CAR-T Cells in the Treatment of Ovarian Cancer: A Promising Cell Therapy
Abstract
:1. Introduction
2. CAR-T Cells Preparation
3. Antigen Targets
3.1. ERBB2
3.2. MSLN
3.3. MUC16
3.4. EPCAM
3.5. FOLR1
3.6. AMHR2
3.7. ANXA2
3.8. TPBG
3.9. CD24
3.10. PDCD1 and CD274
4. Challenges for CAR-T-Cell Therapy
4.1. Off-Target Effects
4.2. Tumor Antigen Escape
4.3. Heterogeneity of Ovarian Tumors
4.4. Immune Checkpoint
4.5. T-Cell Exhaustion
4.6. Efficiency of CAR-T Cells after Reaching the Tumor Site Is Difficult to Guarantee
4.7. TME of Tumor Immunosuppressive Cells
4.7.1. Immunosuppressive Cells
4.7.2. Reactive Oxygen Species (ROS)
4.7.3. Metabolites
4.7.4. Cytokines
4.7.5. PH Value
4.7.6. Hypoxia
4.8. Cytokine Release Syndrome (CRS)
5. Strategies to Enhance the Efficacy of CAR-T Cells
5.1. Armored CAR-T
5.2. Dual Receptors and Chemokine Receptors
5.3. NK Cell Receptors
5.4. Improving CAR-T-Cell Persistence
5.5. Tumor Vessel-Targeted CAR-T Cells
5.6. Promoting Homing and Aggregation of CAR-T Cells in Tumor Tissue
5.7. Multiantigen-Targeted CAR-T Cells
5.8. Combination with Checkpoint Blockers
6. CAR-T Cells in Combination with Radiotherapy or Chemotherapy
6.1. CAR-T-Cell Therapy in Combination with Chemotherapy
6.1.1. Chemotherapy Improves the Efficacy of CAR-T Therapy
6.1.2. Chemotherapy Enhances the Sensitivity of Tumor Cells to Immunotherapy
6.1.3. Chemotherapy Improves Tumor Antigen Expression and Recognition
6.1.4. Chemotherapy Relieves Immunosuppressive Cells
6.1.5. Chemotherapy Improves CAR-T-Cell Durability
6.1.6. CAR-T Cells Improve the Treatment Effect of Chemotherapy
6.2. CAR-T-Cell Therapy in Combination with Radiotherapy
6.2.1. Radiotherapy Improves the Efficacy of CAR-T Therapy
6.2.2. Radiation Increases the Sensitivity of Tumor Cells to Tumor-Specific Cytotoxic Lymphocytes
6.2.3. Radiotherapy Improves CAR-T-Cell Delivery
6.2.4. Radiation Therapy Improves Tumor Antigen Presentation
6.2.5. CAR-T Cells Improve the Treatment Effect of Radiotherapy
6.3. CAR-T-Cell Therapy in Combination with Radiation and Chemotherapy
7. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Yan, W.; Hu, H.; Tang, B. Advances of Chimeric Antigen Receptor T Cell Therapy in Ovarian Cancer. OncoTargets Ther. 2019, 12, 8015–8022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- International Agency for Research on Cancer. Survival, Incidence, and Mortality over Time. Available online: https://gco.iarc.fr/survival/survmark/ (accessed on 23 December 2022).
- Yan, L.; Liu, B. Critical factors in chimeric antigen receptor-modified T-cell (CAR-T) therapy for solid tumors. OncoTargets Ther. 2018, 12, 193–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Majzner, R.G.; Mackall, C.L. Tumor Antigen Escape from CAR T-cell Therapy. Cancer Discov. 2018, 8, 1219–1226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dudley, M.E.; Wunderlich, J.R.; Robbins, P.F.; Yang, J.C.; Hwu, P.; Schwartzentruber, D.J.; Topalian, S.L.; Sherry, R.; Restifo, N.P.; Hubicki, A.M.; et al. Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science 2002, 298, 850–854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rosenberg, S.A.; Yang, J.C.; Sherry, R.M.; Kammula, U.S.; Hughes, M.S.; Phan, G.Q.; Citrin, D.E.; Restifo, N.P.; Robbins, P.F.; Wunderlich, J.R.; et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin. Cancer Res. 2011, 17, 4550–4557. [Google Scholar] [CrossRef] [Green Version]
- Goff, S.L.; Dudley, M.E.; Citrin, D.E.; Somerville, R.P.; Wunderlich, J.R.; Danforth, D.N.; Zlott, D.A.; Yang, J.C.; Sherry, R.M.; Kammula, U.S.; et al. Randomized, prospective evaluation comparing intensity of lymphodepletion before adoptive transfer of tumor-infiltrating lymphocytes for patients with metastatic melanoma. J. Clin. Oncol. 2016, 34, 2389–2397. [Google Scholar] [CrossRef]
- Kuwana, Y.; Asakura, Y.; Utsunomiya, N.; Nakanishi, M.; Arata, Y.; Itoh, S.; Nagase, F.; Kurosawa, Y. Expression of chimeric receptor composed of immunoglobulin-derived V resions and T-cell receptor-derived C regions. Biochem. Biophys. Res. Commun. 1987, 149, 960–968. [Google Scholar] [CrossRef]
- Gross, G.; Waks, T.; Eshhar, Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc. Natl. Acad. Sci. USA 1989, 86, 10024–10028. [Google Scholar] [CrossRef] [Green Version]
- Miliotou, A.N.; Papadopoulou, L.C. CAR T-cell Therapy: A New Era in Cancer Immunotherapy. Curr. Pharm. Biotechnol. 2018, 19, 5–18. [Google Scholar] [CrossRef]
- Odunsi, K. Immunotherapy in ovarian cancer. Ann. Oncol. 2017, 28, viii1–viii7. [Google Scholar] [CrossRef]
- Wagner, J.; Wickman, E.; DeRenzo, C.; Gottschalk, S. CAR T Cell Therapy for Solid Tumors: Bright Future or Dark Reality? Mol. Ther. 2020, 28, 2320–2339. [Google Scholar] [CrossRef]
- Zhang, C.; Liu, J.; Zhong, J.F.; Zhang, X. Engineering CAR-T cells. Biomark. Res. 2017, 5, 22. [Google Scholar] [CrossRef] [Green Version]
- Sadelain, M.; Brentjens, R.; Riviere, I. The basic principles of chimeric antigen receptor design. Cancer Discov. 2013, 3, 388–398. [Google Scholar] [CrossRef] [Green Version]
- Geldres, C.; Savoldo, B.; Dotti, G. Chimeric antigen receptor-redirected T cells return to the bench. Semin. Immunol. 2016, 28, 3–9. [Google Scholar] [CrossRef] [Green Version]
- Ma, S.; Li, X.; Wang, X.; Cheng, L.; Li, Z.; Zhang, C.; Ye, Z.; Qian, Q. Current Progress in CAR-T Cell Therapy for Solid Tumors. Int. J. Biol. Sci. 2019, 15, 2548–2560. [Google Scholar] [CrossRef] [Green Version]
- Martin, V.; Cappuzzo, F.; Mazzucchelli, L.; Frattini, M. HER2 in solid tumors: More than 10 years under the microscope; where are we now? Future Oncol. 2014, 10, 1469–1486. [Google Scholar] [CrossRef]
- Chang, K.-L.; Lee, M.-Y.; Chao, W.-R.; Han, C.-P. The status of Her2 amplification and Kras mutations in mucinous ovarian carcinoma. Hum. Genom. 2016, 10, 40. [Google Scholar] [CrossRef] [Green Version]
- Sun, M.; Shi, H.; Liu, C.; Liu, J.; Liu, X.; Sun, Y. Construction and evaluation of a novel humanized HER2-specific chimeric receptor. Breast Cancer Res. 2014, 16, R61. [Google Scholar] [CrossRef] [Green Version]
- Hassan, R.; Thomas, A.; Alewine, C.; Le, D.T.; Jaffee, E.M.; Pastan, I. Mesothelin Immunotherapy for Cancer: Ready for Prime Time? J. Clin. Oncol. 2016, 34, 4171–4179. [Google Scholar] [CrossRef] [Green Version]
- Beatty, G.L.; Haas, A.R.; Maus, M.V.; Torigian, D.A.; Soulen, M.C.; Plesa, G.; Chew, A.; Zhao, Y.; Levine, B.L.; Albelda, S.M.; et al. Mesothelin-specific chimeric antigen receptor mRNA-engineered T cells induce anti-tumor activity in solid malignancies. Cancer Immunol. Res. 2014, 2, 112–120. [Google Scholar] [CrossRef] [Green Version]
- Morello, A.; Sadelain, M.; Adusumilli, P.S. Mesothelin-Targeted CARs: Driving T Cells to Solid Tumors. Cancer Discov. 2016, 6, 133–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Okła, K.; Surówka, J.; Frąszczak, K.; Czerwonka, A.; Kaławaj, K.; Wawruszak, A.; Kotarski, J.; Wertel, I. Assessment of the clinicopathological relevance of mesothelin level in plasma, peritoneal fluid, and tumor tissue of epithelial ovarian cancer patients. Tumor Biol. 2018, 40, 1010428318804937. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Adusumilli, P.S.; Cherkassky, L.; Villena-Vargas, J.; Colovos, C.; Servais, E.; Plotkin, J.; Jones, D.R.; Sadelain, M. Regional delivery of mesothelin-targeted CAR T cell therapy generates potent and long-lasting CD4-dependent tumor immunity. Sci. Transl. Med. 2014, 6, 261ra151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Q.; Liu, G.; Liu, J.; Yang, M.; Fu, J.; Liu, G.; Li, D.; Gu, Z.; Zhang, L.; Pan, Y.; et al. The antitumor capacity of mesothelin-CAR-T cells in targeting solid tumors in mice. Mol. Ther. Oncolytics 2021, 20, 556–568. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Rao, T.D.; Tian, H.; Ma, X.; Yan, X.; Thapi, S.; Schultz, N.; Rosales, N.; Monette, S.; Wang, A.; et al. Expression of the Carboxy-Terminal Portion of MUC16/CA125 Induces Transformation and Tumor Invasion. PLoS ONE 2015, 10, e0126633. [Google Scholar] [CrossRef] [Green Version]
- Chekmasova, A.A.; Rao, T.D.; Nikhamin, Y.; Park, K.J.; Levine, D.A.; Spriggs, D.R.; Brentjens, R.J. Successful eradication of established peritoneal ovarian tumors in SCID-Beige mice following adoptive transfer of T cells genetically targeted to the MUC16 antigen. Clin. Cancer Res. 2010, 16, 3594–3606. [Google Scholar] [CrossRef] [Green Version]
- Fu, J.; Shang, Y.; Qian, Z.; Hou, J.; Yan, F.; Liu, G.; Li, D.; Tian, X. Chimeric Antigen receptor-T (CAR-T) cells targeting Epithelial cell adhesion molecule (EpCAM) can inhibit tumor growth in ovarian cancer mouse model. J. Vet. Med. Sci. 2021, 83, 241–247. [Google Scholar] [CrossRef]
- Kurosaki, A.; Hasegawa, K.; Kato, T.; Abe, K.; Hanaoka, T.; Miyara, A.; O’Shannessy, D.J.; Somers, E.B.; Yasuda, M.; Sekino, T.; et al. Serum folate receptor alpha as a biomarker for ovarian cancer: Implications for diagnosis, prognosis and predicting its local tumor expression. Int. J. Cancer 2016, 138, 1994–2002. [Google Scholar] [CrossRef] [Green Version]
- Zuo, S.; Wen, Y.; Panha, H.; Dai, G.; Wang, L.; Ren, X.; Fu, K. Modification of cytokine-induced killer cells with folate receptor alpha (FRalpha)-specific chimeric antigen receptors enhances their antitumor immunity toward FRalpha-positive ovarian cancers. Mol. Immunol. 2017, 85, 293–304. [Google Scholar] [CrossRef]
- Kandalaft, L.E.; Powell, D.J., Jr.; Coukos, G. A phase I clinical trial of adoptive transfer of folate receptor-alpha redirected autologous T cells for recurrent ovarian cancer. J. Transl. Med. 2012, 10, 157. [Google Scholar] [CrossRef] [Green Version]
- Ramos, C.A.; Rouce, R.; Robertson, C.S.; Reyna, A.; Narala, N.; Vyas, G.; Mehta, B.; Zhang, H.; Dakhova, O.; Carrum, G.; et al. In Vivo Fate and Activity of Second- versus Third-Generation CD19-Specific CAR-T Cells in B Cell Non-Hodgkin’s Lymphomas. Mol. Ther. 2018, 26, 2727–2737. [Google Scholar] [CrossRef] [Green Version]
- Leong, L.; Heng Liang, T.; Cua, S.; Yong, K.S.M.; Chen, Q.; Choo, A. Preclinical Activity of Embryonic Annexin A2-Specific Chimeric Antigen Receptor T Cells Against Ovarian Cancer. Int. J. Mol. Sci. 2020, 21, 381. [Google Scholar] [CrossRef] [Green Version]
- Guo, C.; Dong, E.; Lai, Q.; Zhou, S.; Zhang, G.; Wu, M.; Yue, X.; Tao, Y.; Peng, Y.; Ali, J.; et al. Effective antitumor activity of 5T4-specific CAR-T cells against ovarian cancer cells in vitro and xenotransplanted tumors in vivo. MedComm 2020, 1, 338–350. [Google Scholar] [CrossRef]
- Klapdor, R.; Wang, S.; Morgan, M.; Doerk, T.; Hacker, U.; Hillemanns, P.; Buening, H.; Schambach, A. Characterization of a Novel Third-Generation Anti-CD24-CAR against Ovarian Cancer. Int. J. Mol. Sci. 2019, 20, 660. [Google Scholar] [CrossRef] [Green Version]
- Chardin, L.; Leary, A. Immunotherapy in Ovarian Cancer: Thinking Beyond PD-1/PD-L1. Front. Oncol. 2021, 11, 795547. [Google Scholar] [CrossRef]
- Cherkassky, L.; Morello, A.; Villena-Vargas, J.; Feng, Y.; Dimitrov, D.S.; Jones, D.R.; Sadelain, M.; Adusumilli, P.S. Human CAR T cells with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated inhibition. J. Clin. Investig. 2016, 126, 3130–3144. [Google Scholar] [CrossRef] [Green Version]
- Moon, E.K.; Ranganathan, R.; Eruslanov, E.; Kim, S.; Newick, K.; O’Brien, S.; Lo, A.; Liu, X.J.; Zhao, Y.B.; Albelda, S.M. Blockade of Programmed Death 1 Augments the Ability of Human T Cells Engineered to Target NY-ESO-1 to Control Tumor Growth after Adoptive Transfer. Clin. Cancer Res. 2016, 22, 436–477. [Google Scholar] [CrossRef] [Green Version]
- Rafiq, S.; Yeku, O.O.; Jackson, H.J.; Purdon, T.J.; van Leeuwen, D.G.; Drakes, D.J.; Song, M.; Miele, M.M.; Li, Z.; Wang, P.; et al. Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo. Nat. Biotechnol. 2018, 36, 847–856. [Google Scholar] [CrossRef]
- Fang, J.; Ding, N.; Guo, X.; Sun, Y.; Zhang, Z.; Xie, B.; Li, Z.; Wang, H.; Mao, W.; Lin, Z.; et al. αPD-1-mesoCAR-T cells partially inhibit the growth of advanced/refractory ovarian cancer in a patient along with daily apatinib. J. Immunother. Cancer 2021, 9, e001162. [Google Scholar] [CrossRef]
- Li, T.; Wang, J. Therapeutic effect of dual CAR-T targeting PDL1 and MUC16 antigens on ovarian cancer cells in mice. BMC Cancer 2020, 20, 678. [Google Scholar] [CrossRef]
- O’Rourke, D.M.; Nasrallah, M.P.; Desai, A.; Melenhorst, J.J.; Mansfield, K.; Morrissette, J.J.D.; Martinez-Lage, M.; Brem, S.; Maloney, E.; Shen, A.; et al. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci. Transl. Med. 2017, 9, eaaa0984. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gardner, R.; Wu, D.; Cherian, S.; Fang, M.; Hanafi, L.A.; Finney, O.; Smithers, H.; Jensen, M.C.; Riddell, S.R.; Maloney, D.G.; et al. Acquisition of a CD19-negative myeloid phenotype allows immune escape of MLL-rearranged B-ALL from CD19 CAR-T-cell therapy. Blood 2016, 127, 2406–2410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gardner, R.; Finney, O.; Smithers, H. CD19CAR T cell products of defined CD4:CD8 composition and transgene expression show prolonged persistence and durable MRD-negative remission in pediatric and young adult B-cell ALL. Blood 2016, 128, 219. [Google Scholar] [CrossRef]
- Majzner, R.G.; Heitzeneder, S.; Mackall, C.L. Harnessing the Immunotherapy Revolution for the Treatment of Childhood Cancers. Cancer Cell 2017, 31, 476–485. [Google Scholar] [CrossRef] [Green Version]
- Kosti, P.; Maher, J.; Arnold, J.N. Perspectives on chimeric antigen receptor T-cell immunotherapy for solid tumors. Front. Immunol. 2018, 9, 1104. [Google Scholar] [CrossRef] [Green Version]
- Buchbinder, E.I.; Desai, A. CTLA-4 and PD-1 pathways similarities, differences, and implications of their inhibition. Am. J. Clin. Oncol. Cancer Clin. Trials 2016, 39, 98–106. [Google Scholar] [CrossRef] [Green Version]
- Anderson, A.C.; Joller, N.; Kuchroo, V.K. Lag-3, Tim-3, and TIGIT: Co-inhibitory Receptors with Specialized Functions in Immune Regulation. Immunity 2016, 44, 989–1004. [Google Scholar] [CrossRef] [Green Version]
- Parry, R.V.; Chemnitz, J.M.; Frauwirth, K.A.; Lanfranco, A.R.; Braunstein, I.; Kobayashi, S.V.; Linsley, P.S.; Thompson, C.B.; Riley, J.L. CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol. Cell. Biol. 2005, 25, 9543–9553. [Google Scholar] [CrossRef] [Green Version]
- Mirzaei, H.R.; Rodriguez, A.; Shepphird, J.; Brown, C.E.; Badie, B. Chimeric antigen receptors T cell therapy in solid tumor: Challenges and clinical applications. Front. Immunol. 2017, 8, 1850. [Google Scholar] [CrossRef] [Green Version]
- Johnson, L.A.; June, C.H. Driving gene-engineered T cell immunotherapy of cancer. Cell Res. 2017, 27, 38–58. [Google Scholar] [CrossRef] [Green Version]
- Maus, M.V.; Haas, A.R.; Beatty, G.L.; Albelda, S.M.; Levine, B.L.; Liu, X.; Zhao, Y.; Kalos, M.; June, C.H. T cells expressing chimeric antigen receptors can cause anaphylaxis in humans. Cancer Immunol. Res. 2013, 1, 26–31. [Google Scholar] [CrossRef] [Green Version]
- Butte, M.J.; Peña-Cruz, V.; Kim, M.J.; Freeman, G.J.; Sharpe, A.H. Interaction of human PD-L1 and B7-1. Mol. Immunol. 2008, 45, 3567–3572. [Google Scholar] [CrossRef] [Green Version]
- Freeman, G.J.; Long, A.J.; Iwai, Y.; Bourque, K.; Chernova, T.; Nishimura, H.; Fitz, L.J.; Malenkovich, N.; Okazaki, T.; Byrne, M.C.; et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J. Exp. Med. 2000, 192, 1027–1034. [Google Scholar] [CrossRef] [Green Version]
- Dong, Y.; Sun, Q.; Zhang, X. PD-1 and its ligands are important immune checkpoints in cancer. Oncotarget 2017, 8, 2171–2186. [Google Scholar] [CrossRef] [Green Version]
- Keir, M.E.; Butte, M.J.; Freeman, G.J.; Sharpe, A.H. PD-1 and its ligands in tolerance and immunity. Annu. Rev. Immunol. 2008, 26, 677–704. [Google Scholar] [CrossRef] [Green Version]
- Egen, J.G.; Kuhns, M.S.; Allison, J.P. CTLA-4: New insights into its biological function and use in tumor immunotherapy. Nat. Immunol. 2002, 3, 611–618. [Google Scholar] [CrossRef]
- Chambers, C.A.; Kuhns, M.S.; Egen, J.G.; Allison, J.P. CTLA-4-mediated inhibition in regulation of T cell responses: Mechanisms and manipulation in tumor immunotherapy. Annu. Rev. Immunol. 2001, 19, 565–594. [Google Scholar] [CrossRef] [Green Version]
- McGowan, E.; Lin, Q.; Ma, G.; Yin, H.; Chen, S.; Lin, Y. PD-1 disrupted CAR-T cells in the treatment of solid tumors: Promises and challenges. Biomed. Pharmacother. 2020, 121, 109625. [Google Scholar] [CrossRef]
- Joller, N.; Kuchroo, V.K. Tim-3, Lag-3, and TIGIT. Curr. Top. Microbiol. Immunol. 2017, 410, 127–156. [Google Scholar] [CrossRef] [Green Version]
- Chen, J.; Lopez-Moyado, I.F.; Seo, H.; Lio, C.J.; Hempleman, L.J.; Sekiya, T.; Yoshimura, A.; Scott-Browne, J.P.; Rao, A. NR4A transcription factors limit CAR T cell function in solid tumours. Nature 2019, 567, 530–534. [Google Scholar] [CrossRef]
- Kang, C.; Jeong, S.Y.; Song, S.Y.; Choi, E.K. The emerging role of myeloid-derived suppressor cells in radiotherapy. Radiat. Oncol. J. 2020, 38, 1–10. [Google Scholar] [CrossRef] [PubMed]
- Gabrilovich, D.I.; Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 2009, 9, 162–174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cassetta, L.; Pollard, J.W. Tumor-associated macrophages. Curr. Biol. 2020, 30, R246–R248. [Google Scholar] [CrossRef] [PubMed]
- Weinberg, F.; Ramnath, N.; Nagrath, D. Reactive oxygen species in the tumor microenvironment: An overview. Cancers 2019, 11, 1191. [Google Scholar] [CrossRef] [Green Version]
- Liou, G.Y.; Storz, P. Reactive oxygen species in cancer. Free Radic. Res. 2010, 44, 479–496. [Google Scholar] [CrossRef] [Green Version]
- Yang, Z.; Guo, J.; Weng, L.; Tang, W.; Jin, S.; Ma, W. Myeloid-derived suppressor cells-new and exciting players in lung cancer. J. Hematol. Oncol. 2020, 13, 10. [Google Scholar] [CrossRef]
- Ohl, K.; Tenbrock, K. Reactive Oxygen Species as Regulators of MDSC-Mediated Immune Suppression. Front. Immunol. 2018, 9, 2499. [Google Scholar] [CrossRef] [Green Version]
- Klaunig, J.E. Oxidative stress and cancer. Curr. Pharm. Des. 2018, 24, 4771–4778. [Google Scholar] [CrossRef]
- Ligtenberg, M.A.; Mougiakakos, D.; Mukhopadhyay, M.; Witt, K.; Lladser, A.; Chmielewski, M.; Riet, T.; Abken, H.; Kiessling, R. Coexpressed Catalase Protects Chimeric Antigen Receptor-Redirected T Cells as well as Bystander Cells from Oxidative Stress-Induced Loss of Antitumor Activity. J. Immunol. 2016, 196, 759–766. [Google Scholar] [CrossRef] [Green Version]
- Baumann, T.; Dunkel, A.; Schmid, C.; Schmitt, S.; Hiltensperger, M.; Lohr, K.; Laketa, V.; Donakonda, S.; Ahting, U.; Lorenz-Depiereux, B.; et al. Regulatory myeloid cells paralyze T cells through cell–cell transfer of the metabolite methylglyoxal. Nat. Immunol. 2020, 21, 555–566. [Google Scholar] [CrossRef]
- de la Cruz-López, K.G.; Castro-Muñoz, L.J.; Reyes-Hernández, D.O.; García-Carrancá, A.; Manzo-Merino, J. Lactate in the Regulation of Tumor Microenvironment and Therapeutic Approaches. Front. Oncol. 2019, 9, 1143. [Google Scholar] [CrossRef] [Green Version]
- Arab, S.; Hadjati, J. Adenosine blockage in tumor microenvironment and improvement of cancer immunotherapy. Immune Netw. 2019, 19, e23. [Google Scholar] [CrossRef]
- Huang, Q.; Xia, J.; Wang, L.; Wang, X.; Ma, X.; Deng, Q.; Lu, Y.; Kumar, M.; Zhou, Z.; Li, L.; et al. MiR-153 suppresses IDO1 expression and enhances CAR T cell immunotherapy. J. Hematol. Oncol. 2018, 11, 58. [Google Scholar] [CrossRef] [Green Version]
- Rodriguez, P.C.; Quiceno, D.G.; Zabaleta, J.; Ortiz, B.; Zea, A.H.; Piazuelo, M.B.; Delgado, A.; Correa, P.; Brayer, J.; Sotomayor, E.M.; et al. Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Res. 2004, 64, 5839–5849. [Google Scholar] [CrossRef] [Green Version]
- Bronte, V.; Serafini, P.; Mazzoni, A.; Segal, D.M.; Zanovello, P. L-arginine metabolism in myeloid cells controls T-lymphocyte functions. Trends Immunol. 2003, 24, 301–305. [Google Scholar] [CrossRef]
- Cheng, J.; Zhao, L.; Zhang, Y.; Qin, Y.; Guan, Y.; Zhang, T.; Liu, C.; Zhou, J. Understanding the Mechanisms of Resistance to CAR T-Cell Therapy in Malignancies. Front. Oncol. 2019, 9, 1237. [Google Scholar] [CrossRef] [Green Version]
- Juillerat, A.; Marechal, A.; Filhol, J.M.; Valogne, Y.; Valton, J.; Duclert, A.; Duchateau, P.; Poirot, L. An oxygen sensitive self-decision making engineered CAR T-cell. Sci. Rep. 2017, 7, 39833. [Google Scholar] [CrossRef] [Green Version]
- Han, S.; Latchoumanin, O.; Wu, G.; Zhou, G.; Hebbard, L.; George, J.; Qiao, L. Recent clinical trials utilizing chimeric antigen receptor T cells therapies against solid tumors. Cancer Lett. 2017, 390, 188–200. [Google Scholar] [CrossRef]
- Bollong, M.J.; Lee, G.; Coukos, J.S.; Yun, H.; Zambaldo, C.; Chang, J.W.; Chin, E.N.; Ahmad, I.; Chatterjee, A.K.; Lairson, L.L.; et al. A metabolite-derived protein modification integrates glycolysis with KEAP1–NRF2 signalling. Nature 2018, 562, 600–604. [Google Scholar] [CrossRef]
- Nokin, M.J.; Durieux, F.; Peixoto, P.; Chiavarina, B.; Peulen, O.; Blomme, A.; Turtoi, A.; Costanza, B.; Smargiasso, N.; Baiwir, D.; et al. Methylglyoxal, a glycolysis side-product, induces Hsp90 glycation and YAP- mediated tumor growth and metastasis. eLife 2016, 5, e19375. [Google Scholar] [CrossRef]
- Xu, X.; Gnanaprakasam, J.N.R.; Sherman, J.; Wang, R. A Metabolism Toolbox for CAR T Therapy. Front. Oncol. 2019, 9, 322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Koneru, M.; Purdon, T.J.; Spriggs, D.; Koneru, S.; Brentjens, R.J. IL-12 secreting tumor-targeted chimeric antigen receptor T cells eradicate ovarian tumors in vivo. OncoImmunology 2015, 4, e994446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Taylor, A.; Verhagen, J.; Blaser, K.; Akdis, M.; Akdis, C.A. Mechanisms of immune suppression by interleukin-10 and transforming growth factor-β: The role of T regulatory cells. Immunology 2006, 117, 433–442. [Google Scholar] [CrossRef] [PubMed]
- Law, A.M.K.; Valdes-Mora, F.; Gallego-Ortega, D. Myeloid-Derived Suppressor Cells as a Therapeutic Target for Cancer. Cells 2020, 9, 561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dennis, K.L.; Blatner, N.R.; Gounari, F.; Khazaie, K. Current status of interleukin-10 and regulatory T-cells in cancer. Curr. Opin. Oncol. 2013, 25, 637–645. [Google Scholar] [CrossRef]
- Massagué, J. TGFβ in Cancer. Cell 2008, 134, 215–230. [Google Scholar] [CrossRef] [Green Version]
- Yeh, H.W.; Lee, S.S.; Chang, C.Y.; Lang, Y.D.; Jou, Y.S. A new switch for TGFβ in cancer. Cancer Res. 2019, 79, 3797–3805. [Google Scholar] [CrossRef] [Green Version]
- Dahmani, A.; Delisle, J.S. TGF-β in T cell biology: Implications for cancer immunotherapy. Cancers 2018, 10, 194. [Google Scholar] [CrossRef] [Green Version]
- Bailey, K.M.; Wojtkowiak, J.W.; Hashim, A.I.; Gillies, R.J. Targeting the Metabolic Microenvironment of Tumors. Adv. Pharmacol. 2012, 65, 63–107. [Google Scholar] [CrossRef] [Green Version]
- Zhang, X.; Lin, Y.; Gillies, R.J. Tumor pH and its measurement. J. Nucl. Med. 2010, 51, 1167–1170. [Google Scholar] [CrossRef] [Green Version]
- Zhou, Y.; Chen, X.; Cao, J.; Gao, H. Overcoming the biological barriers in the tumor microenvironment for improving drug delivery and efficacy. J. Mater. Chem. B 2020, 8, 6765–6781. [Google Scholar] [CrossRef]
- Kato, Y.; Ozawa, S.; Miyamoto, C.; Maehata, Y.; Suzuki, A.; Maeda, T.; Baba, Y. Acidic extracellular microenvironment and cancer. Cancer Cell Int. 2013, 13, 89. [Google Scholar] [CrossRef] [Green Version]
- Ackerman, D.; Simon, M.C. Hypoxia, lipids, and cancer: Surviving the harsh tumor microenvironment. Trends Cell Biol. 2014, 24, 472–478. [Google Scholar] [CrossRef] [Green Version]
- Vaupel, P.; Mayer, A. Hypoxia in cancer: Significance and impact on clinical outcome. Cancer Metastasis Rev. 2007, 26, 225–239. [Google Scholar] [CrossRef]
- Martinez, M.; Moon, E.K. CAR T cells for solid tumors: New strategies for finding, infiltrating, and surviving in the tumor microenvironment. Front. Immunol. 2019, 10, 128. [Google Scholar] [CrossRef] [Green Version]
- Brown, J.M.; Wilson, W.R. Exploiting tumour hypoxia in cancer treatment. Nat. Rev. Cancer 2004, 4, 437–447. [Google Scholar] [CrossRef]
- Digre, A.; Singh, K.; Åbrink, M.; Reijmers, R.M.; Sandler, S.; Vlodavsky, I.; Li, J.P. Overexpression of heparanase enhances T lymphocyte activities and intensifies the inflammatory response in a model of murine rheumatoid arthritis. Sci. Rep. 2017, 7, 46229. [Google Scholar] [CrossRef] [Green Version]
- Shimabukuro-Vornhagen, A.; Gödel, P.; Subklewe, M.; Stemmler, H.J.; Schlößer, H.A.; Schlaak, M.; Kochanek, M.; Böll, B.; von Bergwelt-Baildon, M.S. Cytokine release syndrome. J. ImmunoTher. Cancer 2018, 6, 56. [Google Scholar] [CrossRef] [Green Version]
- Chmielewski, M.; Kopecky, C.; Hombach, A.A.; Abken, H. IL-12 release by engineered T cells expressing chimeric antigen receptors can effectively muster an antigen-independent macrophage response on tumor cells that have shut down tumor antigen expression. Cancer Res. 2011, 71, 5697–5706. [Google Scholar] [CrossRef] [Green Version]
- Kerkar, S.P.; Goldszmid, R.S.; Muranski, P.; Chinnasamy, D.; Yu, Z.; Reger, R.N.; Leonardi, A.J.; Morgan, R.A.; Wang, E.; Marincola, F.M.; et al. IL-12 triggers a programmatic change in dysfunctional myeloid-derived cells within mouse tumors. J. Clin. Investig. 2011, 121, 4746–4757. [Google Scholar] [CrossRef] [Green Version]
- Chinnasamy, D.; Yu, Z.; Kerkar, S.P.; Zhang, L.; Morgan, R.A.; Restifo, N.P.; Rosenberg, S.A. Local delivery of interleukin-12 using T cells targeting VEGF receptor-2 eradicates multiple vascularized tumors in mice. Clin. Cancer Res. 2012, 18, 1672–1683. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pegram, H.J.; Lee, J.C.; Hayman, E.G.; Imperato, G.H.; Tedder, T.F.; Sadelain, M.; Brentjens, R.J. Tumor-targeted T cells modified to secrete IL-12 eradicate systemic tumors without need for prior conditioning. Blood 2012, 119, 4133–4141. [Google Scholar] [CrossRef] [PubMed]
- Krenciute, G.; Prinzing, B.L.; Yi, Z.; Wu, M.F.; Liu, H.; Dotti, G.; Balyasnikova, I.V.; Gottschalk, S. Transgenic expression of IL15 improves antiglioma activity of IL13Rα2-CAR T cells but results in antigen loss variants. Cancer Immunol. Res. 2017, 5, 571–581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hu, B.; Ren, J.; Luo, Y.; Keith, B.; Young, R.M.; Scholler, J.; Zhao, Y.; June, C.H. Augmentation of Antitumor Immunity by Human and Mouse CAR T Cells Secreting IL-18. Cell Rep. 2017, 20, 3025–3033. [Google Scholar] [CrossRef] [Green Version]
- Spolski, R.; Leonard, W.J. Interleukin-21: A double-edged sword with therapeutic potential. Nat. Rev. Drug Discov. 2014, 13, 379–395. [Google Scholar] [CrossRef]
- Wilkie, S.; Burbridge, S.E.; Chiapero-Stanke, L.; Pereira, A.C.P.; Cleary, S.; Van Der Stegen, S.J.C.; Spicer, J.F.; Davies, D.M.; Maher, J. Selective expansion of chimeric antigen receptor-targeted T-cells with potent effector function using interleukin-4. J. Biol. Chem. 2010, 285, 25538–25544. [Google Scholar] [CrossRef] [Green Version]
- Davies, D.M.; Foster, J.; van der Stegen, S.J.C.; Parente-Pereira, A.C.; Chiapero-Stanke, L.; Delinassios, G.J.; Burbridge, S.E.; Kao, V.; Liu, Z.; Bosshard-Carter, L.; et al. Flexible targeting of ErbB dimers that drive tumorigenesis by using genetically engineered T cells. Mol. Med. 2012, 18, 565–576. [Google Scholar] [CrossRef]
- Zhang, J.; Basher, F.; Wu, J.D. NKG2D ligands in tumor immunity: Two sides of a coin. Front. Immunol. 2015, 6, 97. [Google Scholar] [CrossRef] [Green Version]
- Spear, P.; Barber, A.; Rynda-Apple, A.; Sentman, C.L. NKG2D CAR T-cell therapy inhibits the growth of NKG2D ligand heterogeneous tumors. Immunol. Cell Biol. 2013, 91, 435–440. [Google Scholar] [CrossRef] [Green Version]
- Till, B.G.; Jensen, M.C.; Wang, J.; Chen, E.Y.; Wood, B.L.; Greisman, H.A.; Qian, X.; James, S.E.; Raubitschek, A.; Forman, S.J.; et al. Adoptive immunotherapy for indolent non-hodgkin lymphoma and mantle cell lymphoma using genetically modified autologous CD20-specific T cells. Blood 2008, 112, 2261–2271. [Google Scholar] [CrossRef] [Green Version]
- Ajina, A.; Maher, J. Prospects for combined use of oncolytic viruses and CAR T-cells. J. Immunother. Cancer 2017, 5, 90. [Google Scholar] [CrossRef] [Green Version]
- Kim, D.S.; Dastidar, H.; Zhang, C.; Zemp, F.J.; Lau, K.; Ernst, M.; Rakic, A.; Sikdar, S.; Rajwani, J.; Naumenko, V.; et al. Smac mimetics and oncolytic viruses synergize in driving anticancer T-cell responses through complementary mechanisms. Nat. Commun. 2017, 8, 344. [Google Scholar] [CrossRef]
- Scott, E.M.; Duffy, M.R.; Freedman, J.D.; Fisher, K.D.; Seymour, L.W. Solid Tumor Immunotherapy with T Cell Engager-Armed Oncolytic Viruses. Macromol. Biosci. 2018, 18, 1700187. [Google Scholar] [CrossRef]
- Mohammed, S.; Sukumaran, S.; Bajgain, P.; Watanabe, N.; Heslop, H.E.; Rooney, C.M.; Brenner, M.K.; Fisher, W.E.; Leen, A.M.; Vera, J.F. Improving Chimeric Antigen Receptor-Modified T Cell Function by Reversing the Immunosuppressive Tumor Microenvironment of Pancreatic Cancer. Mol. Ther. 2017, 25, 249–258. [Google Scholar] [CrossRef] [Green Version]
- Liu, X.; Ranganathan, R.; Jiang, S.; Fang, C.; Sun, J.; Kim, S.; Newick, K.; Lo, A.; June, C.H.; Zhao, Y.; et al. A chimeric switch-receptor targeting PD1 augments the efficacy of second-generation CAR T cells in advanced solid tumors. Cancer Res. 2016, 76, 1578–1590. [Google Scholar] [CrossRef] [Green Version]
- Alizadeh, D.; Wong, R.A.; Yang, X.; Wang, D.; Pecoraro, J.R.; Kuo, C.F.; Aguilar, B.; Qi, Y.; Ann, D.K.; Starr, R.; et al. IL15 enhances CAR-T cell antitumor activity by reducing mTORC1 activity and preserving their stem cell memory phenotype. Cancer Immunol. Res. 2019, 7, 759–772. [Google Scholar] [CrossRef]
- Cieri, N.; Camisa, B.; Cocchiarella, F.; Forcato, M.; Oliveira, G.; Provasi, E.; Bondanza, A.; Bordignon, C.; Peccatori, J.; Ciceri, F.; et al. IL-7 and IL-15 instruct the generation of human memory stem T cells from naive precursors. Blood 2013, 121, 573–584. [Google Scholar] [CrossRef]
- Chen, J.C.; Chang, Y.W.; Hong, C.C.; Yu, Y.H.; Su, J.L. The role of the VEGF-C/VEGFRs axis in tumor progression and therapy. Int. J. Mol. Sci. 2013, 14, 88–107. [Google Scholar] [CrossRef]
- Fukumura, D.; Xavier, R.; Sugiura, T.; Chen, Y.; Park, E.C.; Lu, N.; Selig, M.; Nielsen, G.; Taksir, T.; Jain, R.K.; et al. Tumor induction of VEGF promoter activity in stromal cells. Cell 1998, 94, 715–725. [Google Scholar] [CrossRef] [Green Version]
- Chinnasamy, D.; Yu, Z.; Theoret, M.R.; Zhao, Y.; Shrimali, R.K.; Morgan, R.A.; Feldman, S.A.; Restifo, N.P.; Rosenberg, S.A. Gene therapy using genetically modified lymphocytes targeting VEGFR-2 inhibits the growth of vascularized syngenic tumors in mice. J. Clin. Investig. 2010, 120, 3953–3968. [Google Scholar] [CrossRef] [Green Version]
- Xia, A.L.; Wang, X.C.; Lu, Y.J.; Lu, X.J.; Sun, B. Chimeric-antigen receptor T (CAR-T) cell therapy for solid tumors: Challenges and opportunities. Oncotarget 2017, 8, 90521–90531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Van Schalkwyk, M.C.I.; Papa, S.E.; Jeannon, J.P.; Urbano, T.G.; Spicer, J.F.; Maher, J. Design of a phase i clinical trial to evaluate intratumoral delivery of ErbB-targeted chimeric antigen receptor T-cells in locally advanced or recurrent head and neck cancer. Hum. Gene Ther. Clin. Dev. 2013, 24, 134–142. [Google Scholar] [CrossRef] [PubMed]
- Choi, B.D.; Suryadevara, C.M.; Gedeon, P.C.; Herndon Ii, J.E.; Sanchez-Perez, L.; Bigner, D.D.; Sampson, J.H. Intracerebral delivery of a third generation EGFRvIII-specific chimeric antigen receptor is efficacious against human glioma. J. Clin. Neurosci. 2014, 21, 189–190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sridhar, P.; Petrocca, F. Regional delivery of chimeric antigen receptor (CAR) T-cells for cancer therapy. Cancers 2017, 9, 92. [Google Scholar] [CrossRef] [Green Version]
- Smith, T.T.; Moffett, H.F.; Stephan, S.B.; Opel, C.F.; Dumigan, A.G.; Jiang, X.; Pillarisetty, V.G.; Pillai, S.P.S.; Wittrup, K.D.; Stephan, M.T. Biopolymers codelivering engineered T cells and STING agonists can eliminate heterogeneous tumors. J. Clin. Investig. 2017, 127, 2176–2191. [Google Scholar] [CrossRef]
- Vignali, D.; Kallikourdis, M. Improving homing in T cell therapy. Cytokine Growth Factor Rev. 2017, 36, 107–116. [Google Scholar] [CrossRef]
- McGettrick, H.M.; Butler, L.M.; Buckley, C.D.; Ed Rainger, G.; Nash, G.B. Tissue stroma as a regulator of leukocyte recruitment in inflammation. J. Leukoc. Biol. 2012, 91, 385–400. [Google Scholar] [CrossRef]
- Chow, M.T.; Luster, A.D. Chemokines in cancer. Cancer Immunol. Res. 2014, 2, 1125–1131. [Google Scholar] [CrossRef] [Green Version]
- Lacy, P. Editorial: Secretion of cytokines and chemokines by innate immune cells. Front. Immunol. 2015, 6, 190. [Google Scholar] [CrossRef] [Green Version]
- Moon, E.K.; Wang, L.C.S.; Bekdache, K.; Lynn, R.C.; Lo, A.; Thorne, S.H.; Albelda, S.M. Intra-tumoral delivery of CXCL11 via a vaccinia virus, but not by modified T cells, enhances the efficacy of adoptive T cell therapy and vaccines. OncoImmunology 2018, 7, e1395997. [Google Scholar] [CrossRef]
- Kershaw, M.H.; Wang, G.; Westwood, J.A.; Pachynski, R.K.; Tiffany, H.L.; Marincola, F.M.; Wang, E.; Young, H.A.; Murphy, P.M.; Hwu, P. Redirecting migration of T cells to chemokine secreted from tumors by genetic modification with CXCR2. Hum. Gene Ther. 2002, 13, 1971–1980. [Google Scholar] [CrossRef]
- Moon, E.K.; Carpenito, C.; Sun, J.; Wang, L.C.S.; Kapoor, V.; Predina, J.; Powell Jr, D.J.; Riley, J.L.; June, C.H.; Albelda, S.M. Expression of a functional CCR2 receptor enhances tumor localization and tumor eradication by retargeted human T cells expressing a mesothelin-specific chimeric antibody receptor. Clin. Cancer Res. 2011, 17, 4719–4730. [Google Scholar] [CrossRef] [Green Version]
- Craddock, J.A.; Lu, A.; Bear, A.; Pule, M.; Brenner, M.K.; Rooney, C.M.; Foster, A.E. Enhanced tumor trafficking of GD2 chimeric antigen receptor T cells by expression of the chemokine receptor CCR2b. J. Immunother. 2010, 33, 780–788. [Google Scholar] [CrossRef] [Green Version]
- Milner, J.J.; Toma, C.; Yu, B.; Zhang, K.; Omilusik, K.; Phan, A.T.; Wang, D.; Getzler, A.J.; Nguyen, T.; Crotty, S.; et al. Runx3 programs CD8(+) T cell residency in non-lymphoid tissues and tumours. Nature 2017, 552, 253–257. [Google Scholar] [CrossRef]
- Nishio, N.; Dotti, G. Oncolytic virus expressing RANTES and IL-15 enhances function of CAR-modified T cells in solid tumors. OncoImmunology 2015, 4, e988098. [Google Scholar] [CrossRef] [Green Version]
- Brown, C.E.; Alizadeh, D.; Starr, R.; Weng, L.; Wagner, J.R.; Naranjo, A.; Ostberg, J.R.; Blanchard, M.S.; Kilpatrick, J.; Simpson, J.; et al. Regression of Glioblastoma after Chimeric Antigen Receptor T-Cell Therapy. N. Engl. J. Med. 2016, 375, 2561–2569. [Google Scholar] [CrossRef]
- Sun, S.; Hao, H.; Yang, G.; Zhang, Y.; Fu, Y. Immunotherapy with CAR-modified T cells: Toxicities and overcoming strategies. J. Immunol. Res. 2018, 2018, 2386187. [Google Scholar] [CrossRef]
- Jackson, H.J.; Brentjens, R.J. Overcoming antigen escape with CAR T-cell therapy. Cancer Discov. 2015, 5, 1238–1240. [Google Scholar] [CrossRef] [Green Version]
- Han, X.; Wang, Y.; Wei, J.; Han, W. Multi-antigen-targeted chimeric antigen receptor T cells for cancer therapy. J. Hematol. Oncol. 2019, 12, 128. [Google Scholar] [CrossRef]
- Rizvi, N.A.; Hellmann, M.D.; Snyder, A.; Kvistborg, P.; Makarov, V.; Havel, J.J.; Lee, W.; Yuan, J.; Wong, P.; Ho, T.S.; et al. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015, 348, 124–128. [Google Scholar] [CrossRef] [Green Version]
- Robert, C.; Schachter, J.; Long, G.V.; Arance, A.; Grob, J.J.; Mortier, L.; Daud, A.; Carlino, M.S.; McNeil, C.; Lotem, M.; et al. Pembrolizumab versus ipilimumab in advanced melanoma. N. Engl. J. Med. 2015, 372, 2521–2532. [Google Scholar] [CrossRef] [PubMed]
- Choudhury, N.; Nakamura, Y. Importance of immunopharmacogenomics in cancer treatment: Patient selection and monitoring for immune checkpoint antibodies. Cancer Sci. 2016, 107, 107–115. [Google Scholar] [CrossRef] [PubMed]
- John, L.B.; Devaud, C.; Duong, C.P.M.; Yong, C.S.; Beavis, P.A.; Haynes, N.M.; Chow, M.T.; Smyth, M.J.; Kershaw, M.H.; Darcy, P.K. Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells. Clin. Cancer Res. 2013, 19, 5636–5646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cogdill, A.P.; Andrews, M.C.; Wargo, J.A. Hallmarks of response to immune checkpoint blockade. Br. J. Cancer 2017, 117, 1–7. [Google Scholar] [CrossRef] [Green Version]
- Li, S.; Siriwon, N.; Zhang, X.; Yang, S.; Jin, T.; He, F.; Kim, Y.J.; Mac, J.; Lu, Z.; Wang, S.; et al. Enhanced cancer immunotherapy by chimeric antigen receptor–modified T cells engineered to secrete checkpoint inhibitors. Clin. Cancer Res. 2017, 23, 6982–6992. [Google Scholar] [CrossRef] [Green Version]
- Serganova, I.; Moroz, E.; Cohen, I.; Moroz, M.; Mane, M.; Zurita, J.; Shenker, L.; Ponomarev, V.; Blasberg, R. Enhancement of PSMA-Directed CAR Adoptive Immunotherapy by PD-1/PD-L1 Blockade. Mol. Ther. Oncolytics 2017, 4, 41–54. [Google Scholar] [CrossRef]
- Gargett, T.; Yu, W.; Dotti, G.; Yvon, E.S.; Christo, S.N.; Hayball, J.D.; Lewis, I.D.; Brenner, M.K.; Brown, M.P. GD2-specific CAR T Cells Undergo Potent Activation and Deletion Following Antigen Encounter but can be Protected from Activation-induced Cell Death by PD-1 Blockade. Mol. Ther. 2016, 24, 1135–1149. [Google Scholar] [CrossRef] [Green Version]
- Thistlethwaite, F.C.; Gilham, D.E.; Guest, R.D.; Rothwell, D.G.; Pillai, M.; Burt, D.J.; Byatte, A.J.; Kirillova, N.; Valle, J.W.; Sharma, S.K.; et al. The clinical efficacy of first-generation carcinoembryonic antigen (CEACAM5)-specific CAR T cells is limited by poor persistence and transient pre-conditioning-dependent respiratory toxicity. Cancer Immunol. Immunother. 2017, 66, 1425–1436. [Google Scholar] [CrossRef] [Green Version]
- Xu, J.; Wang, Y.; Shi, J.; Liu, J.; Li, Q.; Chen, L. Combination therapy: A feasibility strategy for CAR-T cell therapy in the treatment of solid tumors. Oncol. Lett. 2018, 16, 2063–2070. [Google Scholar] [CrossRef] [Green Version]
- Anampa, J.; Chen, A.; Wright, J.; Patel, M.; Pellegrino, C.; Fehn, K.; Sparano, J.A.; Andreopoulou, E. Phase I Trial of Veliparib, a Poly ADP Ribose Polymerase Inhibitor, Plus Metronomic Cyclophosphamide in Metastatic HER2-negative Breast Cancer. Clin. Breast Cancer 2018, 18, e135–e142. [Google Scholar] [CrossRef]
- Vierboom, M.P.M.; Bos, G.M.J.; Ooms, M.; Offringa, R.; Melief, C.J.M. Cyclophosphamide enhances anti-tumor effect of wild-type p53-specific CTL. Int. J. Cancer 2000, 87, 253–260. [Google Scholar] [CrossRef]
- Bracci, L.; Schiavoni, G.; Sistigu, A.; Belardelli, F. Immune-based mechanisms of cytotoxic chemotherapy: Implications for the design of novel and rationale-based combined treatments against cancer. Cell Death Differ. 2014, 21, 15–25. [Google Scholar] [CrossRef] [Green Version]
- Michaud, M.; Martins, I.; Sukkurwala, A.Q.; Adjemian, S.; Ma, Y.; Pellegatti, P.; Shen, S.; Kepp, O.; Scoazec, M.; Mignot, G.; et al. Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science 2011, 334, 1573–1577. [Google Scholar] [CrossRef]
- Alizadeh, D.; Trad, M.; Hanke, N.T.; Larmonier, C.B.; Janikashvili, N.; Bonnotte, B.; Katsanis, E.; Larmonier, N. Doxorubicin eliminates myeloid-derived suppressor cells and enhances the efficacy of adoptive T-cell transfer in breast cancer. Cancer Res. 2014, 74, 104–118. [Google Scholar] [CrossRef] [Green Version]
- Sistigu, A.; Yamazaki, T.; Vacchelli, E.; Chaba, K.; Enot, D.P.; Adam, J.; Vitale, I.; Goubar, A.; Baracco, E.E.; Remédios, C.; et al. Cancer cell–autonomous contribution of type I interferon signaling to the efficacy of chemotherapy. Nat. Med. 2014, 20, 1301–1309. [Google Scholar] [CrossRef]
- Motyka, B.; Korbutt, G.; Pinkoski, M.J.; Heibein, J.A.; Caputo, A.; Hobman, M.; Barry, M.; Shostak, I.; Sawchuk, T.; Holmes, C.F.B.; et al. Mannose 6-phosphate/insulin-like growth factor II receptor is a death receptor for granzyme B during cytotoxic T cell-induced apoptosis. Cell 2000, 103, 491–500. [Google Scholar] [CrossRef] [Green Version]
- Trapani, J.A.; Sutton, V.R.; Thia, K.Y.T.; Li, Y.Q.; Froelich, C.J.; Jans, D.A.; Sandrin, M.S.; Browne, K.A. A clathrin/dynamin- and mannose-6-phosphate receptor-independent pathway for granzyme B-induced cell death. J. Cell Biol. 2003, 160, 223–233. [Google Scholar] [CrossRef] [Green Version]
- Ramakrishnan, R.; Huang, C.; Cho, H.I.; Lloyd, M.; Johnson, J.; Ren, X.; Altiok, S.; Sullivan, D.; Weber, J.; Celis, E.; et al. Autophagy induced by conventional chemotherapy mediates tumor cell sensitivity to immunotherapy. Cancer Res. 2012, 72, 5483–5493. [Google Scholar] [CrossRef] [Green Version]
- Parente-Pereira, A.C.; Whilding, L.M.; Brewig, N.; Van Der Stegen, S.J.C.; Davies, D.M.; Wilkie, S.; Van Schalkwyk, M.C.I.; Ghaem-Maghami, S.; Maher, J. Synergistic chemoimmunotherapy of epithelial ovarian cancer using ErbB-retargeted T cells combined with carboplatin. J. Immunol. 2013, 191, 2437–2445. [Google Scholar] [CrossRef] [Green Version]
- Whilding, L.M.; Maher, J. ErbB-targeted CAR T-cell immunotherapy of cancer. Immunotherapy 2015, 7, 229–241. [Google Scholar] [CrossRef]
- Proietti, E.; Moschella, F.; Capone, I.; Belardelli, F. Exploitation of the propulsive force of chemotherapy for improving the response to cancer immunotherapy. Mol. Oncol. 2012, 6, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Senovilla, L.; Vitale, I.; Martins, I.; Tailler, M.; Pailleret, C.; Michaud, M.; Galluzzi, L.; Adjemian, S.; Kepp, O.; Niso-Santano, M.; et al. An immunosurveillance mechanism controls cancer cell ploidy. Science 2012, 337, 1678–1684. [Google Scholar] [CrossRef] [PubMed]
- Martins, I.; Tesniere, A.; Kepp, O.; Michaud, M.; Schlemmer, F.; Senovilla, L.; Séror, C.; Métivier, D.; Perfettini, J.L.; Zitvogel, L.; et al. Chemotherapy induces ATP release from tumor cells. Cell Cycle 2009, 8, 3723–3728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Garg, A.D.; Galluzzi, L.; Apetoh, L.; Baert, T.; Birge, R.B.; Bravo-San Pedro, J.M.; Breckpot, K.; Brough, D.; Chaurio, R.; Cirone, M.; et al. Molecular and translational classifications of DAMPs in immunogenic cell death. Front. Immunol. 2015, 6, 588. [Google Scholar] [CrossRef] [Green Version]
- Lutsiak, M.E.C.; Semnani, R.T.; De Pascalis, R.; Kashmiri, S.V.S.; Schlom, J.; Sabzevari, H. Inhibition of CD4+25+ T regulatory cell function implicated in enhanced immune response by low-dose cyclophosphamide. Blood 2005, 105, 2862–2868. [Google Scholar] [CrossRef]
- Heylmann, D.; Bauer, M.; Becker, H.; Van Gool, S.; Bacher, N.; Steinbrink, K.; Kaina, B. Human CD4+CD25+ regulatory T cells are sensitive to low dose cyclophosphamide: Implications for the immune response. PLoS ONE 2013, 8, e83384. [Google Scholar] [CrossRef] [Green Version]
- Kwa, M.; Li, X.; Novik, Y.; Oratz, R.; Jhaveri, K.; Wu, J.; Gu, P.; Meyers, M.; Muggia, F.; Speyer, J.; et al. Serial immunological parameters in a phase II trial of exemestane and low-dose oral cyclophosphamide in advanced hormone receptor-positive breast cancer. Breast Cancer Res. Treat. 2018, 168, 57–67. [Google Scholar] [CrossRef]
- Morgan, R.A.; Yang, J.C.; Kitano, M.; Dudley, M.E.; Laurencot, C.M.; Rosenberg, S.A. Case report of a serious adverse event following the administration of t cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol. Ther. 2010, 18, 843–851. [Google Scholar] [CrossRef]
- Rosenberg, S.A.; Dudley, M.E. Adoptive cell therapy for the treatment of patients with metastatic melanoma. Curr. Opin. Immunol. 2009, 21, 233–240. [Google Scholar] [CrossRef] [Green Version]
- Qiao, J.; Liu, Z.; Fu, Y.X. Adapting conventional cancer treatment for immunotherapy. J. Mol. Med. 2016, 94, 489–495. [Google Scholar] [CrossRef]
- Wang, W.; Kryczek, I.; Dostál, L.; Lin, H.; Tan, L.; Zhao, L.; Lu, F.; Wei, S.; Maj, T.; Peng, D.; et al. Effector T Cells Abrogate Stroma-Mediated Chemoresistance in Ovarian Cancer. Cell 2016, 165, 1092–1105. [Google Scholar] [CrossRef] [Green Version]
- Higgins, J.P.; Bernstein, M.B.; Hodge, J.W. Enhancing immune responses to tumor-associated antigens. Cancer Biol. Ther. 2009, 8, 1440–1449. [Google Scholar] [CrossRef] [Green Version]
- Sharma, A.; Bode, B.; Wenger, R.H.; Lehmann, K.; Sartori, A.A.; Moch, H.; Knuth, A.; von Boehmer, L.; van den Broek, M. Γ-Radiation promotes immunological recognition of cancer cells through increased expression of cancer-testis antigens in vitro and in vivo. PLoS ONE 2011, 6, e28217. [Google Scholar] [CrossRef]
- Lee, Y.; Auh, S.L.; Wang, Y.; Burnette, B.; Wang, Y.; Meng, Y.; Beckett, M.; Sharma, R.; Chin, R.; Tu, T.; et al. Therapeutic effects of ablative radiation on local tumor require CD8 + T cells: Changing strategies for cancer treatment. Blood 2009, 114, 589–595. [Google Scholar] [CrossRef]
- Aymeric, L.; Apetoh, L.; Ghiringhelli, F.; Tesniere, A.; Martins, I.; Kroemer, G.; Smyth, M.J.; Zitvogel, L. Tumor cell death and ATP release prime dendritic cells and efficient anticancer immunity. Cancer Res. 2010, 70, 855–858. [Google Scholar] [CrossRef] [Green Version]
- Zhang, B.; Bowerman, N.A.; Salama, J.K.; Schmidt, H.; Spiotto, M.T.; Schietinger, A.; Yu, P.; Fu, Y.X.; Weichselbaum, R.R.; Rowley, D.A.; et al. Induced sensitization of tumor stroma leads to eradication of established cancer by T cells. J. Exp. Med. 2007, 204, 49–55. [Google Scholar] [CrossRef]
- Matsumura, S.; Wang, B.; Kawashima, N.; Braunstein, S.; Badura, M.; Cameron, T.O.; Babb, J.S.; Schneider, R.J.; Formenti, S.C.; Dustin, M.L.; et al. Radiation-induced CXCL16 release by breast cancer cells attracts effector T cells. J. Immunol. 2008, 181, 3099–3107. [Google Scholar] [CrossRef] [Green Version]
- Ganss, R.; Ryschich, E.; Klar, E.; Arnold, B.; Hämmerling, G.J. Combination of T-cell therapy and trigger of inflammation induces remodeling of the vasculature and tumor eradication. Cancer Res. 2002, 62, 1462–1470. [Google Scholar]
- Liao, Y.P.; Wang, C.C.; Butterfield, L.H.; Economou, J.S.; Ribas, A.; Meng, W.S.; Iwamoto, K.S.; McBride, W.H. Ionizing radiation affects human MART-1 melanoma antigen processing and presentation by dendritic cells. J. Immunol. 2004, 173, 2462–2469. [Google Scholar] [CrossRef] [Green Version]
- Apetoh, L.; Ghiringhelli, F.; Tesniere, A.; Obeid, M.; Ortiz, C.; Criollo, A.; Mignot, G.; Maiuri, M.C.; Ullrich, E.; Saulnier, P.; et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat. Med. 2007, 13, 1050–1059. [Google Scholar] [CrossRef]
- Burnette, B.C.; Liang, H.; Lee, Y.; Chlewicki, L.; Khodarev, N.N.; Weichselbaum, R.R.; Fu, Y.X.; Auh, S.L. The efficacy of radiotherapy relies upon induction of type I interferon-dependent innate and adaptive immunity. Cancer Res. 2011, 71, 2488–2496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Crouse, J.; Kalinke, U.; Oxenius, A. Regulation of antiviral T cell responses by type i interferons. Nat. Rev. Immunol. 2015, 15, 231–242. [Google Scholar] [CrossRef] [PubMed]
- Diamond, M.S.; Kinder, M.; Matsushita, H.; Mashayekhi, M.; Dunn, G.P.; Archambault, J.M.; Lee, H.; Arthur, C.D.; White, J.M.; Kalinke, U.; et al. Type I interferon is selectively required by dendritic cells for immune rejection of tumors. J. Exp. Med. 2011, 208, 1989–2003. [Google Scholar] [CrossRef] [PubMed]
- Fuertes, M.B.; Kacha, A.K.; Kline, J.; Woo, S.R.; Kranz, D.M.; Murphy, K.M.; Gajewski, T.F. Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8α+ dendritic cells. J. Exp. Med. 2011, 208, 2005–2016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Deng, L.; Liang, H.; Xu, M.; Yang, X.; Burnette, B.; Arina, A.; Li, X.D.; Mauceri, H.; Beckett, M.; Darga, T.; et al. STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors. Immunity 2014, 41, 843–852. [Google Scholar] [CrossRef] [Green Version]
- Lugade, A.A.; Sorensen, E.W.; Gerber, S.A.; Moran, J.P.; Frelinger, J.G.; Lord, E.M. Radiation-induced IFN-γ production within the tumor microenvironment influences antitumor immunity. J. Immunol. 2008, 180, 3132–3139. [Google Scholar] [CrossRef] [Green Version]
- Kang, J.; Demaria, S.; Formenti, S. Current clinical trials testing the combination of immunotherapy with radiotherapy. J. Immunother. Cancer 2016, 4, 51. [Google Scholar] [CrossRef] [Green Version]
- Rodriguez-Ruiz, M.E.; Rodriguez, I.; Garasa, S.; Barbes, B.; Solorzano, J.L.; Perez-Gracia, J.L.; Labiano, S.; Sanmamed, M.F.; Azpilikueta, A.; Bolaños, E.; et al. Abscopal effects of radiotherapy are enhanced by combined immunostimulatory mAbs and are dependent on CD8 T cells and crosspriming. Cancer Res. 2016, 76, 5994–6005. [Google Scholar] [CrossRef] [Green Version]
- Demaria, S.; Ng, B.; Devitt, M.L.; Babb, J.S.; Kawashima, N.; Liebes, L.; Formenti, S.C. Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Int. J. Radiat. Oncol. Biol. Phys. 2004, 58, 862–870. [Google Scholar] [CrossRef]
- Weiss, T.; Weller, M.; Guckenberger, M.; Sentman, C.L.; Roth, P. NKG2D-based CAR T cells and radiotherapy exert synergistic efficacy in glioblastoma. Cancer Res. 2018, 78, 1031–1043. [Google Scholar] [CrossRef] [Green Version]
- Gaipl, U.S.; Multhoff, G.; Scheithauer, H.; Lauber, K.; Hehlgans, S.; Frey, B.; Rödel, F. Kill and spread the word: Stimulation of antitumor immune responses in the context of radiotherapy. Immunotherapy 2014, 6, 597–610. [Google Scholar] [CrossRef]
- Yovino, S.; Grossman, S.A. Severity, etiology and possible consequences of treatment-related lymphopenia in patients with newly diagnosed high-grade gliomas. CNS Oncol. 2012, 1, 149–154. [Google Scholar] [CrossRef]
- Baniyash, M. TCR ζ-chain downregulation: Curtailing an excessive inflammatory immune response. Nat. Rev. Immunol. 2004, 4, 675–687. [Google Scholar] [CrossRef]
- Alanio, C.; Lemaitre, F.; Law, H.K.W.; Hasan, M.; Albert, M.L. Enumeration of human antigen-specific naive CD8+ T cells reveals conserved precursor frequencies. Blood 2010, 115, 3718–3725. [Google Scholar] [CrossRef] [Green Version]
- Yoo, E.J.; Park, J.C.; Kim, E.H.; Park, C.H.; Shim, C.N.; Lee, H.J.; Chung, H.S.; Lee, H.; Shin, S.K.; Lee, S.K.; et al. Prognostic value of neutrophil-to-lymphocyte ratio in patients treated with concurrent chemoradiotherapy for locally advanced oesophageal cancer. Dig. Liver Dis. 2014, 46, 846–853. [Google Scholar] [CrossRef]
- Gazdic, M.; Simovic Markovic, B.; Jovicic, N.; Misirkic-Marjanovic, M.; Djonov, V.; Jakovljevic, V.; Arsenijevic, N.; Lukic, M.L.; Volarevic, V. Mesenchymal Stem Cells Promote Metastasis of Lung Cancer Cells by Downregulating Systemic Antitumor Immune Response. Stem Cells Int. 2017, 2017, 6294717. [Google Scholar] [CrossRef] [Green Version]
- Galon, J.; Costes, A.; Sanchez-Cabo, F.; Kirilovsky, A.; Mlecnik, B.; Lagorce-Pagès, C.; Tosolini, M.; Camus, M.; Berger, A.; Wind, P.; et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 2006, 313, 1960–1964. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Q.; Yang, X.J.; Kundu, S.D.; Pins, M.; Javonovic, B.; Meyer, R.; Kim, S.J.; Greenberg, N.M.; Kuzel, T.; Meagher, R.; et al. Blockade of transforming growth factor-β signaling in tumor-reactive CD8+ T cells activates the antitumor immune response cycle. Mol. Cancer Ther. 2006, 5, 1733–1743. [Google Scholar] [CrossRef] [Green Version]
- Nelson, A.; Nair, S.; Nagaraj, S. CD4+ T cells suppress immune response to cancer: Novel targets for antitumor efforts. Expert Rev. Clin. Immunol. 2012, 8, 401–403. [Google Scholar] [CrossRef]
- Kitayama, J.; Yasuda, K.; Kawai, K.; Sunami, E.; Nagawa, H. Circulating lymphocyte number has a positive association with tumor response in neoadjuvant chemoradiotherapy for advanced rectal cancer. Radiat. Oncol. 2010, 5, 47. [Google Scholar] [CrossRef] [Green Version]
- Schueneman, A.J.; Sugar, E.A.; Uram, J.; Bigelow, E.; Herman, J.M.; Edil, B.H.; Jaffee, E.M.; Zheng, L.; Laheru, D.A. Low total lymphocyte count is associated with poor survival in patients with resected pancreatic adenocarcinoma receiving a gm-csf secreting pancreatic tumor vaccine. Ann. Surg. Oncol. 2013, 20, S725–S730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tang, C.; Liao, Z.; Gomez, D.; Levy, L.; Zhuang, Y.; Gebremichael, R.A.; Hong, D.S.; Komaki, R.; Welsh, J.W. Lymphopenia association with gross tumor volume and lung V5 and its effects on non-small cell lung cancer patient outcomes. Int. J. Radiat. Oncol. Biol. Phys. 2014, 89, 1084–1091. [Google Scholar] [CrossRef] [PubMed]
- Buka, D.; Dvořák, J.; Sitorová, V.; Hátlová, J.; Richter, I.; Sirák, I. Changes in the CD8+ density of tumor infiltrating lymphocytes after neoadjuvant radiochemotherapy in patients with rectal adenocarcinoma. Klin. Onkol. 2016, 29, 204–209. [Google Scholar] [CrossRef] [PubMed]
- Zitvogel, L.; Kepp, O.; Kroemer, G. Immune parameters affecting the efficacy of chemotherapeutic regimens. Nat. Rev. Clin. Oncol. 2011, 8, 151–160. [Google Scholar] [CrossRef]
- Aranda, F.; Buqué, A.; Bloy, N.; Castoldi, F.; Eggermont, A.; Cremer, I.; Fridman, W.H.; Fucikova, J.; Galon, J.; Spisek, R.; et al. Trial Watch: Adoptive cell transfer for oncological indications. OncoImmunology 2015, 4, e1046673. [Google Scholar] [CrossRef] [Green Version]
- Sathyanarayanan, V.; Neelapu, S.S. Cancer immunotherapy: Strategies for personalization and combinatorial approaches. Mol. Oncol. 2015, 9, 2043–2053. [Google Scholar] [CrossRef] [Green Version]
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
Zhang, X.-W.; Wu, Y.-S.; Xu, T.-M.; Cui, M.-H. CAR-T Cells in the Treatment of Ovarian Cancer: A Promising Cell Therapy. Biomolecules 2023, 13, 465. https://doi.org/10.3390/biom13030465
Zhang X-W, Wu Y-S, Xu T-M, Cui M-H. CAR-T Cells in the Treatment of Ovarian Cancer: A Promising Cell Therapy. Biomolecules. 2023; 13(3):465. https://doi.org/10.3390/biom13030465
Chicago/Turabian StyleZhang, Xi-Wen, Yi-Shi Wu, Tian-Min Xu, and Man-Hua Cui. 2023. "CAR-T Cells in the Treatment of Ovarian Cancer: A Promising Cell Therapy" Biomolecules 13, no. 3: 465. https://doi.org/10.3390/biom13030465
APA StyleZhang, X. -W., Wu, Y. -S., Xu, T. -M., & Cui, M. -H. (2023). CAR-T Cells in the Treatment of Ovarian Cancer: A Promising Cell Therapy. Biomolecules, 13(3), 465. https://doi.org/10.3390/biom13030465