B-cell performance in chemotherapy: Unravelling the mystery of B-cell therapeutic potential

Zizhuo Li , Anqi Lin , Zhifei Gao , Aimin Jiang , Minying Xiong , Jiapeng Song , Zaoqu Liu , Quan Cheng , Jian Zhang , Peng Luo

Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (7) : e1761

PDF
Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (7) : e1761 DOI: 10.1002/ctm2.1761
REVIEW

B-cell performance in chemotherapy: Unravelling the mystery of B-cell therapeutic potential

Author information +
History +
PDF

Abstract

• Chemotherapy can inhibit B-cell proliferation and alter subset distributions and functions, including factor secretion, receptor signalling, and costimulation.

• Chemotherapy can modulate complex B-cell–T-cell interactions with variable effects on anti-tumour immunity

• Targeting B-cell surface markers or signalling improves chemotherapy responses, blocks immune evasion and inhibits tumour growth.

• Critical knowledge gaps remain regarding B-cell interactions in TME, B-cell chemoresistance mechanisms, TLS biology, heterogeneity, spatial distributions, chemotherapy drug selection and B-cell targets that future studies should address.

Keywords

anti-tumour therapy / B cells / chemotherapy / neoadjuvant chemotherapy (NACT) / targeting B cells / tumour microenvironment (TME)

Cite this article

Download citation ▾
Zizhuo Li, Anqi Lin, Zhifei Gao, Aimin Jiang, Minying Xiong, Jiapeng Song, Zaoqu Liu, Quan Cheng, Jian Zhang, Peng Luo. B-cell performance in chemotherapy: Unravelling the mystery of B-cell therapeutic potential. Clinical and Translational Medicine, 2024, 14(7): e1761 DOI:10.1002/ctm2.1761

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

TruffiM, Sorrentino L, CorsiF. Fibroblasts in the tumor microenvironment. Adv Exp Med Biol. 2020;1234:15-29.

[2]

Downs-CannerSM, MeierJ, VincentBG, Serody JS. B cell function in the tumor microenvironment. Annu Rev Immunol. 2022;40:169-193.

[3]

WangS-S, LiuW, LyD, et al. Tumor-infiltrating B cells: their role and application in anti-tumor immunity in lung cancer. Cell Mol Immunol. 2019;16:6-18.

[4]

RastogiI, JeonD, MosemanJE, et al. Role of B cells as antigen presenting cells. Front Immunol. 2022;13:954936.

[5]

JacquelotN, Tellier J, NuttSl, BelzGt. Tertiary lymphoid structures and B lymphocytes in cancer prognosis and response to immunotherapies. Oncoimmunology. 2021;10:1900508.

[6]

Conejo-GarciaJR, BiswasS, ChaurioR, Rodriguez PC. Neglected no more: B cell-mediated anti-tumor immunity. Semin Immunol. 2023;65:101707.

[7]

TokunagaR, NaseemM, LoJH, et al. B cell and B cell-related pathways for novel cancer treatments. Cancer Treat Rev. 2019;73:10-19.

[8]

LohrM, EdlundK, BotlingJ, et al. The prognostic relevance of tumour-infiltrating plasma cells and immunoglobulin kappa C indicates an important role of the humoral immune response in non-small cell lung cancer. Cancer Lett. 2013;333:222-228.

[9]

MichaudD, Steward CR, MirlekarB, Pylayeva-GuptaY. Regulatory B cells in cancer. Immunol Rev. 2021;299:74-92.

[10]

SarvariaA, Madrigal JA, SaudemontA. B cell regulation in cancer and anti-tumor immunity. Cell Mol Immunol. 2017;14:662-674.

[11]

AndersonNM, SimonMC. The tumor microenvironment. Curr Biol. 2020;30:R921-R925.

[12]

HouZ, LiuJ, JinZ, et al. Use of chemotherapy to treat hepatocellular carcinoma. Biosci Trends. 2022;16:31-45.

[13]

PassaroA, AttiliI, De MarinisF. Neoadjuvant chemotherapy plus immunotherapy in early-stage resectable non-small-cell lung cancer. J Clin Oncol. 2022;40:2871-2877.

[14]

JPM | Free. Full-Text | Breast Cancer Treatments: Updates and New Challenges. . Accessed 17 Nov 2023.

[15]

McQuadeRM, Stojanovska V, BornsteinJC, NurgaliK. Colorectal cancer chemotherapy: the evolution of treatment and new approaches. Curr Med Chem. 2017;24:1537-1557.

[16]

SlomkowskiM, Kopec-Szlezak J, Fabijanska-MitekJ, et al. B cells CD19+ in patients with B-cell chronic lymphocytic leukaemia and autoimmune haemolytic anaemia. Clin Lab Haematol. 2004;26:385-389.

[17]

MackayIR, Goodyear MD, RiglarC, et al. Effect on immunologic and other indices of adjuvant cytotoxic chemotherapy including melphalan in breast cancer. Cancer. 1984;53:2619-2627.

[18]

FroidevauxS, LoorF. Myeloid and lymphoid cell alterations in normal mice exposed to chemotherapy with doxorubicin and/or the multidrug-resistance reversing agent SDZ PSC 833. Int J Cancer. 1994;59:133-140.

[19]

ShangX, ZhangC, WangK, Wang H. Neoadjuvant chemotherapy remodels the tumor immune microenvironment by increasing activated and cytotoxic T cell, decreasing B cells and macrophages in small cell lung cancer. J Transl Med. 2023;21:645.

[20]

BerdD, Maguire HC, MastrangeloMJ. Potentiation of human cell-mediated and humoral immunity by low-dose cyclophosphamide. Cancer Res. 1984;44:5439-5443.

[21]

GaudreauP-O, NegraoMV, MitchellKG, et al. Neoadjuvant chemotherapy increases cytotoxic T cell, tissue resident memory T cell, and B cell infiltration in resectable NSCLC. J Thorac Oncol. 2021;16:127-139.

[22]

SewellHF, Halbert CF, RobinsRA, et al. Chemotherapy-induced differential changes in lymphocyte subsets and natural-killer-cell function in patients with advanced breast cancer. Int J Cancer. 1993;55:735-738.

[23]

MontfortA, PearceO, ManiatiE, et al. A strong B-cell response is part of the immune landscape in human high-grade serous ovarian metastases. Clin Cancer Res. 2017;;23:250-262.

[24]

HuiZ, RenY, ZhangD, et al. PD-1 blockade potentiates neoadjuvant chemotherapy in NSCLC via increasing CD127+ and KLRG1+ CD8 T cells. NPJ Precis Oncol. 2023;7:48.

[25]

ZandvoortA, Lodewijk ME, KlokPA, et al. Slow recovery of follicular B cells and marginal zone B cells after chemotherapy: implications for humoral immunity. Clin Exp Immunol. 2002;124:172-179.

[26]

VermaR, FosterRE, HorganK, et al. Lymphocyte depletion and repopulation after chemotherapy for primary breast cancer. Breast Cancer Res. 2016;18:10.

[27]

LuY, ZhaoQ, LiaoJ-Y, et al. Complement signals determine opposite effects of B cells in chemotherapy-induced immunity. Cell. 2020;180:1081-1097 e24.

[28]

SakaguchiA, Horimoto Y, OnagiH, et al. Plasma cell infiltration and treatment effect in breast cancer patients treated with neoadjuvant chemotherapy. Breast Cancer Res. 2021;23:99.

[29]

HenriksenJR, Nederby L, DonskovF, et al. Prognostic significance of baseline T cells, B cells and neutrophil-lymphocyte ratio (NLR) in recurrent ovarian cancer treated with chemotherapy. J Ovarian Res. 2020;13:59.

[30]

GermainC, Gnjatic S, TamzalitF, et al. Presence of B cells in tertiary lymphoid structures is associated with a protective immunity in patients with lung cancer. Am J Respir Crit Care Med. 2014;189:832-844.

[31]

NakamuraS, Ohuchida K, OhtsuboY, et al. Single-cell transcriptome analysis reveals functional changes in tumour-infiltrating B lymphocytes after chemotherapy in oesophageal squamous cell carcinoma. Clin Transl Med. 2023;13:e1181.

[32]

EibelH, KrausH, SicH, et al. B cell biology: an overview. Curr Allergy Asthma Rep. 2014;14:434.

[33]

PieperK, Grimbacher B, EibelH. B-cell biology and development. J Allergy Clin Immunol. 2013;131:959-971.

[34]

OhashiPS, DeFranco AL. Making and breaking tolerance. Curr Opin Immunol. 2002;14:744-759.

[35]

NemazeeD, Weigert M. Revising B cell receptors. J Exp Med. 2000;191:1813-1818.

[36]

GayD, Saunders T, CamperS, WeigertM. Receptor editing: an approach by autoreactive B cells to escape tolerance. J Exp Med. 1993;177:999-1008.

[37]

HartleySB, Crosbie J, BrinkR, et al. Elimination from peripheral lymphoid tissues of self-reactive B lymphocytes recognizing membrane-bound antigens. Nature. 1991;353:765-769.

[38]

TiegsSL, Russell DM, NemazeeD. Receptor editing in self-reactive bone marrow B cells. J Exp Med. 1993;177:1009-1020.

[39]

Shapiro-ShelefM, CalameK. Regulation of plasma-cell development. Nat Rev Immunol. 2005;5:230-242.

[40]

EklandEH, Forster R, LippM, CysterJG. Requirements for follicular exclusion and competitive elimination of autoantigen-binding B cells. J Immunol. 2004;172:4700-4708.

[41]

NuttSL, Hodgkin PD, TarlintonDM, CorcoranLM. The generation of antibody-secreting plasma cells. Nat Rev Immunol. 2015;15:160-171.

[42]

LeBienTW, TedderTF. B lymphocytes: how they develop and function. Blood. 2008;112:1570-1580.

[43]

CalameKL, LinK-I, TunyaplinC. Regulatory mechanisms that determine the development and function of plasma cells. Annu Rev Immunol. 2003;21:205-230.

[44]

InoueT, Kurosaki T. Memory B cells. Nat Rev Immunol. 2024;24:5-17.

[45]

SeifertM, Küppers R. Human memory B cells. Leukemia. 2016;30:2283-2292.

[46]

RosserEC, MauriC. Regulatory B cells: origin, phenotype, and function. Immunity. 2015;42:607-612.

[47]

MantovaniA, Marchesi F, JaillonS, et al. Tumor-associated myeloid cells: diversity and therapeutic targeting. Cell Mol Immunol. 2021;18:566-578.

[48]

DangajD, Lanitis E, ZhaoA, et al. Novel recombinant human b7-h4 antibodies overcome tumoral immune escape to potentiate T-cell antitumor responses. Cancer Res. 2013;73:4820-4829.

[49]

DixonKO, TabakaM, SchrammMA, et al. TIM-3 restrains anti-tumour immunity by regulating inflammasome activation. Nature. 2021;595:101-106.

[50]

CasazzaA, LaouiD, WenesM, et al. Impeding macrophage entry into hypoxic tumor areas by Sema3A/Nrp1 signaling blockade inhibits angiogenesis and restores antitumor immunity. Cancer Cell. 2013;24:695-709.

[51]

WenesM, ShangM, Di MatteoM, et al. Macrophage metabolism controls tumor blood vessel morphogenesis and metastasis. Cell Metab. 2016;24:701-715.

[52]

GordonSR, MauteRL, DulkenBW, et al. PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature. 2017;545:495-499.

[53]

QorrajM, BrunsH, BöttcherM, et al. The PD-1/PD-L1 axis contributes to immune metabolic dysfunctions of monocytes in chronic lymphocytic leukemia. Leukemia. 2017;31:470-478.

[54]

SeoWI, LeeCH, JungSJ, et al. Expression of VISTA on tumor-infiltrating immune cells correlated with short intravesical recurrence in non-muscle-invasive bladder cancer. Cancer Immunol Immunother. 2021;70:3113-3122.

[55]

LaskowskiTJ, Biederstädt A, RezvaniK. Natural killer cells in antitumour adoptive cell immunotherapy. Nat Rev Cancer. 2022;22:557-575.

[56]

BluntMD, KhakooSI. Harnessing natural killer cell effector function against cancer. Immunother Adv. 2024;4:ltad031.

[57]

DjaoudZ, ParhamP. HLAs, TCRs, and KIRs, a triumvirate of human cell-mediated immunity. Annu Rev Biochem. 2020;89:717-739.

[58]

PhilipponC, TaoS, ClementD, et al. Allelic variation of KIR and HLA tunes the cytolytic payload and determines functional hierarchy of NK cell repertoires. Blood Adv. 2023;7:4492-4504.

[59]

SoppJ, CraggMS. Deleting malignant B cells with second-generation anti-CD20 antibodies. J Clin Oncol. 2018;36:2323-2325.

[60]

WangW. NK Cell-mediated antibody-dependent cellular cytotoxicity in cancer immunotherapy. Front Immunol. 2015;6:368.

[61]

BöttcherJP, Bonavita E, ChakravartyP, et al. NK cells stimulate recruitment of cDC1 into the tumor microenvironment promoting cancer immune control. Cell. 2018;172:1022-1037 e14.

[62]

KyrysyukO, Wucherpfennig KW. Designing cancer immunotherapies that engage T cells and NK cells. Annu Rev Immunol. 2023;41:17-38.

[63]

TurajAH, Hussain K, CoxKL, et al. Antibody tumor targeting is enhanced by CD27 agonists through myeloid recruitment. Cancer Cell. 2017;32:777-791.

[64]

CichockiF, Bjordahl R, GaidarovaS, et al. iPSC-derived NK cells maintain high cytotoxicity and enhance in vivo tumor control in concert with T cells and anti–PD-1 therapy. Sci Transl Med. 2020;12:eaaz5618.

[65]

LanierLL. Up on the tightrope: natural killer cell activation and inhibition. Nat Immunol. 2008;9:495-502.

[66]

St PaulM, OhashiPS. The roles of CD8+ T cell subsets in antitumor immunity. Trends Cell Biol. 2020;30:695-704.

[67]

JhunjhunwalaS, HammerC, DelamarreL. Antigen presentation in cancer: insights into tumour immunogenicity and immune evasion. Nat Rev Cancer. 2021;21:298-312.

[68]

AhrendsT, BorstJ. The opposing roles of CD4+ T cells in anti-tumour immunity. Immunology. 2018;154:582-592.

[69]

QinL, WaseemTC, SahooA, et al. insights into the molecular mechanisms of T follicular helper-mediated immunity and pathology. Front Immunol. 2018;9:1884.

[70]

CrottyS. T Follicular helper cell biology: a decade of discovery and diseases. Immunity. 2019;50:1132-1148.

[71]

OhDY, FongL. Cytotoxic CD4+ T cells in cancer: expanding the immune effector toolbox. Immunity. 2021;54:2701-2711.

[72]

BraumüllerH, Wieder T, BrennerE, et al. T-helper-1-cell cytokines drive cancer into senescence. Nature. 2013;494:361-365.

[73]

De VisserKE, JoyceJA. The evolving tumor microenvironment: from cancer initiation to metastatic outgrowth. Cancer Cell. 2023;41:374-403.

[74]

SahaiE, Astsaturov I, CukiermanE, et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat Rev Cancer. 2020;20:174-186.

[75]

FeigC, JonesJO, KramanM, et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc Natl Acad Sci USA. 2013;110:20212-20217.

[76]

MariathasanS, TurleySJ, NicklesD, et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature. 2018;554:544-548.

[77]

WuSZ, RodenDL, WangC, et al. Stromal cell diversity associated with immune evasion in human triple-negative breast cancer. EMBO J. 2020;39:e104063.

[78]

SmitB, Stjernswärd J, DowdleE, et al. The lymphocyte: monocyte ratio: B- and T-cell ratio after radiotherapy, chemotherapy and surgery. Int J Rad Oncol*Biol*Phys. 1979;5:1841-1847.

[79]

MaĭborodinIV, Strunkin DN, MaĭborodinaVI, et al. Changes in the aggregated lymphoid nodules and in the mesenterial lymph nodes of rats after the administration of a chemotherapeutic drug complex: response similarities and differences. Morfologiia. 2007;132:68-73.

[80]

KohnkePL, Mactier S, AlmaziJG, et al. Fludarabine and cladribine induce changes in surface proteins on human B-lymphoid cell lines involved with apoptosis, cell survival, and antitumor immunity. J Proteome Res. 2012;11:4436-4448.

[81]

ReillyA, KersunLS, Luning PrakE, et al. Immunologic consequences of chemotherapy for acute myeloid leukemia. J Pediatr Hematol Oncol. 2013;35:46-53.

[82]

EyrichM, Wiegering V, LimA, et al. Immune function in children under chemotherapy for standard risk acute lymphoblastic leukaemia – a prospective study of 20 paediatric patients. Br J Haematol. 2009;147:360-370.

[83]

IrieE, Shirota Y, SuzukiC, et al. Severe hypogammaglobulinemia persisting for 6 years after treatment with rituximab combined chemotherapy due to arrest of B lymphocyte differentiation together with alteration of T lymphocyte homeostasis. Int J Hematol. 2010;91:501-508.

[84]

Cuenca-MicóO, Delgado-González E, AnguianoB, et al. Effects of molecular iodine/chemotherapy in the immune component of breast cancer tumoral microenvironment. biomolecules. 2021;11:1501.

[85]

GoswamiM, PrinceG, BiancottoA, et al. Impaired B cell immunity in acute myeloid leukemia patients after chemotherapy. J Transl Med. 2017;15:155.

[86]

Van TilburgCM, Van Der Velden VHJ, SandersEAM, et al. Reduced versus intensive chemotherapy for childhood acute lymphoblastic leukemia: impact on lymphocyte compartment composition. Leuk Res. 2011;35:484-491.

[87]

van TilburgCM, van Gent R, BieringsMB, et al. Immune reconstitution in children following chemotherapy for haematological malignancies: a long-term follow-up. Br J Haematol. 2011;152:201-210.

[88]

ZandvoortA, Lodewijk ME, KlokPA, et al. After chemotherapy, functional humoral response capacity is restored before complete restoration of lymphoid compartments. Clin Exp Immunol. 2003;131:8-16.

[89]

WhitesideS, Markova M, ChinA, et al. Influence of chemotherapy on allergen-specific IgE. Int Arch Allergy Immunol. 2018;177:145-152.

[90]

MandiliG, CurcioC, BulfamanteS, et al. In pancreatic cancer, chemotherapy increases antitumor responses to tumor-associated antigens and potentiates DNA vaccination. J Immunother Cancer. 2020;8:e001071.

[91]

CarterNA, Vasconcellos R, RosserEC, et al. Mice lacking endogenous IL-10–producing regulatory B cells develop exacerbated disease and present with an increased frequency of Th1/Th17 but a decrease in regulatory T cells. J Immunol. 2011;186:5569-5579.

[92]

KesselA, HajT, PeriR, et al. Human CD19(+)CD25(high) B regulatory cells suppress proliferation of CD4(+) T cells and enhance Foxp3 and CTLA-4 expression in T-regulatory cells. Autoimmun Rev. 2012;11:670-677.

[93]

ZirakzadehAA, KinnJ, KrantzD, et al. Doxorubicin enhances the capacity of B cells to activate T cells in urothelial urinary bladder cancer. Clin Immunol. 2017;176:63-70.

[94]

ZiebartA, HuberU, JeskeS, et al. The influence of chemotherapy on adenosine-producing B cells in patients with head and neck squamous cell carcinoma. Oncotarget. 2018;9:5834-5847.

[95]

YangJ, LiW, LuoF, et al. Low percentage of CD24hiCD27+CD19+ B cells decelerates gastric cancer progression in XELOX-treated patients. Int Immunopharmacol. 2015;26:322-327.

[96]

LiW, SongD, LiH, et al. Reduction in peripheral CD19+CD24hCD27+ B cell frequency predicts favourable clinical course in XELOX-treated patients with advanced gastric cancer. Cell Physiol Biochem. 2017;41:2045-2052.

[97]

LeiC-J, LiuJ-N, WuR, et al. Change of the peripheral blood immune pattern and its correlation with prognosis in patients with liver cancer treated by sorafenib. Asian Pac J Trop Med. 2016;9:592-596.

[98]

LvJ, WeiY, YinJ-H, et al. The tumor immune microenvironment of nasopharyngeal carcinoma after gemcitabine plus cisplatin treatment. Nat Med. 2023;29:1424-1436.

[99]

JiangQ, FuQ, ChangY, et al. CD19+ tumor-infiltrating B-cells prime CD4+ T-cell immunity and predict platinum-based chemotherapy efficacy in muscle-invasive bladder cancer. Cancer Immunol Immunother. 2019;68:45-56.

[100]

HuiZ, ZhangJ, RenY, et al. Single-cell profiling of immune cells after neoadjuvant pembrolizumab and chemotherapy in IIIA non-small cell lung cancer (NSCLC). Cell Death Dis. 2022;13:607.

[101]

LinX, YeL, WangX, et al. Follicular helper T cells remodel the immune microenvironment of pancreatic cancer via secreting CXCL13 and IL-21. Cancers (Basel). 2021;13:3678.

[102]

ZhangF, LiR, YangY, et al. Specific decrease in B-cell-derived extracellular vesicles enhances post-chemotherapeutic CD8+ T cell responses. Immunity. 2019;50:738-750 e7.

[103]

RaskovalovaT, Lokshin A, HuangX, et al. Inhibition of cytokine production and cytotoxic activity of human antimelanoma specific CD8+ and CD4+ T lymphocytes by adenosine-protein kinase A type I signaling. Cancer Res. 2007;67:5949-5956.

[104]

GhiringhelliF, Bruchard M, ChalminF, RébéC. Production of adenosine by ectonucleotidases: a key factor in tumor immunoescape. J Biomed Biotechnol. 2012;2012:1-9.

[105]

ShalapourS, Font-Burgada J, Di CaroG, et al. Immunosuppressive plasma cells impede T-cell-dependent immunogenic chemotherapy. Nature. 2015;521:94-98.

[106]

AffaraNI, Ruffell B, MedlerTR, et al. B cells regulate macrophage phenotype and response to chemotherapy in squamous carcinomas. Cancer Cell. 2014;25:809-821.

[107]

ChenY-L, LinH-W, ChienC-L, et al. BTLA blockade enhances cancer therapy by inhibiting IL-6/IL-10-induced CD19(high) B lymphocytes. J Immunother Cancer. 2019;7:313.

[108]

SunY, Campisi J, HiganoC, et al. Treatment-induced damage to the tumor microenvironment promotes prostate cancer therapy resistance through WNT16B. Nat Med. 2012;18:1359-1368.

[109]

ChienY, Scuoppo C, WangX, et al. Control of the senescence-associated secretory phenotype by NF-κB promotes senescence and enhances chemosensitivity. Genes Dev. 2011;25:2125-2136.

[110]

PeiJ-P, WangY, MaL-P, et al. AXL antibody and AXL-ADC mediate antitumor efficacy via targeting AXL in tumor-intrinsic epithelial-mesenchymal transition and tumor-associated M2-like macrophage. Acta Pharmacol Sin. 2023;44:1290-1303.

[111]

SinghS, LeeN, PedrozaDA, et al. Chemotherapy coupled to macrophage inhibition induces T-cell and B-cell infiltration and durable regression in triple-negative breast cancer. Cancer Res. 2022;82:2281-2297.

[112]

GundersonAJ, KanedaMM, TsujikawaT, et al. Bruton tyrosine kinase-dependent immune cell cross-talk drives pancreas cancer. Cancer Discov. 2016;6:270-285.

[113]

GaoN, Schwartzberg P, WilderJA, et al. B cell induction of IL-13 expression in NK cells: role of CD244 and SLAM-associated protein. J Immunol. 2006;176:2758-2764.

[114]

GaoN, DangT, DunnickWA, et al. Receptors and counterreceptors involved in NK-B cell interactions. J Immunol. 2005;174:4113-4119.

[115]

BlancaIR, BereEW, YoungHA, Ortaldo JR. Human B cell activation by autologous NK cells is regulated by CD40-CD40 ligand interaction: role of memory B cells and CD5+ B cells. J Immunol. 2001;167:6132-6139.

[116]

FridmanWH, Petitprez F, MeylanM, et al. B cells and cancer: to B or not to B? J Exp Med. 2021;218:e20200851.

[117]

XiY, ZhangY, ZhengK, et al. A chemotherapy response prediction model derived from tumor-promoting B and Tregs and proinflammatory macrophages in HGSOC. Front Oncol. 2023;13:1171582.

[118]

CeruttiA. The regulation of IgA class switching. Nat Rev Immunol. 2008;8:421-434.

[119]

AmmiranteM, Shalapour S, KangY, et al. Tissue injury and hypoxia promote malignant progression of prostate cancer by inducing CXCL13 expression in tumor myofibroblasts. Proc Natl Acad Sci USA. 2014;111:14776-14781.

[120]

SchietingerA, Greenberg PD. Tolerance and exhaustion: defining mechanisms of T cell dysfunction. Trends Immunol. 2014;35:51-60.

[121]

JungYJ, WooJS, HwangS-H, et al. Effect of IL-10-producing B cells in peripheral blood and tumor tissue on gastric cancer. Cell Commun Signal. 2023;21:320.

[122]

Jabłońska-TrypućA, MatejczykM, Rosochacki S. Matrix metalloproteinases (MMPs), the main extracellular matrix (ECM) enzymes in collagen degradation, as a target for anticancer drugs. J Enzyme Inhib Med Chem. 2016;31:177-183.

[123]

CardosoAP, PintoML, PintoAT, et al. Matrix metalloproteases as maestros for the dual role of LPS- and IL-10-stimulated macrophages in cancer cell behaviour. BMC Cancer. 2015;15:456.

[124]

KongY, JiangJ, HuangY, et al. Endoplasmic reticulum stress in melanoma pathogenesis and resistance. Biomed Pharmacother. 2022;155:113741.

[125]

ChenX, Cubillos-Ruiz JR. Endoplasmic reticulum stress signals in the tumour and its microenvironment. Nat Rev Cancer. 2021;21:71-88.

[126]

LinL, LinG, LinH, et al. Integrated profiling of endoplasmic reticulum stress-related DERL3 in the prognostic and immune features of lung adenocarcinoma. Front Immunol. 2022;13:906420.

[127]

ZhangL, ZhangR, JinD, et al. Synergistic induction of tertiary lymphoid structures by chemoimmunotherapy in bladder cancer. Br J Cancer. 2024;130:1221-1231.

[128]

SunX, LiuW, SunL, et al. Maturation and abundance of tertiary lymphoid structures are associated with the efficacy of neoadjuvant chemoimmunotherapy in resectable non-small cell lung cancer. J ImmunoTher Cancer. 2022;10:e005531.

[129]

AnY, SunJ-X, XuM-Y, et al. Tertiary lymphoid structure patterns aid in identification of tumor microenvironment infiltration and selection of therapeutic agents in bladder cancer. Front Immunol. 2022;13:1049884.

[130]

FranzL, Alessandrini L, OttavianoG, et al. Postoperative radiotherapy for laryngeal cancer. The prognostic role of programmed death-ligand 1: an immune microenvironment-based cluster analysis. Pathol – Res Pract. 2020;216:153120.

[131]

RemarkR, LupoA, AlifanoM, et al. Immune contexture and histological response after neoadjuvant chemotherapy predict clinical outcome of lung cancer patients. OncoImmunology. 2016;5:e1255394.

[132]

SongIH, HeoS-H, BangWS, et al. Predictive value of tertiary lymphoid structures assessed by high endothelial venule counts in the neoadjuvant setting of triple-negative breast cancer. Cancer Res Treat. 2017;49:399-407.

[133]

Gu-TrantienC, Migliori E, BuisseretL, et al. CXCL13-producing TFH cells link immune suppression and adaptive memory in human breast cancer. JCI Insight. 2017;2:e91487.

[134]

IshigamiE, Sakakibara M, SakakibaraJ, et al. Coexistence of regulatory B cells and regulatory T cells in tumor-infiltrating lymphocyte aggregates is a prognostic factor in patients with breast cancer. Breast Cancer-tokyo. 2019;26:180-189.

[135]

PoschF, SilinaK, LeiblS, et al. Maturation of tertiary lymphoid structures and recurrence of stage II and III colorectal cancer. OncoImmunology. 2018;7:e1378844.

[136]

SiliņaK, Soltermann A, AttarFM, et al. Germinal centers determine the prognostic relevance of tertiary lymphoid structures and are impaired by corticosteroids in lung squamous cell carcinoma. Cancer Res. 2018;78:1308-1320.

[137]

FridmanWH, MeylanM, PetitprezF, et al. B cells and tertiary lymphoid structures as determinants of tumour immune contexture and clinical outcome. Nat Rev Clin Oncol. 2022;19:441-457.

[138]

DelvecchioFR, Fincham REA, SpearS, et al. Pancreatic cancer chemotherapy is potentiated by induction of tertiary lymphoid structures in mice. Cell Mol Gastroenterol Hepatol. 2021;12:1543-1565.

[139]

HuJ, ZhangL, XiaH, et al. Tumor microenvironment remodeling after neoadjuvant immunotherapy in non-small cell lung cancer revealed by single-cell RNA sequencing. Genome Med. 2023;15:14.

[140]

JiangH, YuD, YangP, et al. Revealing the transcriptional heterogeneity of organ-specific metastasis in human gastric cancer using single-cell RNA Sequencing. Clin Transl Med. 2022;12:e730.

[141]

ReuschenbachM, Von Knebel Doeberitz M, WentzensenN. A systematic review of humoral immune responses against tumor antigens. Cancer Immunol Immunother. 2009;58:1535-1544.

[142]

HannaniD, LocherC, YamazakiT, et al. Contribution of humoral immune responses to the antitumor effects mediated by anthracyclines. Cell Death Differ. 2014;21:50-58.

[143]

Lopez De RodasM, Nagineni V, RaviA, et al. Role of tumor infiltrating lymphocytes and spatial immune heterogeneity in sensitivity to PD-1 axis blockers in non-small cell lung cancer. J ImmunoTher Cancer. 2022;10:e004440.

[144]

CorredorG, WangX, ZhouY, et al. Spatial architecture and arrangement of tumor-infiltrating lymphocytes for predicting likelihood of recurrence in early-stage non-small cell lung cancer. Clin Cancer Res. 2019;25:1526-1534.

[145]

WalterCU, BillerBJ, LanaSE, et al. Effects of chemotherapy on immune responses in dogs with cancer. J Vet Intern Med. 2006;20:342-347. 342:eocoir]2.0.co;2

[146]

Sordo-BahamondeC, Lorenzo-Herrero S, Granda-DíazR, et al. Beyond the anti-PD-1/PD-L1 era: promising role of the BTLA/HVEM axis as a future target for cancer immunotherapy. Mol Cancer. 2023;22:142.

[147]

SchilderRJ, Powderly JD, ParkH, et al. Phase Ia dose-escalation study of the anti-BTLA antibody icatolimab as a monotherapy in patients with advanced solid tumor. J Clin Oncol;40.

[148]

HenneganT, BurtonEM, SimpsonL, et al. Expanded cohort and extended follow-up of neoadjuvant plus adjuvant (neo + adj) dabrafenib (D) and trametinib (T) in patients (pts) with surgically resectable stage (stg) III/IV melanoma. J Clin Oncol. 2023;41:9583-9583.

[149]

DhoS, ChoE, LeeJ, et al. A novel therapeutic anti‑CD55 monoclonal antibody inhibits the proliferation and metastasis of colorectal cancer cells. Oncol Rep. 2019;42:2686-2693.

[150]

RozkiewiczD, Hermanowicz JM, KwiatkowskaI, et al. Bruton's tyrosine kinase inhibitors (BTKIs): review of preclinical studies and evaluation of clinical trials. Molecules. 2023;28:2400.

RIGHTS & PERMISSIONS

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

AI Summary AI Mindmap
PDF

153

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/