Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 FGFR4 targeting by single domain antibodies is feasable in vitro. Now it has to be shown that it is feasable in vivo, and that a therapeutic effect can be achieved. + 3266 word(s) 3266 2020-11-16 09:25:54 |
2 format correct -1724 word(s) 1542 2020-11-24 10:30:25 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Alijaj, N.; Moutel, S.; Gouveia, Z.L.; Gray, M.; Roveri, M.; Dzhumashev, D.; Weber, F.; Meier, G.; Luciani, P.; Rössler, J.K.; et al. Novel FGFR4-Targeting Single-Domain Antibodies. Encyclopedia. Available online: https://encyclopedia.pub/entry/3190 (accessed on 27 April 2024).
Alijaj N, Moutel S, Gouveia ZL, Gray M, Roveri M, Dzhumashev D, et al. Novel FGFR4-Targeting Single-Domain Antibodies. Encyclopedia. Available at: https://encyclopedia.pub/entry/3190. Accessed April 27, 2024.
Alijaj, Nagjie, Sandrine Moutel, Zelia L. Gouveia, Maxim Gray, Maurizio Roveri, Dzhangar Dzhumashev, Florian Weber, Gianmarco Meier, Paola Luciani, Jochen K. Rössler, et al. "Novel FGFR4-Targeting Single-Domain Antibodies" Encyclopedia, https://encyclopedia.pub/entry/3190 (accessed April 27, 2024).
Alijaj, N., Moutel, S., Gouveia, Z.L., Gray, M., Roveri, M., Dzhumashev, D., Weber, F., Meier, G., Luciani, P., Rössler, J.K., Schäfer, B.W., Perez, F., & Bernasconi, M. (2020, November 24). Novel FGFR4-Targeting Single-Domain Antibodies. In Encyclopedia. https://encyclopedia.pub/entry/3190
Alijaj, Nagjie, et al. "Novel FGFR4-Targeting Single-Domain Antibodies." Encyclopedia. Web. 24 November, 2020.
Novel FGFR4-Targeting Single-Domain Antibodies
Edit

FGFR4, member of the Fibroblast Growth Factor Receptor family, is a promising therapeutic target for rhabdomyosarcoma, the most frequent pediatric soft tissue sarcoma. FGFR4 single domain Antibodies specific for FGFR4 were selected, characterized, and showed promising results against rhabdomyosarcoma cell lines as single agents, as targeting moieties for liposomes, and as antigen binding domain in Chimeric Antigen Receptor modified T cells.

FGFR4 Rhabdomyosarcoma targeted drug delivery nanobodies single domain antibodies CAR T cells chimeric antigen receptor

1. Introduction

1.1. Rhabdomyosarcoma

Approximately 50% of children and adolescents diagnosed with soft tissue sarcoma suffer from rhabdomyosarcoma (RMS), a striated-muscle lineage malignancy with variable pathologies[1]. The two major subtypes of the tumor are embryonal RMS (ERMS) and alveolar RMS (ARMS), accounting for 60% and 20% of all cases, respectively. ARMS is more aggressive and is characterized by a chromosomal translocation resulting in a PAX3-FOXO1 gene fusion, whereas ERMS is associated with different tumor-promoting mutations and chromosome number aberrations[2]. Surgery, radiation, and multi-drug chemotherapy composed of vincristine (VCR), actinomycin D and cyclophosphamide are the standard treatments for RMS [1]. The overall survival rates for RMS patients have improved within the last few decades but the prognostic outcome is still very poor for high-risk patients, including those presenting metastatic diseases, ARMS subtype, or diagnosis in adulthood[3][4]. The treatment goes along with high toxicity and many who survive RMS will experience long-term adverse effects as adults[5]. Therefore, there is an urgent need for new targeted therapies to improve overall survival rates, and to overcome long-term side effects.

1.2. Liposomal Drug Delivery - Passive

Nanovesicle-mediated chemotherapeutic drug delivery offers the possibility to increase the therapeutic effect in the tumor and to decrease side effects in healthy tissues[6][7][8][9]. Passive accumulation of nanoparticles in the tumors has been attributed to the so-called “enhanced permeability and retention” (EPR) effect[10]. Fast-growing solid tumors display a leaky vascular architecture and a lack of functional lymphatics, enabling the size-dependent passive extravasation and accumulation of nanoparticles in the interstitial space of the tumor[11]. Most recently, this dogma has been challenged by findings showing that the great majority of nanoparticles enter tumors using an active process through endothelial cells[12]. Liposomal formulations of chemotherapeutic drugs have demonstrated the safety and improved pharmacokinetic properties of the drug[13]. Prominent examples are liposomal doxorubicin (Doxil), daunorubicin (DaunoXome), and VCR (Marqibo) which have contributed to reducing side-effects compared to the free drug[14][15][16]. However, liposomal formulations have not, so far, been able to increase the therapeutic effect of the encapsulated drug.

1.3. Liposomal Drug Delivery - Active

One possibility to increase the therapeutic effect of the encapsulated drug is to modify the liposomal surface with tumor targeting-ligands, such as peptides[17], antibodies or antibody fragments[18], for active targeting to cancer cells. Single-domain antibodies (sdAb), first discovered in camelids[19], are the smallest possible antibody fragments (15 kDa) derived from heavy-chain antibodies. They are characterized by affinities comparable to conventional bivalent antibodies, as well as by high solubility, tissue penetration, and stability[20]. Previously, we developed the optimal formulation of liposomal VCR[21], and we investigated its pharmacokinetic and biodistribution in a mouse model engrafted with human RMS cells, revealing longer plasma circulation time and enhanced tumor accumulation of the liposomal drug compared to free VCR. Now, to further improve tumor accumulation of the liposomes in RMS by active targeting, we selected and investigated novel RMS-targeting sdAb.

1.4. Fibroblast Growth Factor Receptor 4 (FGFR4)

FGFR4 belongs to the family of receptor tyrosine kinases and is involved in myogenesis and muscle regeneration by promoting cell survival and differentiation[22][23]. FGFR4 is absent in normal differentiated muscles and is specifically overexpressed in RMS[24], as well as in other tumors, such as hepatocellular carcinomas, head and neck squamous cell carcinomas and basal-like breast cancer[25][26][27][28]. Therefore, FGFR4 represents a promising candidate for targeted therapies in RMS.

1.5. Chimeric Antigen Receptor (CAR) T cell

Another approach that can benefit from specific tumor targeting and that may improve the therapeutic outcome for RMS patients, is represented by chimeric antigen receptor (CAR) T cells. These cytolytic T cells are engineered with an extracellular antigen-binding domain recognizing specifically surface antigens on tumor cells. The intracellular part of the receptors is composed of T cell receptor signaling and costimulatory domains[29]. Tremendous clinical success has been achieved in the treatment of hematological malignancies with CAR T cells targeting CD19[30][31], CD22[32] and the B cell maturation antigen (BCMA)[33]. The application of CAR T cells for solid tumors has been more challenging, due to the lack of ideal tumor-specific target molecules, and also due to the strong immunosuppressive tumor microenvironment (TME) of solid tumors. Nevertheless, preclinical studies of CD276 (B7-H3) CAR T cells in pediatric solid tumors demonstrated good activity[34], and encouraging results have been reported for RMS CAR T cells targeting HER2 led to remission in a child with refractory metastatic RMS[35].

2. Strategies for RMS Therapy by Targeting FGFR4 with sdAb

Researchers selected four FGFR4-binding sdAb and tested them in vitro for (a) inhibitory activity of FGFR4 signaling; (b) active delivery as liposome conjugates, and (c) cell-mediated immunotherapy as CAR constructs. Surface plasmon resonance spectroscopy of sdAb binding to FGFR4 revealed strong affinities in the order of nano- to picomolar. 

2.1. sdAb Inhibitory Activity on FGFR4 Signaling

The four selected sdAb A8, B1, B5 and F8 not only bind to FGFR4 expressed on RMS cells but are also able to block the FGF19-FGFR4-MAPK signaling axis. In ARMS, FGFR4 is a direct target gene of the fusion protein PAX3-FOXO1 [41], and in ERMS FGFR4 is frequently amplified with 12% of the tumors harboring activating mutations of the receptor[42][43][44]. In RMS, besides overexpression, FGFR4 has been shown to harbor activating mutations in over 6% of all tumors, resulting in constitutive tumor-promoting signaling within the cells[2][45][46]. Eventhough we did not observe a toxic effect on cultured RMS cells, it is tempting to speculate that FGFR4 signaling could still represent a therapeutic target for sdAb in RMS. Moreover, FGFR4 is not only implicated in RMS tumorigenesis, but drives tumor progression in FGF19 expressing hepatocellular carcinomas, head and neck squamous cell carcinomas, and basal-like breast cancer[25][26][27][28]. It is also estimated that 0.5% of all tumors display abnormalities in FGFR4[47]. The selected sdAb could therefore also serve as possible therapeutic approach for cancers other than RMS.

2.2. Active Delivery of Liposomes-sdAb Conjugates

Both free and liposome-conjugated sdAb bound specifically to Rh4-FR4wt cells and showed, except for uncoupled B5 sdAb, no binding to Rh4-FR4ko cells. Nevertheless, recombinant B5 binding to Rh4-FR4ko cells in FACS experiments was only 0.25 times higher than mCh control sdAb, whereas on Rh4-FR4wt it was 2 times higher. Affinity measurements revealed binding of only A8 and B5 another FGFR-member, FGFR2. The binding affinity of A8 to FGFR2 was in the micromolar range and therefore very low when compared to the binding affinity to FGFR4. The fast koff rates further highlighted its weak binding to FGFR2. In contrast, B5 showed high affinities to FGFR2 in the low nanomolar range, thus similar to its affinity for FGFR4. Rh4 cells do express FGFR2, but protein levels are lower compared to FGFR4. Moreover, the Rh4-FR4ko cells have reduced FGFR1 and FGFR2 protein levels compared to Rh4-FR4wt. The reasons for the lower expression level of FGFR1 and FGFR2 in Rh4-FR4ko are not completely clear but could be due to a clonal effect or to regulatory loops. Therefore, it is well possible that the binding of sdAb A8 and B5 to cell surface FGFR2 would be only detectable above a certain expression level.

The formulation of liposomal VCR was modified from the previously established one[21]by the introduction of DSPE-PEG-maleimide at 1 mol%. As expected, the resulting physico-chemical properties of the liposomes and the drug loading efficiency were comparable. SdAb coupling to the surface was performed as described by Oliveira and colleagues[48]with 0.4 nmol sdAb per μmol of total lipids and it resulted in high coupling efficiencies. Among various conditions of the coupling reaction tested, we also tested higher sdAb-to-lipid ratios, but this resulted in precipitation of the liposomes. The fraction of uncoupled sdAb in the liposome suspension was negligible and it did not apparently interfere with binding on cells.

Confocal microscopy of Rh4-FR4wt cells incubated with the fluorescent FGFR4-targeting liposomes showed a very specific internalization after 2 h of incubation, represented by dot-like structures within the cells, which were absent in Rh4-FR4ko cells. This indicates a rather fast internalization process which can represent an advantage for a drug delivery platform to highly vascularized tumors.

2.3. Cell-Mediated Immunotherapy as CAR Constructs

Importantly, researchers were able to verify the selective cell-mediated cytotoxicity of sdAb-based FGFR4 CAR T cells towards Rh4-FR4wt. Although some differences in cytotoxic efficiencies between three CD8+ T cell donors were observed, all FGFR4-CAR Ts showed the same specific trend. Real-time cell analysis revealed no or lower effects of FGFR CAR Ts on Rh4-FR4ko, comparable to that of control CD19 CAR Ts.

The immune-based treatment of RMS with FGFR4 CAR Ts holds promising potential, since RMS tumors display aberrantly high FGFR4 expression compared to healthy tissues[42]. It has been shown that high antigen densities above a certain threshold level are required for effective CAR T cell activation, offering a therapeutic window for RMS treatment[49][50]. Further studies will be required to test FGFR4 CAR Ts efficiency in a RMS in vivo model.

References

  1. Skapek, S.X.; Ferrari, A.; Gupta, A.A.; Lupo, P.J.; Butler, E.; Shipley, J.; Barr, F.G.; Hawkins, D.S. Rhabdomyosarcoma. Nat. Rev. Dis. Prim. 2019, 5, 1–19.
  2. Hern, J.F.; Chen, L.; Chmielecki, J.; Wei, J.S.; Patidar, R.; Rosenberg, M.; Ambrogio, L.; Auclair, D.; Wang, J.; Song, Y.K.; et al. Comprehensive Genomic Analysis of Rhabdomyosarcoma Reveals a Landscape of Alterations Affecting a Common Genetic Axis in Fusion-Positive and Fusion-Negative Tumors. Cancer Discov. 2014, 4, 216–231.
  3. Van Gaal, J.C.; Van Der Graaf, W.T.A.; Rikhof, B.; Van Hoesel, Q.G.C.M.; Teerenstra, S.; Suurmeijer, A.J.H.; Flucke, U.E.; Loeffen, J.L.C.M.; Sleijfer, S.; De Bont, E.S.J.M. The impact of age on outcome of embryonal and alveolar rhabdomyosarcoma patients. A multicenter study. Anticancer Res. 2012, 32, 4485–4497.
  4. Sultan, I.; Qaddoumi, I.; Yaser, S.; Rodriguez-Galindo, C.; Ferrari, A. Comparing Adult and Pediatric Rhabdomyosarcoma in the Surveillance, Epidemiology and End Results Program, 1973 to 2005: An Analysis of 2600 Patients. J. Clin. Oncol. 2009, 27, 3391–3397.
  5. Punyko, J.A.; Mertens, A.C.; Gurney, J.G.; Yasui, Y.; Donaldson, S.S.; Rodeberg, D.A.; Raney, R.B.; Stovall, M.; Sklar, C.A.; Robison, L.L.; et al. Long-term medical effects of childhood and adolescent rhabdomyosarcoma: A report from the childhood cancer survivor study. Pediatr. Blood Cancer 2005, 44, 643–653.
  6. Rodríguez-Nogales, C.; González-Fernández, Y.; Aldaz, A.; Couvreur, P.; Blanco-Prieto, M.J. Nanomedicines for Pediatric Cancers. ACS Nano 2018, 12, 7482–7496.
  7. Li, Z.; Tan, S.; Li, S.; Shen, Q.; Wang, K. Cancer drug delivery in the nano era: An overview and perspectives. Oncol. Rep. 2017, 38, 611–624.
  8. Kumari, P.; Ghosh, B.; Biswas, S. Nanocarriers for cancer-targeted drug delivery. J. Drug Target. 2015, 24, 179–191.
  9. Ferrari, M. Cancer nanotechnology: Opportunities and challenges. Nat. Rev. Cancer 2005, 5, 161–171.
  10. Matsumura, Y.; Maeda, H. A new concept for macromolecular therapeutics in cancer chemotherapy: Mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. Cancer Res. 1986, 46, 6387–6392.
  11. Jain, R.K. Transport of Molecules, Particles, and Cells in Solid Tumors. Annu. Rev. Biomed. Eng. 1999, 1, 241–263.
  12. Sindhwani, S.; Syed, A.M.; Ngai, J.; Kingston, B.R.; Maiorino, L.; Rothschild, J.; Macmillan, P.; Zhang, Y.; Rajesh, N.U.; Hoang, T.; et al. The entry of nanoparticles into solid tumours. Nat. Mater. 2020, 19, 566–575.
  13. Bulbake, U.; Doppalapudi, S.; Kommineni, N.; Khan, W. Liposomal Formulations in Clinical Use: An Updated Review. Pharmaceutics 2017, 9, 12.
  14. Shah, N.N.; Merchant, M.S.; Cole, D.E.; Jayaprakash, N.; Bernstein, D.; Delbrook, C.; Richards, K.; Widemann, B.C.; Wayne, A.S. Vincristine Sulfate Liposomes Injection (VSLI, Marqibo®): Results From a Phase I Study in Children, Adolescents, and Young Adults With Refractory Solid Tumors or Leukemias. Pediatr. Blood Cancer 2016, 63, 997–1005.
  15. Gill, P.S.; Wernz, J.; Scadden, D.T.; Cohen, P.; Mukwaya, G.M.; Von Roenn, J.H.; Jacobs, M.; Kempin, S.; Silverberg, I.; Gonzales, G.; et al. Randomized phase III trial of liposomal daunorubicin versus doxorubicin, bleomycin, and vincristine in AIDS-related Kaposi’s sarcoma. J. Clin. Oncol. 1996, 14, 2353–2364.
  16. O’Brien, M.E.R.; Wigler, N.; Inbar, M.; Rosso, R.; Grischke, E.; Santoro, A.; Catane, R.; Kieback, D.G.; Tomczak, P.; Ackland, S.P.; et al. Reduced cardiotoxicity and comparable efficacy in a phase IIItrial of pegylated liposomal doxorubicin HCl(CAELYX™/Doxil®) versus conventional doxorubicin forfirst-line treatment of metastatic breast cancer. Ann. Oncol. 2004, 15, 440–449.
  17. Roveri, M.; Bernasconi, M.; Leroux, J.-C.; Luciani, P. Peptides for tumor-specific drug targeting: State of the art and beyond. J. Mater. Chem. B 2017, 5, 4348–4364.
  18. Kirpotin, D.B.; Drummond, D.C.; Shao, Y.; Shalaby, M.R.; Hong, K.; Nielsen, U.B.; Marks, J.D.; Benz, C.; Park, J.W. Antibody Targeting of Long-Circulating Lipidic Nanoparticles Does Not Increase Tumor Localization but Does Increase Internalization in Animal Models. Cancer Res. 2006, 66, 6732–6740.
  19. Hamers-Casterman, C.; Atarhouch, T.; Muyldermans, S.; Robinson, G.; Hammers, C.; Songa, E.B.; Bendahman, N.; Hammers, R. Naturally occurring antibodies devoid of light chains. Nature 1993, 363, 446–448.
  20. Oliveira, S.; Heukers, R.; Sornkom, J.; Kok, R.J.; van Bergen En Henegouwen, P.M.P. Targeting tumors with nanobodies for cancer imaging and therapy. J. Control. Release 2013, 172, 607–617.
  21. Roveri, M.; Pfohl, A.; Jaaks, P.; Alijaj, N.; Leroux, J.-C.; Luciani, P.; Bernasconi, M. Prolonged circulation and increased tumor accumulation of liposomal vincristine in a mouse model of rhabdomyosarcoma. Nanomedicine 2017, 12, 1135–1151.
  22. Zhao, P.; Caretti, G.; Mitchell, S.; McKeehan, W.L.; Boskey, A.L.; Pachman, L.M.; Sartorelli, V.; Hoffman, E.P. Fgfr4 Is Required for Effective Muscle Regenerationin Vivo. J. Biol. Chem. 2006, 281, 429–438.
  23. Marics, I.; Padilla, F.; Guillemot, J.-F.; Scaal, M.; Marcelle, C. FGFR4 signaling is a necessary step in limb muscle differentiation. Development 2002, 129, 4559–4569.
  24. Khan, J.; Wei, J.S.; Ringnér, M.; Saal, L.H.; Ladanyi, M.; Westermann, F.; Berthold, F.; Schwab, M.; Antonescu, C.R.; Peterson, C.; et al. Classification and diagnostic prediction of cancers using gene expression profiling and artificial neural networks. Nat. Med. 2001, 7, 673–679.
  25. Tiong, K.H.; Tan, B.S.; Choo, H.L.; Chung, F.F.-L.; Hii, L.-W.; Tan, S.H.; Khor, N.T.W.; Wong, S.F.; See, S.-J.; Tan, Y.-F.; et al. Fibroblast growth factor receptor 4 (FGFR4) and fibroblast growth factor 19 (FGF19) autocrine enhance breast cancer cells survival. Oncotarget 2016, 7, 57633–57650.
  26. Gao, L.; Lang, L.; Zhao, X.; Shay, C.; Shull, A.Y.; Teng, Y. FGF19 amplification reveals an oncogenic dependency upon autocrine FGF19/FGFR4 signaling in head and neck squamous cell carcinoma. Oncogene 2018, 38, 2394–2404.
  27. Sawey, E.T.; Chanrion, M.; Cai, C.; Wu, G.; Zhang, J.; Zender, L.; Zhao, A.; Busuttil, R.W.; Yee, H.; Stein, L.; et al. Identification of a Therapeutic Strategy Targeting Amplified FGF19 in Liver Cancer by Oncogenomic Screening. Cancer Cell 2011, 19, 347–358.
  28. French, D.M.; Lin, B.C.; Wang, M.; Adams, C.; Shek, T.; Hötzel, K.; Bolon, B.; Ferrando, R.; Blackmore, C.; Schroeder, K.; et al. Targeting FGFR4 Inhibits Hepatocellular Carcinoma in Preclinical Mouse Models. PLoS ONE 2012, 7, e36713.
  29. June, C.H.; O’Connor, R.S.; Kawalekar, O.U.; Ghassemi, S.; Milone, M.C. CAR T cell immunotherapy for human cancer. Science 2018, 359, 1361–1365.
  30. Gill, S.; Maus, M.V.; Porter, D.L. Chimeric antigen receptor T cell therapy: 25years in the making. Blood Rev. 2016, 30, 157–167.
  31. Kochenderfer, J.N.; Dudley, M.E.; Feldman, S.A.; Wilson, W.H.; Spaner, D.E.; Maric, I.; Stetler-Stevenson, M.; Phan, G.Q.; Hughes, M.S.; Sherry, R.M.; et al. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor–transduced T cells. Blood 2012, 119, 2709–2720.
  32. Fry, T.J.; Shah, N.N.; Orentas, R.J.; Stetler-Stevenson, M.; Yuan, C.M.; Ramakrishna, S.; Wolters, P.; Martin, S.; Delbrook, C.; Yates, B.; et al. CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy. Nat. Med. 2018, 24, 20–28.
  33. Ali, S.A.; Shi, V.; Maric, I.; Wang, M.; Stroncek, D.F.; Rose, J.J.; Brudno, J.N.; Stetler-Stevenson, M.; Feldman, S.A.; Hansen, B.G.; et al. T cells expressing an anti–B-cell maturation antigen chimeric antigen receptor cause remissions of multiple myeloma. Blood 2016, 128, 1688–1700.
  34. Majzner, R.G.; Theruvath, J.L.; Nellan, A.; Heitzeneder, S.; Cui, Y.; Mount, C.W.; Rietberg, S.P.; Linde, M.H.; Xu, P.; Rota, C.; et al. CAR T Cells Targeting B7-H3, a Pan-Cancer Antigen, Demonstrate Potent Preclinical Activity Against Pediatric Solid Tumors and Brain Tumors. Clin. Cancer Res. 2019, 25, 2560–2574.
  35. Hegde, M.; Joseph, S.K.; Pashankar, F.; Derenzo, C.; Sanber, K.; Navai, S.; Byrd, T.T.; Hicks, J.; Xu, M.L.; Gerken, C.; et al. Tumor response and endogenous immune reactivity after administration of HER2 CAR T cells in a child with metastatic rhabdomyosarcoma. Nat. Commun. 2020, 11, 1–15.
  36. Moutel, S.; Bery, N.; Bernard, V.; Keller, L.; Lemesre, E.; De Marco, A.; Ligat, L.; Rain, J.-C.; Favre, G.; Olichon, A.; et al. NaLi-H1: A universal synthetic library of humanized nanobodies providing highly functional antibodies and intrabodies. eLife 2016, 5, e16228.
  37. Moutel, S.; Perez, F. Synthetic Single Domain Antibody. European Patent Application Number EP20305536, 20 May 2020.
  38. Turner, N.; Grose, R. Fibroblast growth factor signalling: From development to cancer. Nat. Rev. Cancer 2010, 10, 116–129.
  39. Sapra, P.; Allen, T.M. Internalizing antibodies are necessary for improved therapeutic efficacy of antibody-targeted liposomal drugs. Cancer Res. 2002, 62, 7190–7194.
  40. Porter, D.L.; Levine, B.L.; Kalos, M.; Bagg, A.; June, C.H. Chimeric Antigen Receptor–Modified T Cells in Chronic Lymphoid Leukemia. N. Engl. J. Med. 2011, 365, 725–733.
  41. Cao, L.; Yu, Y.; Bilke, S.; Walker, R.L.; Mayeenuddin, L.H.; Azorsa, D.O.; Yang, F.; Pineda, M.; Helman, L.J.; Meltzer, P.S. Genome-Wide Identification of PAX3-FKHR Binding Sites in Rhabdomyosarcoma Reveals Candidate Target Genes Important for Development and Cancer. Cancer Res. 2010, 70, 6497–6508.
  42. Vi, J.G.T.; Cheuk, A.T.; Tsang, P.S.; Chung, J.-Y.; Song, Y.K.; Desai, K.; Yu, Y.; Chen, Q.-R.; Shah, K.; Youngblood, V.; et al. Identification of FGFR4-activating mutations in human rhabdomyosarcomas that promote metastasis in xenotransplanted models. J. Clin. Investig. 2009, 119, 3395–3407.
  43. Sun, X.; Guo, W.; Shen, J.K.; Mankin, H.J.; Hornicek, F.J.; Duan, Z. Rhabdomyosarcoma: Advances in Molecular and Cellular Biology. Sarcoma 2015, 2015, 1–14.
  44. Seki, M.; Nishimura, R.; Yoshida, K.; Shimamura, T.; Shiraishi, Y.; Sato, Y.; Kato, M.; Chiba, K.; Tanaka, H.; Hoshino, N.; et al. Integrated genetic and epigenetic analysis defines novel molecular subgroups in rhabdomyosarcoma. Nat. Commun. 2015, 6, 7557.
  45. Crose, L.E.; Etheridge, K.T.; Chen, C.; Belyea, B.; Talbot, L.J.; Bentley, R.C.; Linardic, C.M. FGFR4 blockade exerts distinct antitumorigenic effects in human embryonal versus alveolar rhabdomyosarcoma. Clin. Cancer Res. 2012, 18, 3780–3790.
  46. Li, S.Q.; Cheuk, A.T.; Shern, J.F.; Song, Y.K.; Hurd, L.; Liao, H.; Wei, J.S.; Khan, J. Targeting Wild-Type and Mutationally Activated FGFR4 in Rhabdomyosarcoma with the Inhibitor Ponatinib (AP24534). PLoS ONE 2013, 8, e76551.
  47. Helsten, T.; Schwaederle, M.; Kurzrock, R. Fibroblast growth factor receptor signaling in hereditary and neoplastic disease: Biologic and clinical implications. Cancer Metastasis Rev. 2015, 34, 479–496.
  48. Oliveira, S.; Schiffelers, R.M.; Van Der Veeken, J.; Van Der Meel, R.; Vongpromek, R.; van Bergen en Henegouwen, P.M.P.; Storm, G.; Roovers, R.C. Downregulation of EGFR by a novel multivalent nanobody-liposome platform. J. Control. Release 2010, 145, 165–175.
  49. Caruso, H.G.; Hurton, L.V.; Najjar, A.; Rushworth, D.; Ang, S.; Olivares, S.; Mi, T.; Switzer, K.; Singh, H.; Huls, H.; et al. Tuning Sensitivity of CAR to EGFR Density Limits Recognition of Normal Tissue While Maintaining Potent Antitumor Activity. Cancer Res. 2015, 75, 3505–3518.
  50. Walker, A.J.; Majzner, R.G.; Zhang, L.; Wanhainen, K.; Long, A.H.; Nguyen, S.M.; Lopomo, P.; Vigny, M.; Fry, T.J.; Orentas, R.J.; et al. Tumor Antigen and Receptor Densities Regulate Efficacy of a Chimeric Antigen Receptor Targeting Anaplastic Lymphoma Kinase. Mol. Ther. 2017, 25, 2189–2201.
More
Information
Subjects: Oncology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , , , , , , ,
View Times: 293
Revisions: 2 times (View History)
Update Date: 24 Nov 2020
1000/1000