Cancer-Associated B Cells in Sarcoma
Abstract
:Simple Summary
Abstract
1. A Historical Perspective
“It is by no means inconceivable that small accumulations of tumour cells may develop and, because of their possession of new antigenic potentialities, provoke an effective immunological reaction with regression of the tumour and no clinical hint of its existence. It has also been suggested that the result of surgery for cancer may to a large extent be determined by the degree of resistance, presumably immunological in nature, against the tumour cells.”- Sir Macfarlane Burnet, 1957 [1]
2. B Cells in the Sarcoma Microenvironment
2.1. Intra-Tumoral B Cells in Sarcoma
2.2. Tertiary Lymphoid Structures
3. Sarcoma Immunotherapy and B Cells
Tertiary Lymphoid Structures and Immunotherapy
4. Sarcoma Antigens and Anti-Tumor Antibodies
4.1. Cancer-Testis Antigens and Translocation-Positive Sarcoma
4.2. Intratumoral Sarcoma Immunoglobulin
5. Pro-Tumor Relationships of B Cells in Sarcoma
6. Next Steps
7. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Burnet, M. Cancer: A biological approach. III. Viruses associated with neoplastic conditions. IV. Practical applications. Br. Med. J. 1957, 1, 841–847. [Google Scholar] [CrossRef]
- Cooper, M.D. The early history of B cells. Nat. Rev. Immunol. 2015, 15, 191–197. [Google Scholar] [CrossRef] [Green Version]
- Cooper, M.D.; Peterson, R.D.; Good, R.A. Delineation of the thymic and bursal lymphoid systems in the chicken. Nature 1965, 205, 143–146. [Google Scholar] [CrossRef]
- Morton, D.L.; Malmgren, R.A. Human osteosarcomas: Immunologic evidence suggesting an associated infectious agent. Science 1968, 162, 1279–1281. [Google Scholar] [CrossRef]
- Dunn, G.P.; Bruce, A.T.; Ikeda, H.; Old, L.J.; Schreiber, R.D. Cancer immunoediting: From immunosurveillance to tumor escape. Nat. Immunol. 2002, 3, 991–998. [Google Scholar] [CrossRef]
- Eilber, F.R.; Morton, D.L. Demonstration in sarcoma patients of anti-tumor antibodies which fix only human complement. Nature 1970, 225, 1137–1138. [Google Scholar] [CrossRef]
- McBride, R.A.; Watanabe, D.H.; Schierman, L.W. Role of B cells in the expression of genetic resistance to growth of Rous sarcoma in the chicken. Eur. J. Immunol. 1978, 8, 147–149. [Google Scholar] [CrossRef]
- Glass, D.R.; Tsai, A.G.; Oliveria, J.P.; Hartmann, F.J.; Kimmey, S.C.; Calderon, A.A.; Borges, L.; Glass, M.C.; Wagar, L.E.; Davis, M.M.; et al. An Integrated Multi-omic Single-Cell Atlas of Human B Cell Identity. Immunity 2020, 53, 217–232.e5. [Google Scholar] [CrossRef]
- Garaud, S.; Buisseret, L.; Solinas, C.; Gu-Trantien, C.; de Wind, A.; Van den Eynden, G.; Naveaux, C.; Lodewyckx, J.N.; Boisson, A.; Duvillier, H.; et al. Tumor infiltrating B-cells signal functional humoral immune responses in breast cancer. JCI Insight 2019, 5, e129641. [Google Scholar] [CrossRef] [Green Version]
- Wouters, M.C.A.; Nelson, B.H. Prognostic Significance of Tumor-Infiltrating B Cells and Plasma Cells in Human Cancer. Clin. Cancer Res. 2018, 24, 6125–6135. [Google Scholar] [CrossRef]
- Tsagozis, P.; Augsten, M.; Zhang, Y.; Li, T.; Hesla, A.; Bergh, J.; Haglund, F.; Tobin, N.P.; Ehnman, M. An immunosuppressive macrophage profile attenuates the prognostic impact of CD20-positive B cells in human soft tissue sarcoma. Cancer Immunol. Immunother. 2019, 68, 927–936. [Google Scholar] [CrossRef] [Green Version]
- Zou, M.X.; Lv, G.H.; Wang, X.B.; Huang, W.; Li, J.; Jiang, Y.; She, X.L. Clinical Impact of the Immune Microenvironment in Spinal Chordoma: Immunoscore as an Independent Favorable Prognostic Factor. Neurosurgery 2019, 84, E318–E333. [Google Scholar] [CrossRef]
- Duan, W.; Zhang, B.; Li, X.; Chen, W.; Jia, S.; Xin, Z.; Jian, Q.; Jian, F.; Chou, D.; Chen, Z. Single-cell transcriptome profiling reveals intra-tumoral heterogeneity in human chordomas. Cancer Immunol. Immunother. 2022, 71, 2185–2195. [Google Scholar] [CrossRef]
- Sorbye, S.W.; Kilvaer, T.; Valkov, A.; Donnem, T.; Smeland, E.; Al-Shibli, K.; Bremnes, R.M.; Busund, L.T. Prognostic impact of lymphocytes in soft tissue sarcomas. PLoS ONE 2011, 6, e14611. [Google Scholar] [CrossRef] [Green Version]
- Smolle, M.A.; Herbsthofer, L.; Goda, M.; Granegger, B.; Brcic, I.; Bergovec, M.; Scheipl, S.; Prietl, B.; El-Heliebi, A.; Pichler, M.; et al. Influence of tumor-infiltrating immune cells on local control rate, distant metastasis, and survival in patients with soft tissue sarcoma. Oncoimmunology 2021, 10, 1896658. [Google Scholar] [CrossRef]
- Chen, L.; Oke, T.; Siegel, N.; Cojocaru, G.; Tam, A.J.; Blosser, R.L.; Swailes, J.; Ligon, J.A.; Lebid, A.; Morris, C.; et al. The Immunosuppressive Niche of Soft-Tissue Sarcomas is Sustained by Tumor-Associated Macrophages and Characterized by Intratumoral Tertiary Lymphoid Structures. Clin. Cancer Res. 2020, 26, 4018–4030. [Google Scholar] [CrossRef] [Green Version]
- Petitprez, F.; de Reyniès, A.; Keung, E.Z.; Chen, T.W.; Sun, C.M.; Calderaro, J.; Jeng, Y.M.; Hsiao, L.P.; Lacroix, L.; Bougoüin, A.; et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature 2020, 577, 556–560. [Google Scholar] [CrossRef]
- Bruno, T.C. New predictors for immunotherapy responses sharpen our view of the tumour microenvironment. Nature 2020, 577, 474–476. [Google Scholar] [CrossRef] [Green Version]
- Tokunaga, R.; Zhang, W.; Naseem, M.; Puccini, A.; Berger, M.D.; Soni, S.; McSkane, M.; Baba, H.; Lenz, H.J. CXCL9, CXCL10, CXCL11/CXCR3 axis for immune activation—A target for novel cancer therapy. Cancer Treat. Rev. 2018, 63, 40–47. [Google Scholar] [CrossRef]
- Cabrita, R.; Lauss, M.; Sanna, A.; Donia, M.; Skaarup Larsen, M.; Mitra, S.; Johansson, I.; Phung, B.; Harbst, K.; Vallon-Christersson, J.; et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature 2020, 577, 561–565. [Google Scholar] [CrossRef]
- Rubtsov, A.V.; Rubtsova, K.; Kappler, J.W.; Jacobelli, J.; Friedman, R.S.; Marrack, P. CD11c-Expressing B Cells Are Located at the T Cell/B Cell Border in Spleen and Are Potent APCs. J. Immunol. 2015, 195, 71–79. [Google Scholar] [CrossRef] [Green Version]
- Watts, C.; Davidson, H.W. Endocytosis and recycling of specific antigen by human B cell lines. EMBO J. 1988, 7, 1937–1945. [Google Scholar] [CrossRef]
- Bruno, T.C.; Ebner, P.J.; Moore, B.L.; Squalls, O.G.; Waugh, K.A.; Eruslanov, E.B.; Singhal, S.; Mitchell, J.D.; Franklin, W.A.; Merrick, D.T.; et al. Antigen-Presenting Intratumoral B Cells Affect CD4+TIL Phenotypes in Non-Small Cell Lung Cancer Patients. Cancer Immunol. Res. 2017, 5, 898–907. [Google Scholar] [CrossRef] [Green Version]
- Gnjatic, S.; Atanackovic, D.; Matsuo, M.; Jäger, E.; Lee, S.Y.; Valmori, D.; Chen, Y.T.; Ritter, G.; Knuth, A.; Old, L.J. Cross-presentation of HLA class I epitopes from exogenous NY-ESO-1 polypeptides by nonprofessional APCs. J. Immunol. 2003, 170, 1191–1196. [Google Scholar] [CrossRef] [Green Version]
- de Taeye, S.W.; Rispens, T.; Vidarsson, G. The Ligands for Human IgG and Their Effector Functions. Antibodies 2019, 8, 30. [Google Scholar] [CrossRef] [Green Version]
- Davies, A.M.; Sutton, B.J. Human IgG4: A structural perspective. Immunol. Rev. 2015, 268, 139–159. [Google Scholar] [CrossRef] [Green Version]
- Sharonov, G.V.; Serebrovskaya, E.O.; Yuzhakova, D.V.; Britanova, O.V.; Chudakov, D.M. B cells, plasma cells and antibody repertoires in the tumour microenvironment. Nat. Rev. Immunol. 2020, 20, 294–307. [Google Scholar] [CrossRef]
- Michaud, D.; Steward, C.R.; Mirlekar, B.; Pylayeva-Gupta, Y. Regulatory B cells in cancer. Immunol. Rev. 2021, 299, 74–92. [Google Scholar] [CrossRef]
- Maglioco, A.; Machuca, D.G.; Badano, M.N.; Nannini, P.; Camerano, G.V.; Costa, H.; Meiss, R.; Ruggiero, R.A.; Giordano, M.; Dran, G.I. B cells inhibit the antitumor immunity against an established murine fibrosarcoma. Oncol. Lett. 2017, 13, 3225–3232. [Google Scholar] [CrossRef] [Green Version]
- Cyster, J.G.; Allen, C.D.C. B Cell Responses: Cell Interaction Dynamics and Decisions. Cell 2019, 177, 524–540. [Google Scholar] [CrossRef]
- Maglioco, A.; Machuca, D.G.; Camerano, G.; Costa, H.A.; Ruggiero, R.; Dran, G.I. Regulatory B cells present in lymph nodes draining a murine tumor. Medicina 2014, 74, 185–188. [Google Scholar]
- Italiano, A.; Bessede, A.; Pulido, M.; Bompas, E.; Piperno-Neumann, S.; Chevreau, C.; Penel, N.; Bertucci, F.; Toulmonde, M.; Bellera, C.; et al. Pembrolizumab in soft-tissue sarcomas with tertiary lymphoid structures: A phase 2 PEMBROSARC trial cohort. Nat. Med. 2022, 28, 1199–1206. [Google Scholar] [CrossRef]
- Schumacher, T.N.; Thommen, D.S. Tertiary lymphoid structures in cancer. Science 2022, 375, eabf9419. [Google Scholar] [CrossRef]
- Sautès-Fridman, C.; Petitprez, F.; Calderaro, J.; Fridman, W.H. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat. Rev. Cancer 2019, 19, 307–325. [Google Scholar] [CrossRef]
- Laumont, C.M.; Banville, A.C.; Gilardi, M.; Hollern, D.P.; Nelson, B.H. Tumour-infiltrating B cells: Immunological mechanisms, clinical impact and therapeutic opportunities. Nat. Rev. Cancer 2022, 22, 414–430. [Google Scholar] [CrossRef]
- Dieu-Nosjean, M.C.; Goc, J.; Giraldo, N.A.; Sautès-Fridman, C.; Fridman, W.H. Tertiary lymphoid structures in cancer and beyond. Trends Immunol. 2014, 35, 571–580. [Google Scholar] [CrossRef]
- Jenks, S.A.; Cashman, K.S.; Woodruff, M.C.; Lee, F.E.H.; Sanz, I. Extrafollicular responses in humans and SLE. Immunol. Rev. 2019, 288, 136–148. [Google Scholar] [CrossRef]
- Kroeger, D.R.; Milne, K.; Nelson, B.H. Tumor-Infiltrating Plasma Cells Are Associated with Tertiary Lymphoid Structures, Cytolytic T-Cell Responses, and Superior Prognosis in Ovarian Cancer. Clin. Cancer Res. 2016, 22, 3005–3015. [Google Scholar] [CrossRef] [Green Version]
- Sautès-Fridman, C.; Lawand, M.; Giraldo, N.A.; Kaplon, H.; Germain, C.; Fridman, W.H.; Dieu-Nosjean, M.C. Tertiary Lymphoid Structures in Cancers: Prognostic Value, Regulation, and Manipulation for Therapeutic Intervention. Front. Immunol. 2016, 7, 407. [Google Scholar] [CrossRef] [Green Version]
- Meylan, M.; Petitprez, F.; Becht, E.; Bougoüin, A.; Pupier, G.; Calvez, A.; Giglioli, I.; Verkarre, V.; Lacroix, G.; Verneau, J.; et al. Tertiary lymphoid structures generate and propagate anti-tumor antibody-producing plasma cells in renal cell cancer. Immunity 2022, 55, 527–541.e5. [Google Scholar]
- Castino, G.F.; Cortese, N.; Capretti, G.; Serio, S.; Di Caro, G.; Mineri, R.; Magrini, E.; Grizzi, F.; Cappello, P.; Novelli, F.; et al. Spatial distribution of B cells predicts prognosis in human pancreatic adenocarcinoma. Oncoimmunology 2015, 5, e1085147. [Google Scholar] [CrossRef] [Green Version]
- Germain, C.; Gnjatic, S.; Tamzalit, F.; Knockaert, S.; Remark, R.; Goc, J.; Lepelley, A.; Becht, E.; Katsahian, S.; Bizouard, G.; 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. [Google Scholar] [CrossRef]
- Di Caro, G.; Bergomas, F.; Grizzi, F.; Doni, A.; Bianchi, P.; Malesci, A.; Laghi, L.; Allavena, P.; Mantovani, A.; Marchesi, F. Occurrence of tertiary lymphoid tissue is associated with T-cell infiltration and predicts better prognosis in early-stage colorectal cancers. Clin. Cancer Res. 2014, 20, 2147–2158. [Google Scholar] [CrossRef] [Green Version]
- Cipponi, A.; Mercier, M.; Seremet, T.; Baurain, J.F.; Théate, I.; van den Oord, J.; Stas, M.; Boon, T.; Coulie, P.G.; van Baren, N. Neogenesis of lymphoid structures and antibody responses occur in human melanoma metastases. Cancer Res. 2012, 72, 3997–4007. [Google Scholar] [CrossRef] [Green Version]
- Giraldo, N.A.; Becht, E.; Pagès, F.; Skliris, G.; Verkarre, V.; Vano, Y.; Mejean, A.; Saint-Aubert, N.; Lacroix, L.; Natario, I.; et al. Orchestration and Prognostic Significance of Immune Checkpoints in the Microenvironment of Primary and Metastatic Renal Cell Cancer. Clin. Cancer Res. 2015, 21, 3031–3040. [Google Scholar] [CrossRef] [Green Version]
- von Bergwelt-Baildon, M.S.; Vonderheide, R.H.; Maecker, B.; Hirano, N.; Anderson, K.S.; Butler, M.O.; Xia, Z.; Zeng, W.Y.; Wucherpfennig, K.W.; Nadler, L.M.; et al. Human primary and memory cytotoxic T lymphocyte responses are efficiently induced by means of CD40-activated B cells as antigen-presenting cells: Potential for clinical application. Blood 2002, 99, 3319–3325. [Google Scholar] [CrossRef] [Green Version]
- Rossetti, R.A.M.; Lorenzi, N.P.C.; Yokochi, K.; Rosa, M.B.S.F.; Benevides, L.; Margarido, P.F.R.; Baracat, E.C.; Carvalho, J.P.; Villa, L.L.; Lepique, A.P. B lymphocytes can be activated to act as antigen presenting cells to promote anti-tumor responses. PLoS ONE 2018, 13, e0199034. [Google Scholar] [CrossRef] [Green Version]
- Hoption Cann, S.A.; van Netten, J.P.; van Netten, C. Dr William Coley and tumour regression: A place in history or in the future. Postgrad. Med. J. 2003, 79, 672–680. [Google Scholar]
- Coley, W.B., II. Contribution to the Knowledge of Sarcoma. Ann. Surg. 1891, 14, 199–220. [Google Scholar] [CrossRef]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
- D’Angelo, S.P.; Shoushtari, A.N.; Agaram, N.P.; Kuk, D.; Qin, L.X.; Carvajal, R.D.; Dickson, M.A.; Gounder, M.; Keohan, M.L.; Schwartz, G.K.; et al. Prevalence of tumor-infiltrating lymphocytes and PD-L1 expression in the soft tissue sarcoma microenvironment. Hum. Pathol. 2015, 46, 357–365. [Google Scholar] [CrossRef] [Green Version]
- Italiano, A.; Bellera, C.; D’Angelo, S. PD1/PD-L1 targeting in advanced soft-tissue sarcomas: A pooled analysis of phase II trials. J. Hematol. Oncol. 2020, 13, 55. [Google Scholar] [CrossRef]
- Klemen, N.D.; Kelly, C.M.; Bartlett, E.K. The emerging role of immunotherapy for the treatment of sarcoma. J. Surg. Oncol. 2021, 123, 730–738. [Google Scholar] [CrossRef]
- Groisberg, R.; Hong, D.S.; Behrang, A.; Hess, K.; Janku, F.; Piha-Paul, S.; Naing, A.; Fu, S.; Benjamin, R.; Patel, S.; et al. Characteristics and outcomes of patients with advanced sarcoma enrolled in early phase immunotherapy trials. J. Immunother. Cancer 2017, 5, 100. [Google Scholar] [CrossRef] [Green Version]
- Maki, R.G.; Jungbluth, A.A.; Gnjatic, S.; Schwartz, G.K.; D’Adamo, D.R.; Keohan, M.L.; Wagner, M.J.; Scheu, K.; Chiu, R.; Ritter, E.; et al. A Pilot Study of Anti-CTLA4 Antibody Ipilimumab in Patients with Synovial Sarcoma. Sarcoma 2013, 2013, 168145. [Google Scholar] [CrossRef] [Green Version]
- Yarchoan, M.; Hopkins, A.; Jaffee, E.M. Tumor Mutational Burden and Response Rate to PD-1 Inhibition. N. Engl. J. Med. 2017, 377, 2500–2501. [Google Scholar] [CrossRef] [PubMed]
- Chalmers, Z.R.; Connelly, C.F.; Fabrizio, D.; Gay, L.; Ali, S.M.; Ennis, R.; Schrock, A.; Campbell, B.; Shlien, A.; Chmielecki, J.; et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017, 9, 34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tawbi, H.A.; Burgess, M.; Bolejack, V.; Van Tine, B.A.; Schuetze, S.M.; Hu, J.; D’Angelo, S.; Attia, S.; Riedel, R.F.; Priebat, D.A.; et al. Pembrolizumab in advanced soft-tissue sarcoma and bone sarcoma (SARC028): A multicentre, two-cohort, single-arm, open-label, phase 2 trial. Lancet Oncol. 2017, 18, 1493–1501, Erratum in: Lancet Oncol. 2017, 18, e711. [Google Scholar] [CrossRef] [PubMed]
- Burgess, M.A.; Bolejack, V.; Schuetze, S.; Tine, B.A.V.; Attia, S.; Riedel, R.F.; Hu, J.S.; Davis, L.E.; Okuno, S.H.; Priebat, D.A.; et al. Clinical activity of pembrolizumab (P) in undifferentiated pleomorphic sarcoma (UPS) and dedifferentiated/pleomorphic liposarcoma (LPS): Final results of SARC028 expansion cohorts. J. Clin. Oncol. 2019, 37, 11015. [Google Scholar] [CrossRef]
- Paijens, S.T.; Vledder, A.; de Bruyn, M.; Nijman, H.W. Tumor-infiltrating lymphocytes in the immunotherapy era. Cell Mol. Immunol. 2021, 18, 842–859. [Google Scholar] [CrossRef]
- Keung, E.Z.; Burgess, M.; Salazar, R.; Parra, E.R.; Rodrigues-Canales, J.; Bolejack, V.; Van Tine, B.A.; Schuetze, S.M.; Attia, S.; Riedel, R.F.; et al. Correlative Analyses of the SARC028 Trial Reveal an Association Between Sarcoma-Associated Immune Infiltrate and Response to Pembrolizumab. Clin. Cancer Res. 2020, 26, 1258–1266. [Google Scholar] [CrossRef]
- Sharpe, A.H.; Pauken, K.E. The diverse functions of the PD1 inhibitory pathway. Nat. Rev. Immunol. 2018, 18, 153–167. [Google Scholar] [CrossRef]
- Helmink, B.A.; Reddy, S.M.; Gao, J.; Zhang, S.; Basar, R.; Thakur, R.; Yizhak, K.; Sade-Feldman, M.; Blando, J.; Han, G.; et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature 2020, 577, 549–555. [Google Scholar] [CrossRef]
- Kinker, G.S.; Vitiello, G.A.F.; Ferreira, W.A.S.; Chaves, A.S.; Cordeiro de Lima, V.C.; Medina, T.D.S. B Cell Orchestration of Anti-tumor Immune Responses: A Matter of Cell Localization and Communication. Front. Cell Dev. Biol. 2021, 9, 678127. [Google Scholar] [CrossRef]
- Patil, N.S.; Nabet, B.Y.; Müller, S.; Koeppen, H.; Zou, W.; Giltnane, J.; Au-Yeung, A.; Srivats, S.; Cheng, J.H.; Takahashi, C.; et al. Intratumoral plasma cells predict outcomes to PD-L1 blockade in non-small cell lung cancer. Cancer Cell 2022, 40, 289–300.e4. [Google Scholar] [CrossRef]
- Rouanne, M.; Arpaia, N.; Marabelle, A. CXCL13 shapes tertiary lymphoid structures and promotes response to immunotherapy in bladder cancer. Eur. J. Cancer 2021, 151, 245–248. [Google Scholar] [CrossRef]
- Toulmonde, M.; Penel, N.; Adam, J.; Chevreau, C.; Blay, J.Y.; Le Cesne, A.; Bompas, E.; Piperno-Neumann, S.; Cousin, S.; Grellety, T.; et al. Use of PD-1 Targeting, Macrophage Infiltration, and IDO Pathway Activation in Sarcomas: A Phase 2 Clinical Trial. JAMA Oncol. 2018, 4, 93–97. [Google Scholar] [CrossRef]
- Katayama, H.; Boldt, C.; Ladd, J.J.; Johnson, M.M.; Chao, T.; Capello, M.; Suo, J.; Mao, J.; Manson, J.E.; Prentice, R.; et al. An Autoimmune Response Signature Associated with the Development of Triple-Negative Breast Cancer Reflects Disease Pathogenesis. Cancer Res. 2015, 75, 3246–3254. [Google Scholar] [CrossRef] [Green Version]
- Ladd, J.J.; Chao, T.; Johnson, M.M.; Qiu, J.; Chin, A.; Israel, R.; Pitteri, S.J.; Mao, J.; Wu, M.; Amon, L.M.; et al. Autoantibody signatures involving glycolysis and splicesome proteins precede a diagnosis of breast cancer among postmenopausal women. Cancer Res. 2013, 73, 1502–1513. [Google Scholar] [CrossRef] [Green Version]
- Le Naour, F.; Brichory, F.; Misek, D.E.; Bréchot, C.; Hanash, S.M.; Beretta, L. A distinct repertoire of autoantibodies in hepatocellular carcinoma identified by proteomic analysis. Mol. Cell. Proteom. 2002, 1, 197–203. [Google Scholar] [CrossRef] [Green Version]
- Mattioni, M.; Soddu, S.; Prodosmo, A.; Visca, P.; Conti, S.; Alessandrini, G.; Facciolo, F.; Strigari, L. Prognostic role of serum p53 antibodies in lung cancer. BMC Cancer 2015, 15, 148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tsou, P.; Katayama, H.; Ostrin, E.J.; Hanash, S.M. The Emerging Role of B Cells in Tumor Immunity. Cancer Res. 2016, 76, 5597–5601. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weinberg, R.A. The Biology of Cancer, 1st ed.; W.W. Norton & Company: New York, NY, USA, 2007. [Google Scholar]
- Mazor, R.D.; Nathan, N.; Gilboa, A.; Stoler-Barak, L.; Moss, L.; Solomonov, I.; Hanuna, A.; Divinsky, Y.; Shmueli, M.D.; Hezroni, H.; et al. Tumor-reactive antibodies evolve from non-binding and autoreactive precursors. Cell 2022, 185, 1208–1222.e21. [Google Scholar] [CrossRef] [PubMed]
- Wei, R.; Dean, D.C.; Thanindratarn, P.; Hornicek, F.J.; Guo, W.; Duan, Z. Cancer testis antigens in sarcoma: Expression, function and immunotherapeutic application. Cancer Lett. 2020, 479, 54–60. [Google Scholar] [CrossRef] [PubMed]
- Zou, C.; Shen, J.; Tang, Q.; Yang, Z.; Yin, J.; Li, Z.; Xie, X.; Huang, G.; Lev, D.; Wang, J. Cancer-testis antigens expressed in osteosarcoma identified by gene microarray correlate with a poor patient prognosis. Cancer 2012, 118, 1845–1855. [Google Scholar] [CrossRef]
- Iura, K.; Maekawa, A.; Kohashi, K.; Ishii, T.; Bekki, H.; Otsuka, H.; Yamada, Y.; Yamamoto, H.; Harimaya, K.; Iwamoto, Y.; et al. Cancer-testis antigen expression in synovial sarcoma: NY-ESO-1, PRAME, MAGEA4, and MAGEA1. Hum. Pathol. 2017, 61, 130–139. [Google Scholar] [CrossRef]
- Hemminger, J.A.; Toland, A.E.; Scharschmidt, T.J.; Mayerson, J.L.; Guttridge, D.C.; Iwenofu, O.H. Expression of cancer-testis antigens MAGEA1, MAGEA3, ACRBP, PRAME, SSX2, and CTAG2 in myxoid and round cell liposarcoma. Mod. Pathol. 2014, 27, 1238–1245. [Google Scholar] [CrossRef] [Green Version]
- Iura, K.; Kohashi, K.; Ishii, T.; Maekawa, A.; Bekki, H.; Otsuka, H.; Yamada, Y.; Yamamoto, H.; Matsumoto, Y.; Iwamoto, Y.; et al. MAGEA4 expression in bone and soft tissue tumors: Its utility as a target for immunotherapy and diagnostic marker combined with NY-ESO-1. Virchows Arch. 2017, 471, 383–392. [Google Scholar] [CrossRef]
- Morgan, R.A.; Chinnasamy, N.; Abate-Daga, D.; Gros, A.; Robbins, P.F.; Zheng, Z.; Dudley, M.E.; Feldman, S.A.; Yang, J.C.; Sherry, R.M.; et al. Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. J. Immunother. 2013, 36, 133–151. [Google Scholar] [CrossRef] [Green Version]
- Shurell, E.; Vergara-Lluri, M.E.; Li, Y.; Crompton, J.G.; Singh, A.; Bernthal, N.; Wu, H.; Eilber, F.C.; Dry, S.M. Comprehensive adipocytic and neurogenic tissue microarray analysis of NY-ESO-1 expression—A promising immunotherapy target in malignant peripheral nerve sheath tumor and liposarcoma. Oncotarget 2016, 7, 72860–72867. [Google Scholar] [CrossRef] [Green Version]
- Lee, S.Y.; Obata, Y.; Yoshida, M.; Stockert, E.; Williamson, B.; Jungbluth, A.A.; Chen, Y.T.; Old, L.J.; Scanlan, M.J. Immunomic analysis of human sarcoma. Proc. Natl. Acad. Sci. USA 2003, 100, 2651–2656. [Google Scholar] [CrossRef] [Green Version]
- Somaiah, N.; Block, M.S.; Kim, J.W.; Shapiro, G.I.; Do, K.T.; Hwu, P.; Eder, J.P.; Jones, R.L.; Lu, H.; Ter Meulen, J.H.; et al. First-in-Class, First-in-Human Study Evaluating LV305, a Dendritic-Cell Tropic Lentiviral Vector, in Sarcoma and Other Solid Tumors Expressing NY-ESO-1. Clin. Cancer Res. 2019, 25, 5808–5817. [Google Scholar] [CrossRef] [Green Version]
- Rettig, W.J.; Garin-Chesa, P.; Healey, J.H.; Su, S.; Ozer, H.L.; Schwab, M.; Albino, A.P.; Old, L.J. Regulation and heteromeric structure of the fibroblast activation protein in normal and transformed cells of mesenchymal and neuroectodermal origin. Cancer Res. 1993, 53, 3327–3335. [Google Scholar]
- Ding, L.; Yem, L.; Xu, J.; Jiang, W.G. Impact of fibroblast activation protein on osteosarcoma cell lines in vitro. Oncol. Lett. 2014, 7, 699–704. [Google Scholar] [CrossRef]
- Chen, M.; Xiang, R.; Wen, Y.; Xu, G.; Wang, C.; Luo, S.; Yin, T.; Wei, X.; Shao, B.; Liu, N.; et al. A whole-cell tumor vaccine modified to express fibroblast activation protein induces antitumor immunity against both tumor cells and cancer-associated fibroblasts. Sci. Rep. 2015, 5, 14421. [Google Scholar] [CrossRef] [Green Version]
- Eilber, F.C.; Dry, S.M. Diagnosis and management of synovial sarcoma. J. Surg. Oncol. 2008, 97, 314–320. [Google Scholar] [CrossRef]
- Kawaguchi, S.; Tsukahara, T.; Ida, K.; Kimura, S.; Murase, M.; Kano, M.; Emori, M.; Nagoya, S.; Kaya, M.; Torigoe, T.; et al. SYT-SSX breakpoint peptide vaccines in patients with synovial sarcoma: A study from the Japanese Musculoskeletal Oncology Group. Cancer Sci. 2012, 103, 1625–1630. [Google Scholar] [CrossRef]
- Dagher, R.; Long, L.M.; Read, E.J.; Leitman, S.F.; Carter, C.S.; Tsokos, M.; Goletz, T.J.; Avila, N.; Berzofsky, J.A.; Helman, L.J.; et al. Pilot trial of tumor-specific peptide vaccination and continuous infusion interleukin-2 in patients with recurrent Ewing sarcoma and alveolar rhabdomyosarcoma: An inter-institute NIH study. Med. Pediatr. Oncol. 2002, 38, 158–164. [Google Scholar] [CrossRef]
- Hu-Lieskovan, S.; Heidel, J.D.; Bartlett, D.W.; Davis, M.E.; Triche, T.J. Sequence-specific knockdown of EWS-FLI1 by targeted, nonviral delivery of small interfering RNA inhibits tumor growth in a murine model of metastatic Ewing’s sarcoma. Cancer Res. 2005, 65, 8984–8992. [Google Scholar] [CrossRef] [Green Version]
- Liu, H.; Huang, L.; Luo, J.; Chen, W.; Zhang, Z.; Liao, X.; Dai, M.; Shu, Y.; Cao, K. Prediction and identification of B cell epitopes derived from EWS/FLI-l fusion protein of Ewing’s sarcoma. Med. Oncol. 2012, 29, 3421–3430. [Google Scholar] [CrossRef]
- Kaumaya, P.T. B-cell epitope peptide cancer vaccines: A new paradigm for combination immunotherapies with novel checkpoint peptide vaccine. Future Oncol. 2020, 16, 1767–1791. [Google Scholar] [CrossRef] [PubMed]
- Matsuzaki, J.; Gnjatic, S.; Mhawech-Fauceglia, P.; Beck, A.; Miller, A.; Tsuji, T.; Eppolito, C.; Qian, F.; Lele, S.; Shrikant, P.; et al. Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc. Natl. Acad. Sci. USA 2010, 107, 7875–7880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kametani, Y.; Miyamoto, A.; Tsuda, B.; Tokuda, Y. B Cell Epitope-Based Vaccination Therapy. Antibodies 2015, 4, 225–239. [Google Scholar] [CrossRef] [Green Version]
- Kaumaya, P.T. Bridging oncology and immunology: Expanding horizons with innovative peptide vaccines and peptidomimetics. Immunotherapy 2013, 5, 1159–1163. [Google Scholar] [CrossRef] [PubMed]
- Punt, C.J.; Barbuto, J.A.; Zhang, H.; Grimes, W.J.; Hatch, K.D.; Hersh, E.M. Anti-tumor antibody produced by human tumor-infiltrating and peripheral blood B lymphocytes. Cancer Immunol. Immunother. 1994, 38, 225–232. [Google Scholar] [CrossRef]
- Erdag, G.; Schaefer, J.T.; Smolkin, M.E.; Deacon, D.H.; Shea, S.M.; Dengel, L.T.; Patterson, J.W.; Slingluff, C.L., Jr. Immunotype and immunohistologic characteristics of tumor-infiltrating immune cells are associated with clinical outcome in metastatic melanoma. Cancer Res. 2012, 72, 1070–1080. [Google Scholar] [CrossRef] [Green Version]
- Lohr, M.; Edlund, K.; Botling, J.; Hammad, S.; Hellwig, B.; Othman, A.; Berglund, A.; Lambe, M.; Holmberg, L.; Ekman, S. 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. [Google Scholar] [CrossRef]
- Bolotin, D.A.; Poslavsky, S.; Davydov, A.N.; Frenkel, F.E.; Fanchi, L.; Zolotareva, O.I.; Hemmers, S.; Putintseva, E.V.; Obraztsova, A.S.; Shugay, M. Antigen receptor repertoire profiling from RNA-seq data. Nat. Biotechnol. 2017, 35, 908–911. [Google Scholar] [CrossRef]
- Pavoni, E.; Monteriù, G.; Santapaola, D.; Petronzelli, F.; Anastasi, A.M.; Pelliccia, A.; D’Alessio, V.; De Santis, R.; Minenkova, O. Tumor-infiltrating B lymphocytes as an efficient source of highly specific immunoglobulins recognizing tumor cells. BMC Biotechnol. 2007, 7, 70. [Google Scholar] [CrossRef] [Green Version]
- Coronella, J.A.; Spier, C.; Welch, M.; Trevor, K.T.; Stopeck, A.T.; Villar, H.; Hersh, E.M. Antigen-driven oligoclonal expansion of tumor-infiltrating B cells in infiltrating ductal carcinoma of the breast. J. Immunol. 2002, 169, 1829–1836. [Google Scholar] [CrossRef] [Green Version]
- Isaeva, O.I.; Sharonov, G.V.; Serebrovskaya, E.O.; Turchaninova, M.A.; Zaretsky, A.R.; Shugay, M.; Chudakov, D.M. Intratumoral immunoglobulin isotypes predict survival in lung adenocarcinoma subtypes. J. Immunother. Cancer 2019, 7, 279. [Google Scholar] [CrossRef]
- Karagiannis, P.; Gilbert, A.E.; Josephs, D.H.; Ali, N.; Dodev, T.; Saul, L.; Correa, I.; Roberts, L.; Beddowes, E.; Koers, A.; et al. IgG4 subclass antibodies impair antitumor immunity in melanoma. J. Clin. Investig. 2013, 123, 1457–1474. [Google Scholar] [CrossRef] [Green Version]
- Fujimoto, M.; Yoshizawa, A.; Sumiyoshi, S.; Sonobe, M.; Kobayashi, M.; Koyanagi, I.; Aini, W.; Tsuruyama, T.; Date, H.; Haga, H. Stromal plasma cells expressing immunoglobulin G4 subclass in non-small cell lung cancer. Hum. Pathol. 2013, 44, 1569–1576. [Google Scholar] [CrossRef] [Green Version]
- Welinder, C.; Jirström, K.; Lehn, S.; Nodin, B.; Marko-Varga, G.; Blixt, O.; Danielsson, L.; Jansson, B. Intra-tumour IgA1 is common in cancer and is correlated with poor prognosis in bladder cancer. Heliyon 2016, 2, e00143. [Google Scholar] [CrossRef]
- Smolle, M.A.; Herbsthofer, L.; Granegger, B.; Goda, M.; Brcic, I.; Bergovec, M.; Scheipl, S.; Prietl, B.; Pichler, M.; Gerger, A. T-regulatory cells predict clinical outcome in soft tissue sarcoma patients: A clinico-pathological study. Br. J. Cancer 2021, 125, 717–724. [Google Scholar] [CrossRef]
- Qiu, X.; Zhu, X.; Zhang, L.; Mao, Y.; Zhang, J.; Hao, P.; Li, G.; Lv, P.; Li, Z.; Sun, X.; et al. Human epithelial cancers secrete immunoglobulin g with unidentified specificity to promote growth and survival of tumor cells. Cancer Res. 2003, 63, 6488–6495. [Google Scholar]
- Chen, Z.; Huang, X.; Ye, J.; Pan, P.; Cao, Q.; Yang, B.; Li, Z.; Su, M.; Huang, C.; Gu, J. Immunoglobulin G is present in a wide variety of soft tissue tumors and correlates well with proliferation markers and tumor grades. Cancer 2010, 116, 1953–1963. [Google Scholar] [CrossRef]
- Chen, Z.; Li, J.; Xiao, Y.; Zhang, J.; Zhao, Y.; Liu, Y.; Ma, C.; Qiu, Y.; Luo, J.; Huang, G.; et al. Immunoglobulin G locus events in soft tissue sarcoma cell lines. PLoS ONE 2011, 6, e21276. [Google Scholar] [CrossRef] [Green Version]
- Premkumar, K.; Shankar, B.S. TGF-βR inhibitor SB431542 restores immune suppression induced by regulatory B-T cell axis and decreases tumour burden in murine fibrosarcoma. Cancer Immunol. Immunother. 2021, 70, 153–168. [Google Scholar] [CrossRef]
- Fridman, W.H.; Meylan, M.; Petitprez, F.; Sun, C.M.; Italiano, A.; Sautès-Fridman, C. B cells and tertiary lymphoid structures as determinants of tumour immune contexture and clinical outcome. Nat. Rev. Clin. Oncol. 2022, 19, 441–457. [Google Scholar] [CrossRef]
- Joshi, N.S.; Akama-Garren, E.H.; Lu, Y.; Lee, D.Y.; Chang, G.P.; Li, A.; DuPage, M.; Tammela, T.; Kerper, N.R.; Farago, A.F.; et al. Regulatory T Cells in Tumor-Associated Tertiary Lymphoid Structures Suppress Anti-tumor T Cell Responses. Immunity 2015, 43, 579–590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Delvecchio, F.R.; Fincham, R.E.A.; Spear, S.; Clear, A.; Roy-Luzarraga, M.; Balkwill, F.R.; Gribben, J.G.; Bombardieri, M.; Hodivala-Dilke, K.; Capasso, M.; et al. Pancreatic Cancer Chemotherapy Is Potentiated by Induction of Tertiary Lymphoid Structures in Mice. Cell Mol. Gastroenterol. Hepatol. 2021, 12, 1543–1565. [Google Scholar] [CrossRef] [PubMed]
- Hildebrand, K.M.; Singla, A.K.; McNeil, R.; Marritt, K.L.; Hildebrand, K.N.; Zemp, F.; Rajwani, J.; Itani, D.; Bose, P.; Mahoney, D.J.; et al. The KrasG12D;Trp53fl/fl murine model of undifferentiated pleomorphic sarcoma is macrophage dense, lymphocyte poor, and resistant to immune checkpoint blockade. PLoS ONE 2021, 16, e0253864. [Google Scholar] [CrossRef] [PubMed]
- Kirsch, D.G.; Dinulescu, D.M.; Miller, J.B.; Grimm, J.; Santiago, P.M.; Young, N.P.; Nielsen, G.P.; Quade, B.J.; Chaber, C.J.; Schultz, C.P.; et al. A spatially and temporally restricted mouse model of soft tissue sarcoma. Nat. Med. 2007, 13, 992–997. [Google Scholar] [CrossRef]
- Filderman, J.N.; Appleman, M.; Chelvanambi, M.; Taylor, J.L.; Storkus, W.J. STINGing the Tumor Microenvironment to Promote Therapeutic Tertiary Lymphoid Structure Development. Front. Immunol. 2021, 12, 690105. [Google Scholar] [CrossRef]
- Yagawa, Y.; Robertson-Tessi, M.; Zhou, S.L.; Anderson, A.R.A.; Mulé, J.J.; Mailloux, A.W. Systematic Screening of Chemokines to Identify Candidates to Model and Create Ectopic Lymph Node Structures for Cancer Immunotherapy. Sci. Rep. 2017, 7, 15996. [Google Scholar] [CrossRef] [Green Version]
- Edris, B.; Weiskopf, K.; Volkmer, A.K.; Volkmer, J.P.; Willingham, S.B.; Contreras-Trujillo, H.; Liu, J.; Majeti, R.; West, R.B.; Fletcher, J.A.; et al. Antibody therapy targeting the CD47 protein is effective in a model of aggressive metastatic leiomyosarcoma. Proc. Natl. Acad. Sci. USA 2012, 109, 6656–6661. [Google Scholar] [CrossRef] [Green Version]
- Al Shihabi, A.; Davarifar, A.; Nguyen, H.T.L.; Tavanaie, N.; Nelson, S.D.; Yanagawa, J.; Federman, N.; Bernthal, N.; Hornicek, F.; Soragni, A. Personalized chordoma organoids for drug discovery studies. Sci. Adv. 2022, 8, eabl3674. [Google Scholar] [CrossRef]
- Li, Q.; Lao, X.; Pan, Q.; Ning, N.; Yet, J.; Xu, Y.; Li, S.; Chang, A.E. Adoptive transfer of tumor reactive B cells confers host T-cell immunity and tumor regression. Clin. Cancer Res. 2011, 17, 4987–4995. [Google Scholar] [CrossRef]
Study | Histology | B Cell Marker | Detection Method | Number of Positive Samples | Prognostic Association |
---|---|---|---|---|---|
Sorbye [14] | Mixed non-GIST STS | CD20 | IHC | 22/105 | Positive correlation with disease-specific survival |
Tsagozis et al. [11] | Mixed STS | CD20 | IHC | 13/30 | Positive correlation with metastasis-free survival and overall survival |
Tsagozis et al. [11] | Mixed STS (SARC TCGA dataset) | MS4A1 RNA expression | Transcriptional data from SARC TCGA | Not reported | Overall survival |
Zou et al. [12] | Chordoma | CD20 | IHC | 19/54 | None |
Smolle et al. [15] | Mixed STS | CD20 | IHC | n = 188, number positive not reported | Negatively associated with local recurrence |
Chen et al. [16] | Rhabdomyosarcoma and UPS | CD20 and CD3 to identify TLS | IHC | UPS: 2/34 RMS: 19/47 | Not assessed |
Petitprez et al. [17] | Mixed STS (SARC TCGA, GEO accessions GSE21050, GSE21122 and GSE30929 datasets) | BANK1, CD19, CD22, CD79A, CR2, FCRL2, IGKC, MS4A1 and PAX5 | Transcriptional data from TCGA and GEO | Not reported | Positively correlated with overall survival and response to pembrolizumab |
Italiano et al. [32] | Mixed STS | CD20 and CD3 to identify TLS | IHC | 30/249 in prospective group 1/41 in all-comers retrospective group | Positively correlated with six-month, non-progression rate and progression-free survival |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Kendal, J.K.; Shehata, M.S.; Lofftus, S.Y.; Crompton, J.G. Cancer-Associated B Cells in Sarcoma. Cancers 2023, 15, 622. https://doi.org/10.3390/cancers15030622
Kendal JK, Shehata MS, Lofftus SY, Crompton JG. Cancer-Associated B Cells in Sarcoma. Cancers. 2023; 15(3):622. https://doi.org/10.3390/cancers15030622
Chicago/Turabian StyleKendal, Joseph K., Michael S. Shehata, Serena Y. Lofftus, and Joseph G. Crompton. 2023. "Cancer-Associated B Cells in Sarcoma" Cancers 15, no. 3: 622. https://doi.org/10.3390/cancers15030622
APA StyleKendal, J. K., Shehata, M. S., Lofftus, S. Y., & Crompton, J. G. (2023). Cancer-Associated B Cells in Sarcoma. Cancers, 15(3), 622. https://doi.org/10.3390/cancers15030622