nomenclature of hepatic, pancreatic, and biliary organoids. Cell Stem Cell. 2021;28(5):816-832.

[214]

SrivastavaV, HuyckeTR, PhongKT, et al. Organoid models for mammary gland dynamics and breast cancer. Curr Opin Cell Biol. 2020;66:51-58.

[215]

KrollKT, MataMM, HomanKA, et al. Immune-infiltrated kidney organoid-on-chip model for assessing T cell bispecific antibodies. Proc Natl Acad Sci U S A. 2023;120(35):e1989645176.

[216]

DekkersJF, van Vliet EJ, SachsN, et al. Long-term culture, genetic manipulation and xenotransplantation of human normal and breast cancer organoids. Nat Protoc. 2021;16(4):1936-1965.

[217]

DohlmanAB, KlugJ, MeskoM, et al. A pan-cancer mycobiome analysis reveals fungal involvement in gastrointestinal and lung tumors. Cell. 2022;185(20):3807-3822.

[218]

GhaddarB, BiswasA, HarrisC, et al. Tumor microbiome links cellular programs and immunity in pancreatic cancer. Cancer Cell. 2022;40(10):1240-1253.

[219]

HoshinoA, Costa-Silva B, ShenTL, et al. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527(7578):329-335.

[220]

MaiTT, Hamaï A, HienzschA, et al. Salinomycin kills cancer stem cells by sequestering iron in lysosomes. Nat Chem. 2017;9(10):1025-1033.

[221]

GaleanoNJ, WuH, LaCourseKD, et al. Effect of the intratumoral microbiota on spatial and cellular heterogeneity in cancer. Nature. 2022;611(7937):810-817.

[222]

RiquelmeE, ZhangY, ZhangL, et al. Tumor microbiome diversity and composition influence pancreatic cancer outcomes. Cell. 2019;178(4):795-806.

[223]

FuA, YaoB, DongT, et al. Tumor-resident intracellular microbiota promotes metastatic colonization in breast cancer. Cell. 2022;185(8):1356-1372.

[224]

Narunsky-HazizaL, Sepich-Poore GD, LivyatanI, et al. Pan-cancer analyses reveal cancer-type-specific fungal ecologies and bacteriome interactions. Cell. 2022;185(20):3789-3806.

RIGHTS & PERMISSIONS

2024 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

163

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/