Targeting novel regulated cell death: Ferroptosis, pyroptosis and necroptosis in anti-PD-1/PD-L1 cancer immunotherapy

Li Yu , Ke Huang , Yixiang Liao , Lingzhi Wang , Gautam Sethi , Zhaowu Ma

Cell Proliferation ›› 2024, Vol. 57 ›› Issue (8) : e13644

PDF
Cell Proliferation ›› 2024, Vol. 57 ›› Issue (8) : e13644 DOI: 10.1002/cpr.13644
REVIEW

Targeting novel regulated cell death: Ferroptosis, pyroptosis and necroptosis in anti-PD-1/PD-L1 cancer immunotherapy

Author information +
History +
PDF

Abstract

Chemotherapy, radiotherapy, and immunotherapy represent key tumour treatment strategies. Notably, immune checkpoint inhibitors (ICIs), particularly anti-programmed cell death 1 (PD1) and anti-programmed cell death ligand 1 (PD-L1), have shown clinical efficacy in clinical tumour immunotherapy. However, the limited effectiveness of ICIs is evident due to many cancers exhibiting poor responses to this treatment. An emerging avenue involves triggering non-apoptotic regulated cell death (RCD), a significant mechanism driving cancer cell death in diverse cancer treatments. Recent research demonstrates that combining RCD inducers with ICIs significantly enhances their antitumor efficacy across various cancer types. The use of anti-PD-1/PD-L1 immunotherapy activates CD8+ T cells, prompting the initiation of novel RCD forms, such as ferroptosis, pyroptosis, and necroptosis. However, the functions and mechanisms of non-apoptotic RCD in anti-PD1/PD-L1 therapy remain insufficiently explored. This review summarises the emerging roles of ferroptosis, pyroptosis, and necroptosis in anti-PD1/PD-L1 immunotherapy. It emphasises the synergy between nanomaterials and PD-1/PD-L1 inhibitors to induce non-apoptotic RCD in different cancer types. Furthermore, targeting cell death signalling pathways in combination with anti-PD1/PD-L1 therapies holds promise as a prospective immunotherapy strategy for tumour treatment.

Cite this article

Download citation ▾
Li Yu, Ke Huang, Yixiang Liao, Lingzhi Wang, Gautam Sethi, Zhaowu Ma. Targeting novel regulated cell death: Ferroptosis, pyroptosis and necroptosis in anti-PD-1/PD-L1 cancer immunotherapy. Cell Proliferation, 2024, 57(8): e13644 DOI:10.1002/cpr.13644

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

DesaiAP, Adashek JJ, ReussJE, WestHJ, Mansfield AS. Perioperative immune checkpoint inhibition in early-stage non-small cell lung cancer: a review. JAMA Oncol. 2023;9:135-142.

[2]

EmilojuOE, Sinicrope FA. Neoadjuvant immune checkpoint inhibitor therapy for localized deficient mismatch repair colorectal cancer: a review. JAMA Oncol. 2023;9:1708-1715.

[3]

WrightJJ, PowersAC, JohnsonDB. Endocrine toxicities of immune checkpoint inhibitors. Nat Rev Endocrinol. 2021;17:389-399.

[4]

UpadhayaS, Neftelinov ST, HodgeJ, CampbellJ. Challenges and opportunities in the PD1/PDL1 inhibitor clinical trial landscape. Nat Rev Drug Discov. 2022;21:482-483.

[5]

GuptaB, Sadaria D, WarrierVU, et al. Plant lectins and their usage in preparing targeted nanovaccines for cancer immunotherapy. Semin Cancer Biol. 2022;80:87-106.

[6]

DoroshowDB, BhallaS, BeasleyMB, et al. PD-L1 as a biomarker of response to immune-checkpoint inhibitors. Nat Rev Clin Oncol. 2021;18:345-362.

[7]

KornepatiAVR, Vadlamudi RK, CurielTJ. Programmed death ligand 1 signals in cancer cells. Nat Rev Cancer. 2022;22:174-189.

[8]

KroemerG, Galassi C, ZitvogelL, GalluzziL. Immunogenic cell stress and death. Nat Immunol. 2022;23:487-500.

[9]

StrasserA, VauxDL. Cell death in the origin and treatment of cancer. Mol Cell. 2020;78:1045-1054.

[10]

GalluzziL, VitaleI, AaronsonSA, et al. Molecular mechanisms of cell death: recommendations of the nomenclature committee on cell death 2018. Cell Death Differ. 2018;25:486-541.

[11]

PengF, LiaoM, QinR, et al. Regulated cell death (RCD) in cancer: key pathways and targeted therapies. Signal Transduct Target Ther. 2022;7:286.

[12]

KistM, VucicD. Cell death pathways: intricate connections and disease implications. EMBO J. 2021;40:e106700.

[13]

ZhangC, LiuN. Ferroptosis, necroptosis, and pyroptosis in the occurrence and development of ovarian cancer. Front Immunol. 2022;13:920059.

[14]

ChenX, ZehHJ, KangR, Kroemer G, TangD. Cell death in pancreatic cancer: from pathogenesis to therapy. Nat Rev Gastroenterol Hepatol. 2021;18:804-823.

[15]

GaoW, WangX, ZhouY, Wang X, YuY. Autophagy, ferroptosis, pyroptosis, and necroptosis in tumor immunotherapy. Signal Transduct Target Ther. 2022;7:196.

[16]

NiuX, ChenL, LiY, HuZ, HeF. Ferroptosis, necroptosis, and pyroptosis in the tumor microenvironment: Perspectives for immunotherapy of SCLC. Semin Cancer Biol. 2022;86:273-285.

[17]

TangR, XuJ, ZhangB, et al. Ferroptosis, necroptosis, and pyroptosis in anticancer immunity. J Hematol Oncol. 2020;13:110.

[18]

ZhangH, DaiZ, WuW, et al. Regulatory mechanisms of immune checkpoints PD-L1 and CTLA-4 in cancer. J Exp Clin Cancer Res. 2021;40:184.

[19]

TangL, WangJ, LinN, et al. Immune checkpoint inhibitor-associated colitis: from mechanism to management. Front Immunol. 2021;12:800879.

[20]

RowshanravanB, Halliday N, SansomDM. CTLA-4: a moving target in immunotherapy. Blood. 2018;131:58-67.

[21]

SchachterJ, RibasA, LongGV, et al. Pembrolizumab versus ipilimumab for advanced melanoma: final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006). Lancet. 2017;390:1853-1862.

[22]

BaderJE, VossK, RathmellJC. Targeting metabolism to improve the tumor microenvironment for cancer immunotherapy. Mol Cell. 2020;78:1019-1033.

[23]

LiJ, CheM, ZhangB, Zhao K, WanC, YangK. The association between the neuroendocrine system and the tumor immune microenvironment: emerging directions for cancer immunotherapy. Biochim Biophys Acta Rev Cancer. 2023;1878:189007.

[24]

MayesPA, HanceKW, HoosA. The promise and challenges of immune agonist antibody development in cancer. Nat Rev Drug Discov. 2018;17:509-527.

[25]

RibasA, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 2018;359:1350-1355.

[26]

PardollDM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252-264.

[27]

RobertC. A decade of immune-checkpoint inhibitors in cancer therapy. Nat Commun. 2020;11:3801.

[28]

GongJ, Chehrazi-Raffle A, ReddiS, SalgiaR. Development of PD-1 and PD-L1 inhibitors as a form of cancer immunotherapy: a comprehensive review of registration trials and future considerations. J Immunother Cancer. 2018;6:8.

[29]

ChinaiJM, Janakiram M, ChenF, ChenW, KaplanM, ZangX. New immunotherapies targeting the PD-1 pathway. Trends Pharmacol Sci. 2015;36:587-595.

[30]

WarrierVU, Makandar AI, GargM, et al. Engineering anti-cancer nanovaccine based on antigen cross-presentation. Biosci Rep. 2019;39:BSR20193220.

[31]

Heras-MurilloI, Adán-Barrientos I, GalánM, WculekSK, SanchoD. Dendritic cells as orchestrators of anticancer immunity and immunotherapy. Nat Rev Clin Oncol. 2024;21:257-277.

[32]

VeselyMD, ZhangT, ChenL. Resistance mechanisms to anti-PD cancer immunotherapy. Annu Rev Immunol. 2022;40:45-74.

[33]

XuY, ChenC, GuoY, HuS, SunZ. Effect of CRISPR/Cas9-edited PD-1/PD-L1 on tumor immunity and immunotherapy. Front Immunol. 2022;13:848327.

[34]

HuangQ, WuX, WangZ, et al. The primordial differentiation of tumor-specific memory CD8(+) T cells as bona fide responders to PD-1/PD-L1 blockade in draining lymph nodes. Cell. 2022;185:4049-4066.e25.

[35]

LiZZ, ZhongNN, CaoLM, et al. Nanoparticles targeting lymph nodes for cancer immunotherapy: strategies and influencing factors. Small. 2024;e2308731.doi:10.1002/smll.202308731

[36]

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.

[37]

O’Sullivan CoyneG, MadanRA, GulleyJL. Nivolumab: promising survival signal coupled with limited toxicity raises expectations. J Clin Oncol. 2014;32:986-988.

[38]

WilkinsonE. Nivolumab success in untreated metastatic melanoma. Lancet Oncol. 2015;16:e9.

[39]

LarkinJ, Chiarion-Sileni V, GonzalezR, et al. Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med. 2019;381:1535-1546.

[40]

InmanBA, LongoTA, RamalingamS, Harrison MR. Atezolizumab: a PD-L1-blocking antibody for bladder cancer. Clin Cancer Res. 2017;23:1886-1890.

[41]

SidawayP. Skin cancer: Avelumab effective against Merkel-cell carcinoma. Nat Rev Clin Oncol. 2016;13:652.

[42]

American Association for CancerResearch. Three drugs approved for urothelial carcinoma by FDA. Cancer Discov. 2017;7:659-660.

[43]

DaskivichTJ, Belldegrun A. Words of Wisdom. Re: safety, activity, and immune correlates of anti-PD-1 antibody in cancer. Eur Urol. 2015;67:816-817.

[44]

Ramos-CasalsM, Sisó-Almirall A. Immune-related adverse events of immune checkpoint inhibitors. Ann Intern Med. 2024;177:ITC17-ITC32.

[45]

LiaoP, WangW, WangW, et al. CD8(+) T cells and fatty acids orchestrate tumor ferroptosis and immunity via ACSL4. Cancer Cell. 2022;40:365-378.e6.

[46]

TanY, ChenQ, LiX, et al. Pyroptosis: a new paradigm of cell death for fighting against cancer. J Exp Clin Cancer Res. 2021;40:153.

[47]

BayırH, DixonSJ, TyurinaYY, Kellum JA, KaganVE. Ferroptotic mechanisms and therapeutic targeting of iron metabolism and lipid peroxidation in the kidney. Nat Rev Nephrol. 2023;19:315-336.

[48]

DixonSJ, Lemberg KM, LamprechtMR, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149:1060-1072.

[49]

KaganVE, MaoG, QuF, et al. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat Chem Biol. 2017;13:81-90.

[50]

DixonSJ, Olzmann JA. The cell biology of ferroptosis. Nat Rev Mol Cell Biol. 2024.doi:10.1038/s41580-024-00703-5

[51]

PedreraL, Espiritu RA, RosU, et al. Ferroptotic pores induce Ca(2+) fluxes and ESCRT-III activation to modulate cell death kinetics. Cell Death Differ. 2021;28:1644-1657.

[52]

LiuJ, XiaX, HuangP. xCT: a critical molecule that links cancer metabolism to redox signaling. Mol Ther. 2020;28:2358-2366.

[53]

DaiE, ChenX, LinkermannA, et al. A guideline on the molecular ecosystem regulating ferroptosis. Nat Cell Biol. 2024.doi:10.1038/s41556-024-01360-8

[54]

KennedyL, SandhuJK, HarperME, Cuperlovic-Culf M. Role of glutathione in cancer: from mechanisms to therapies. Biomolecules. 2020;10:1429.

[55]

StockwellBR, JiangX, GuW. Emerging mechanisms and disease relevance of ferroptosis. Trends Cell Biol. 2020;30:478-490.

[56]

KirtoniaA, SethiG, GargM. The multifaceted role of reactive oxygen species in tumorigenesis. Cell Mol Life Sci. 2020;77:4459-4483.

[57]

LiuJ, HongM, LiY, ChenD, WuY, HuY. Programmed cell death tunes tumor immunity. Front Immunol. 2022;13:847345.

[58]

ZhouX, ZouL, LiaoH, et al. Abrogation of HnRNP L enhances anti-PD-1 therapy efficacy via diminishing PD-L1 and promoting CD8(+) T cell-mediated ferroptosis in castration-resistant prostate cancer. Acta Pharm Sin B. 2022;12:692-707.

[59]

RothlinCV, HilleTD, GhoshS. Determining the effector response to cell death. Nat Rev Immunol. 2021;21:292-304.

[60]

XuYJ, ZhengL, HuYW, WangQ. Pyroptosis and its relationship to atherosclerosis. Clin Chim Acta. 2018;476:28-37.

[61]

KomadaT, MuruveDA. The role of inflammasomes in kidney disease. Nat Rev Nephrol. 2019;15:501-520.

[62]

FangY, TianS, PanY, et al. Pyroptosis: a new frontier in cancer. Biomed Pharmacother. 2020;121:109595.

[63]

SilkeJ, VinceJ. IAPs and cell death. Curr Top Microbiol Immunol. 2017;403:95-117.

[64]

Fernández-DuranI, QuintanillaA, Tarrats N, et al. Cytoplasmic innate immune sensing by the caspase-4 non-canonical inflammasome promotes cellular senescence. Cell Death Differ. 2022;29:1267-1282.

[65]

ShiJ, ZhaoY, WangK, et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature. 2015;526:660-665.

[66]

WangY, GaoW, ShiX, et al. Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature. 2017;547:99-103.

[67]

ChenKW, Demarco B, HeiligR, et al. Extrinsic and intrinsic apoptosis activate pannexin-1 to drive NLRP3 inflammasome assembly. EMBO J. 2019;38:e101638.

[68]

KayagakiN, StoweIB, LeeBL, et al. Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature. 2015;526:666-671.

[69]

SuL, ChenY, HuangC, et al. Targeting Src reactivates pyroptosis to reverse chemoresistance in lung and pancreatic cancer models. Sci Transl Med. 2023;15:eabl7895.

[70]

WangW, ZhangL, SunZ. Eliciting pyroptosis to fuel cancer immunotherapy: mechanisms and strategies. Cancer Biol Med. 2022;19:948-964.

[71]

WangQ, WangY, DingJ, et al. A bioorthogonal system reveals antitumour immune function of pyroptosis. Nature. 2020;579:421-426.

[72]

LuY, HeW, HuangX, et al. Strategies to package recombinant adeno-associated virus expressing the N-terminal gasdermin domain for tumor treatment. Nat Commun. 2021;12:7155.

[73]

PasparakisM, Vandenabeele P. Necroptosis and its role in inflammation. Nature. 2015;517:311-320.

[74]

ConradM, AngeliJP, VandenabeeleP, StockwellBR. Regulated necrosis: disease relevance and therapeutic opportunities. Nat Rev Drug Discov. 2016;15:348-366.

[75]

DegterevA, HitomiJ, GermscheidM, et al. Identification of RIP1 kinase as a specific cellular target of necrostatins. Nat Chem Biol. 2008;4:313-321.

[76]

YanJ, WanP, ChoksiS, Liu ZG. Necroptosis and tumor progression. Trends Cancer. 2022;8:21-27.

[77]

CaiZ, Jitkaew S, ZhaoJ, et al. Plasma membrane translocation of trimerized MLKL protein is required for TNF-induced necroptosis. Nat Cell Biol. 2014;16:55-65.

[78]

KaczmarekA, Vandenabeele P, KryskoDV. Necroptosis: the release of damage-associated molecular patterns and its physiological relevance. Immunity. 2013;38:209-223.

[79]

LiuZG, JiaoD. Necroptosis, tumor necrosis and tumorigenesis. Cell Stress. 2019;4:1-8.

[80]

HöckendorfU, Yabal M, HeroldT, et al. RIPK3 restricts myeloid leukemogenesis by promoting cell death and differentiation of leukemia initiating cells. Cancer Cell. 2016;30:75-91.

[81]

KooGB, MorganMJ, LeeDG, et al. Methylation-dependent loss of RIP3 expression in cancer represses programmed necrosis in response to chemotherapeutics. Cell Res. 2015;25:707-725.

[82]

SchumacherTN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348:69-74.

[83]

SnyderAG, Hubbard NW, MessmerMN, et al. Intratumoral activation of the necroptotic pathway components RIPK1 and RIPK3 potentiates antitumor immunity. Sci Immunol. 2019;4:eaaw2004.

[84]

ZengQ, MaX, SongY, Chen Q, JiaoQ, ZhouL. Targeting regulated cell death in tumor nanomedicines. Theranostics. 2022;12:817-841.

[85]

LongGV, Swetter SM, MenziesAM, GershenwaldJE, Scolyer RA. Cutaneous melanoma. Lancet. 2023;402:485-502.

[86]

SunY, LyuB, YangC, et al. An enzyme-responsive and transformable PD-L1 blocking peptide-photosensitizer conjugate enables efficient photothermal immunotherapy for breast cancer. Bioact Mater. 2023;22:47-59.

[87]

XieL, LiJ, WangG, et al. Phototheranostic metal-phenolic networks with Antiexosomal PD-L1 enhanced ferroptosis for synergistic immunotherapy. J Am Chem Soc. 2022;144:787-797.

[88]

WangG, XieL, LiB, et al. A nanounit strategy reverses immune suppression of exosomal PD-L1 and is associated with enhanced ferroptosis. Nat Commun. 2021;12:5733.

[89]

XuY, GuoY, ZhangC, et al. Fibronectin-coated metal-phenolic networks for cooperative tumor chemo-/Chemodynamic/immune therapy via enhanced ferroptosis-mediated immunogenic cell death. ACS Nano. 2022;16:984-996.

[90]

LiuP, ShiX, PengY, Hu J, DingJ, ZhouW. Anti-PD-L1 DNAzyme loaded photothermal Mn(2+)/Fe(3+) hybrid metal-phenolic networks for cyclically amplified tumor ferroptosis-immunotherapy. Adv Healthc Mater. 2022;11:e2102315.

[91]

ZhangF, LiF, LuGH, et al. Engineering magnetosomes for ferroptosis/immunomodulation synergism in cancer. ACS Nano. 2019;13:5662-5673.

[92]

ZhangM, QinX, ZhaoZ, et al. A self-amplifying nanodrug to manipulate the Janus-faced nature of ferroptosis for tumor therapy. Nanoscale Horiz. 2022;7:198-210.

[93]

MaX, XiaoL, LiuL, et al. CD36-mediated ferroptosis dampens intratumoral CD8(+) T cell effector function and impairs their antitumor ability. Cell Metab. 2021;33:1001-1012.e5.

[94]

WangS, YiX, WuZ, et al. CAMKK2 defines ferroptosis sensitivity of melanoma cells by regulating AMPK–NRF2 pathway. J Invest Dermatol. 2022;142:189-200.e8.

[95]

XuJ, SongF, LyuH, et al. Subtype-specific 3D genome alteration in acute myeloid leukaemia. Nature. 2022;611:387-398.

[96]

NewellLF, CookRJ. Advances in acute myeloid leukemia. BMJ. 2021;375:n2026.

[97]

CaoK, DuY, BaoX, et al. Glutathione-bioimprinted nanoparticles targeting of N6-methyladenosine FTO demethylase as a strategy against leukemic stem cells. Small. 2022;18:e2106558.

[98]

LiQ, SuR, BaoX, et al. Glycyrrhetinic acid nanoparticles combined with ferrotherapy for improved cancer immunotherapy. Acta Biomater. 2022;144:109-120.

[99]

YangF, XiaoY, DingJH, et al. Ferroptosis heterogeneity in triple-negative breast cancer reveals an innovative immunotherapy combination strategy. Cell Metab. 2023;35:84-100.e8.

[100]

JiangQ, WangK, ZhangX, et al. Platelet membrane-camouflaged magnetic nanoparticles for ferroptosis-enhanced cancer immunotherapy. Small. 2020;16:e2001704.

[101]

JiangZ, LimSO, YanM, et al. TYRO3 induces anti-PD-1/PD-L1 therapy resistance by limiting innate immunity and tumoral ferroptosis. J Clin Invest. 2021;131:e139434.

[102]

ConcheC, Finkelmeier F, PešićM, et al. Combining ferroptosis induction with MDSC blockade renders primary tumours and metastases in liver sensitive to immune checkpoint blockade. Gut. 2023;72:1774-1782.

[103]

LiangH, WuX, ZhaoG, Feng K, NiK, SunX. Renal clearable ultrasmall single-crystal Fe nanoparticles for highly selective and effective ferroptosis therapy and immunotherapy. J Am Chem Soc. 2021;143:15812-15823.

[104]

HanW, DuanX, NiK, LiY, ChanC, Lin W. Co-delivery of dihydroartemisinin and pyropheophorbide-iron elicits ferroptosis to potentiate cancer immunotherapy. Biomaterials. 2022;280:121315.

[105]

FanF, LiuP, BaoR, et al. A dual PI3K/HDAC inhibitor induces immunogenic ferroptosis to potentiate cancer immune checkpoint therapy. Cancer Res. 2021;81:6233-6245.

[106]

DengJ, ZhouM, LiaoT, et al. Targeting cancer cell ferroptosis to reverse immune checkpoint inhibitor therapy resistance. Front Cell Dev Biol. 2022;10:818453.

[107]

HsiehCH, HsiehHC, ShihFS, et al. An innovative NRF2 nano-modulator induces lung cancer ferroptosis and elicits an immunostimulatory tumor microenvironment. Theranostics. 2021;11:7072-7091.

[108]

TangB, ZhuJ, WangY, et al. Targeted xCT-mediated ferroptosis and Protumoral polarization of macrophages is effective against HCC and enhances the efficacy of the anti-PD-1/L1 response. Adv Sci. 2023;10:e2203973.

[109]

WanC, SunY, TianY, et al. Irradiated tumor cell-derived microparticles mediate tumor eradication via cell killing and immune reprogramming. Sci Adv. 2020;6:eaay9789.

[110]

ZhangZ, ZhangY, XiaS, et al. Gasdermin E suppresses tumour growth by activating anti-tumour immunity. Nature. 2020;579:415-420.

[111]

ZhouW, LiuH, YuanZ, et al. Targeting the mevalonate pathway suppresses ARID1A-inactivated cancers by promoting pyroptosis. Cancer Cell. 2023;41:740-756.e10.

[112]

WangH, HeZ, GaoY, et al. Dual-pronged attack: pH-driven membrane-anchored NIR dual-type Nano-photosensitizer excites immunogenic pyroptosis and sequester immune checkpoint for enhanced prostate cancer photo-immunotherapy. Adv Sci. 2023;10:e2302422.

[113]

WeiX, XieF, ZhouX, et al. Role of pyroptosis in inflammation and cancer. Cell Mol Immunol. 2022;19:971-992.

[114]

ZhangS, ZhangY, FengY, et al. Biomineralized two-enzyme nanoparticles regulate tumor Glycometabolism inducing tumor cell pyroptosis and robust antitumor immunotherapy. Adv Mater. 2022;34:e2206851.

[115]

RenY, FengM, HaoX, et al. USP48 stabilizes Gasdermin E to promote pyroptosis in cancer. Cancer Res. 2023;83:1074-1093.

[116]

ZhivakiD, Borriello F, ChowOA, et al. Inflammasomes within hyperactive murine dendritic cells stimulate Long-lived T cell-mediated anti-tumor immunity. Cell Rep. 2020;33:108381.

[117]

BlascoMT, GomisRR. PD-L1 controls cancer pyroptosis. Nat Cell Biol. 2020;22:1157-1159.

[118]

HouJ, ZhaoR, XiaW, et al. PD-L1-mediated gasdermin C expression switches apoptosis to pyroptosis in cancer cells and facilitates tumour necrosis. Nat Cell Biol. 2020;22:1264-1275.

[119]

GongY, FanZ, LuoG, et al. The role of necroptosis in cancer biology and therapy. Mol Cancer. 2019;18:100.

[120]

DongD, WangW, WangH, Chen L, LiuT. Expression patterns of necroptosis-related genes: predicting prognosis and immunotherapeutic effects in cutaneous melanoma. J Oncol. 2022;2022:5722599.

[121]

DeAntoneoC, DanthiP, BalachandranS. Reovirus activated cell death pathways. Cells. 2022;11:1757.

[122]

KangT, HuangY, ZhuQ, et al. Necroptotic cancer cells-mimicry nanovaccine boosts anti-tumor immunity with tailored immune-stimulatory modality. Biomaterials. 2018;164:80-97.

[123]

ZhangT, YinC, FedorovA, et al. ADAR1 masks the cancer immunotherapeutic promise of ZBP1-driven necroptosis. Nature. 2022;606:594-602.

[124]

ZhaoZ, ZhangG, SunY, WinotoA. Necroptotic-susceptible dendritic cells exhibit enhanced antitumor activities in mice. Immun Inflamm Dis. 2020;8:468-479.

[125]

XiaGQ, LeiTR, YuTB, ZhouPH. Nanocarrier-based activation of necroptotic cell death potentiates cancer immunotherapy. Nanoscale. 2021;13:1220-1230.

[126]

GaihaGD, McKimKJ, WoodsM, et al. Dysfunctional HIV-specific CD8+ T cell proliferation is associated with increased caspase-8 activity and mediated by necroptosis. Immunity. 2014;41:1001-1012.

[127]

LomphithakT, Akara-Amornthum P, MurakamiK, et al. Tumor necroptosis is correlated with a favorable immune cell signature and programmed death-ligand 1 expression in cholangiocarcinoma. Sci Rep. 2021;11:11743.

[128]

SchoenfeldAJ, Hellmann MD. Acquired resistance to immune checkpoint inhibitors. Cancer Cell. 2020;37:443-455.

[129]

HsuJ, Hodgins JJ, MaratheM, et al. Contribution of NK cells to immunotherapy mediated by PD-1/PD-L1 blockade. J Clin Invest. 2018;128:4654-4668.

[130]

YokoiT, ObaT, KajiharaR, Abrams SI, ItoF. Local, multimodal intralesional therapy renders distant brain metastases susceptible to PD-L1 blockade in a preclinical model of triple-negative breast cancer. Sci Rep. 2021;11:21992.

[131]

BaiR, CuiJ. Burgeoning exploration of the role of natural killer cells in anti-PD-1/PD-L1 therapy. Front Immunol. 2022;13:886931.

[132]

WangY, ZhangH, LiuC, et al. Immune checkpoint modulators in cancer immunotherapy: recent advances and emerging concepts. J Hematol Oncol. 2022;15:111.

[133]

HackSP, ZhuAX, WangY. Augmenting anticancer immunity through combined targeting of angiogenic and PD-1/PD-L1 pathways: challenges and opportunities. Front Immunol. 2020;11:598877.

[134]

KikuchiH, MatsuiA, MoritaS, et al. Increased CD8+ T-cell infiltration and efficacy for multikinase inhibitors after PD-1 blockade in hepatocellular carcinoma. J Natl Cancer Inst. 2022;114:1301-1305.

[135]

TisonA, GaraudS, ChicheL, Cornec D, KostineM. Immune-checkpoint inhibitor use in patients with cancer and pre-existing autoimmune diseases. Nat Rev Rheumatol. 2022;18:641-656.

RIGHTS & PERMISSIONS

2024 The Authors. Cell Proliferation published by Beijing Institute for Stem Cell and Regenerative Medicine and John Wiley & Sons Ltd.

AI Summary AI Mindmap
PDF

173

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/