DCLK1 Monoclonal Antibody-Based CAR-T Cells as a Novel Treatment Strategy against Human Colorectal Cancers
"> Figure 1
Doublecortin-like kinase 1 (DCLK1) surface expression in human colorectal cancer cell lines. HT29, human colorectal cancer (CRC) cells were grown in adherent 2D tissue culture plates and as 3D spheroids. These cells were disassociated and subjected to Fluorescence-activated cell sorting (FACS) using humanized DCLK1 mAb (hCBT-15). HT29 cells grown in 2D culture demonstrated extracellular DCLK1 expression on 9.89% of cells (A—Top panel). HT29 cells grown in 3D matrices demonstrated extracellular DCLK1 expression on 45% of cells (A—bottom panel). The quantitative expression is represented in the bar graph (B). (C,D): HCT116, grown in 2D demonstrated DCLK1 extracellular expression on 20% and 3D demonstrated DCLK1 extracellular on 19.5% of cells. (E,F): LoVo, grown in 2D demonstrated DCLK1 extracellular expression on 22% of cells and 3D demonstrated DCLK1 extracellular on 25% of cells.
"> Figure 2Doublecortin-like kinase 1 (DCLK1) expression in human colorectal cancer patient tissues. (A) Human colorectal cancer (CRC) tissues and adjacent normal tissues were subjected to immunohistochemical analyses for DCLK1 using hCBT-15 mAb. Representative images are presented. Scale bars are presented at the image. (B) The Cancer Genome Atlas (TCGA) data was generated using Illumina HiSeqV2 RNA-sequencing data and was sorted for overall survival of the patients with low or high DCLK1 expression. (C) Moreover, TCGA data was generated and sorted for the expression of various epithelial-mesenchymal transition (EMT)-related transcription factors in the samples with low or high DCLK1 expression indicating that EMT and DCLK1 expression is correlated with poor overall survival. (D) Heatmap of the TCGA analyses for DCLK1 and immunomodulating checkpoints proteins programmed cell death-ligand 1 (PD-L1) and PD-L2. (E) TCGA data were sorted for samples with low or high PD-L1 expression. The samples with high PD-L1 had increased expression of DCLK1, cancer stem cells (CSCs)-associated DCLK1 isoform 2 and isoform 4 (p < 0.01 for all comparisons).
"> Figure 3Expression of chimeric antigen receptors (CARs) in Doublecortin-like kinase 1 (DCLK1) CAR-T cells (CBT-511). (A) The illustration represents the structure of DCLK1-CAR (CBT-511). The second-generation CAR with DCLK1 ScFv, CD28 co-stimulatory domain, and CD-3zeta activation domain was generated. (B) Expression of DCLK1-CAR in T cells transduced with lentiviral DCLK1-CAR by Fluorescence-activated cell sorting (FACS) with F(ab)2 antibody. CBT-511 cells were effectively transduced with DCLK1-CAR and expression of CAR (~20%) was confirmed by FACS with F(ab)2 antibody.
"> Figure 4CBT-511 treatment induced colorectal cancer (CRC) cell cytotoxicity and secretion of Interferon gamma (IFN-γ). Human CRC cells (HT29, HCT116, and LoVo) were subjected to real-time cytotoxicity assays (RTCA). Doublecortin-like kinase 1 (DCLK1) CAR-T cells effectively killed HT29 cells compared to mock CAR-T or T cells alone (A–C). Bar graphs in (A,B): Cytotoxicity of human DCLK1 (hDCLK1)-CAR-T and mouse Dclk1 (mDCLK1)-CAR-T cells versus T cells and Mock-CAR-T cells. The quantitation of RTCA from three independent experiments is shown. All pairwise comparisons were significant (p < 0.0001), except for Mock CAR-T cells versus T cells (p = 0.9363) and hDCLK-1-CAR-T cells 20:1 versus mDCLK-1-CAR-T cells 10:1 (p = 0.0703). Bar graphs in (C): All pairwise comparisons were significant (p < 0.05), except for Mock CAR-T cells versus T cells (p = 0.2017) and Mock CAR-T cells versus mDclk-1-CAR-T cells (p = 0.3769). All p-values were adjusted using Tukey’s method. (D) Treatment of HT29 cells grown in 2D with CBT-511 resulted in a dramatic release of IFN-γ (~25 pg/mL) as compared to mock CAR-T cells (0 pg/mL; p < 0.01). However, treatment of HT29 cells grown in 3D with CBT-511 resulted in an even higher IFN-γ release (~32 pg/mL versus 0 pg/mL; p < 0.01) compared to mock CAR-T. Similar treatment of HCT116 cells with CBT-511 resulted in increased IFN-γ release grown in 2D culture (150 pg/mL) compared to mock CAR-T (0 pg/mL). However, treatment of HCT116 cells grown in 3D with CBT-511 resulted in an even higher IFN-γ release (~250 pg/mL) compare to mock CAR-T (0 pg/mL; p < 0.01). (E) Treatment of LoVo cells with CBT-511 resulted in increased IFN-γ release grown in 2D culture (~50 pg/mL) compared to mock CAR-T (~30 pg/mL). However, treatment of LoVo cells grown in 3D with CBT-511 resulted in an even higher IFN-γ release (~95 pg/mL) compare to mock CAR-T (50 pg/mL). These data taken together suggests that CBT-511 is more active against CRC cells grown in 3D culture consistent with the increased clonogenic capacity of Tumor stem cells (TSCs).
"> Figure 5CBT-511 treatment reduced LoVo, human colorectal cancer (CRC) cell-line induced tumor xenograft growth. (A) LoVo, human CRC cells were injected subcutaneously into NOD Scid gamma (NSG™) mice and the tumors were allowed to grow. On days 7, 14, and 21 post-cell implantations, the mice were injected intravenous (i.v.) with either Doublecortin-like kinase 1 (DCLK1) CAR-T or mock CAR-T cells (1 × 107 cells/mice). Tumor volumes were measured and all the mice were weighted during the experiment period. (B) CBT-511 treatment resulted in a significant (* p = 0.02) decreased tumor xenograft growth compared to treatment with mock CAR-T cells. The mice treated with CBT-511 had smaller tumors compared to mock CAR-T treated mice. (C) There were no significant differences in the animal weights between CBT-511 and mock CAR-T treated mice.
">
Abstract
CAR-T (chimeric antigen receptor T cells) immunotherapy is effective in many hematological cancers; however, efficacy in solid tumors is disappointing. Doublecortin-like kinase 1 (DCLK1) labels tumor stem cells (TSCs) in genetic mouse models of colorectal cancer (CRC). Here, we describe a novel CAR-T targeting DCLK1 (CBT-511; with our proprietary DCLK1 single-chain antibody variable fragment) as a treatment strategy to eradicate CRC TSCs. The cell surface expression of DCLK1 and cytotoxicity of CBT-511 were assessed in CRC cells (HT29, HCT116, and LoVo). LoVo-derived tumor xenografts in NOD Scid gamma (NSG™) mice were treated with CBT-511 or mock CAR-T cells. Adherent CRC cells express surface DCLK1 (two-dimensional, 2D). A 4. 5-fold increase in surface DCLK1 was observed when HT29 cells were grown as spheroids (three-dimensional, 3D). CBT-511 induced cytotoxicity (2D; p < 0. 0001), and increased Interferon gamma (IFN-γ) release in CRC cells (2D) compared to mock CAR-T (p < 0. 0001). Moreover, an even greater increase in IFN-γ release was observed when cells were grown in 3D V体育官网入口. CBT-511 reduced tumor growth by approximately 50 percent compared to mock CAR-T. These data suggest that CRC cells with increased clonogenic capacity express increased surface DCLK1. A DCLK1-targeted CAR-T can induce cytotoxicity in vitro and inhibit xenograft growth in vivo. Keywords: DCLK1; CAR-T; tumor stem cells; immunotherapy; clonogenicity ."V体育安卓版" 1. Introduction
2. Results
2.1. DCLK1 mAb CBT-15 Has High Binding Affinity to DCLK1
2.2. DCLK1 Is Upregulated in Human CRC and CBT-15 Specifically Recognizes DCLK1 in CRC
2.3. CBT-511, CAR-T Cells Generated with DCLK1 CBT-15 Antibody ScFv, Recognizes DCLK1 Protein
"V体育平台登录" 2.4. CBT-511 Effectively Kills CRC Cells and Induces the Secretion of IFN-γ
2.5. CBT-511 Blocks Subcutaneous LoVo CRC Cells-Derived Xenograft Tumor Growth In Vivo
VSports - 3. Discussion
4. Materials and Methods (V体育2025版)
V体育官网入口 - 4.1. Cells, Primary Tissues
4.2. Monoclonal Antibody (V体育官网)
4.3. Generation of CAR-Encoding Lentivirus (V体育官网)
"VSports在线直播" 4.4. Generation and Expansion of CAR-T Cells
4.5. Flow Cytometry
4.6. Cytokine Induction Assay
4.7. Real-Time Cytotoxicity Assay (RTCA)
4.8. Mouse Xenograft Study
V体育平台登录 - 4.9. Statistical Analysis
5. Conclusions
6. Patents
Supplementary Materials
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- American-Cancer-Society. What is the Survival Rates for Colorectal Cancer by Stage? Available online: http://www.cancer.org/cancer/colonandrectumcancer/detailedguide/colorectal-cancer-survival-rates (accessed on 23 June 2019).
- Jordan, C.T.; Guzman, M.L.; Noble, M. Cancer stem cells. N. Engl. J. Med. 2006, 355, 1253–1261. [Google Scholar] [CrossRef] [PubMed]
- Clarke, M.F.; Dick, J.E.; Dirks, P.B.; Eaves, C.J.; Jamieson, C.H.; Jones, D.L.; Visvader, J.; Weissman, I.L.; Wahl, G.M. Cancer stem cells-perspectives on current status and future directions: AACR Workshop on cancer stem cells. Cancer Res. 2006, 66, 9339–9344. [Google Scholar] [CrossRef] [PubMed]
- Beatty, G.L.; Gladney, W.L. Immune escape mechanisms as a guide for cancer immunotherapy. Clin. Cancer Res. 2015, 21, 687–692. [Google Scholar] [CrossRef] [PubMed]
- Markman, J.L.; Shiao, S.L. Impact of the immune system and immunotherapy in colorectal cancer. J. Gastrointest. Oncol. 2015, 6, 208–223. [Google Scholar] [CrossRef]
- Menon, S.; Shin, S.; Dy, G. Advances in cancer immunotherapy in solid tumors. Cancers 2016, 8, 106. [Google Scholar] [CrossRef]
- Yan, X.; Zhang, S.; Deng, Y.; Wang, P.; Hou, Q.; Xu, H. Prognostic factors for checkpoint inhibitor based immunotherapy: An update with new evidences. Front. Pharmacol. 2018, 9, 1050. ["V体育官网入口" Google Scholar] [CrossRef]
- Kouidhi, S.; Ben Ayed, F.; Benammar Elgaaied, A. Targeting tumor metabolism: A new challenge to improve immunotherapy. Front. Immunol. 2018, 9, 353. [V体育2025版 - Google Scholar] [CrossRef]
- Noble, F.; Mellows, T.; McCormick Matthews, L.H.; Bateman, A.C.; Harris, S.; Underwood, T.J.; Byrne, J.P.; Bailey, I.S.; Sharland, D.M.; Kelly, J.J.; et al. Tumour infiltrating lymphocytes correlate with improved survival in patients with oesophageal adenocarcinoma. Cancer Immunol. Immunother. 2016, 65, 651–662. [Google Scholar (V体育2025版)] [CrossRef]
- Fan, F.; Samuel, S.; Evans, K.W.; Lu, J.; Xia, L.; Zhou, Y.; Sceusi, E.; Tozzi, F.; Ye, X.C.; Mani, S.A.; et al. Overexpression of snail induces epithelial-mesenchymal transition and a cancer stem cell-like phenotype in human colorectal cancer cells. Cancer Med. 2012, 1, 5–16. [Google Scholar] [CrossRef]
- Wang, Y.; Liu, Y.; Lu, J.; Zhang, P.; Wang, Y.; Xu, Y.; Wang, Z.; Mao, J.H.; Wei, G. Rapamycin inhibits FBXW7 loss-induced epithelial-mesenchymal transition and cancer stem cell-like characteristics in colorectal cancer cells. Biochem. Biophys. Res. Commun. 2013, 434, 352–356. [Google Scholar] [CrossRef]
- Brabletz, S.; Bajdak, K.; Meidhof, S.; Burk, U.; Niedermann, G.; Firat, E.; Wellner, U.; Dimmler, A.; Faller, G.; Schubert, J.; et al. The ZEB1/miR-200 feedback loop controls Notch signalling in cancer cells. EMBO J. 2011, 30, 770–782. [Google Scholar] [CrossRef] [PubMed]
- Krantz, S.B.; Shields, M.A.; Dangi-Garimella, S.; Munshi, H.G.; Bentrem, D.J. Contribution of epithelial-to-mesenchymal transition and cancer stem cells to pancreatic cancer progression. J. Surg. Res. 2012, 173, 105–112. ["VSports app下载" Google Scholar] [CrossRef] [PubMed]
- Wang, Z.; Li, Y.; Kong, D.; Banerjee, S.; Ahmad, A.; Azmi, A.S.; Ali, S.; Abbruzzese, J.L.; Gallick, G.E.; Sarkar, F.H. Acquisition of epithelial-mesenchymal transition phenotype of gemcitabine-resistant pancreatic cancer cells is linked with activation of the notch signaling pathway. Cancer Res. 2009, 69, 2400–2407. [VSports - Google Scholar] [CrossRef] [PubMed]
- Wellner, U.; Schubert, J.; Burk, U.C.; Schmalhofer, O.; Zhu, F.; Sonntag, A.; Waldvogel, B.; Vannier, C.; Darling, D.; zur Hausen, A.; et al. The EMT-activator ZEB1 promotes tumorigenicity by repressing stemness-inhibiting microRNAs. Nat. Cell Biol. 2009, 11, 1487–1495. ["V体育ios版" Google Scholar] [CrossRef]
- Lamouille, S.; Xu, J.; Derynck, R. Molecular mechanisms of epithelial-mesenchymal transition. Nat. Rev. Mol. Cell Biol. 2014, 15, 178–196. [VSports - Google Scholar] [CrossRef]
- Brabletz, T. EMT and MET in metastasis: Where are the cancer stem cells? Cancer Cell 2012, 22, 699–701. ["V体育平台登录" Google Scholar] [CrossRef]
- Choi, J.E.; Bae, J.S.; Kang, M.J.; Chung, M.J.; Jang, K.Y.; Park, H.S.; Moon, W.S. Expression of epithelial-mesenchymal transition and cancer stem cell markers in colorectal adenocarcinoma: Clinicopathological significance. Oncol. Rep. 2017, 38, 1695–1705. [Google Scholar] [CrossRef]
- Sureban, S.M.; Madhoun, M.F.; May, R.; Qu, D.; Ali, N.; Fazili, J.; Weygant, N.; Chandrakesan, P.; Ding, K.; Lightfoot, S.A.; et al. Plasma DCLK1 is a marker of hepatocellular carcinoma (HCC): Targeting DCLK1 prevents HCC tumor xenograft growth via a microRNA-dependent mechanism. Oncotarget 2015, 6, 37200–37215. ["V体育安卓版" Google Scholar] [CrossRef]
- Sureban, S.M.; May, R.; Lightfoot, S.A.; Hoskins, A.B.; Lerner, M.; Brackett, D.J.; Postier, R.G.; Ramanujam, R.; Mohammed, A.; Rao, C.V.; et al. DCAMKL-1 regulates epithelial-mesenchymal transition in human pancreatic cells through a miR-200a-dependent mechanism. Cancer Res. 2011, 71, 2328–2338. [Google Scholar (V体育平台登录)] [CrossRef]
- Sureban, S.M.; May, R.; Mondalek, F.G.; Qu, D.; Ponnurangam, S.; Pantazis, P.; Anant, S.; Ramanujam, R.P.; Houchen, C.W. Nanoparticle-based delivery of siDCAMKL-1 increases microRNA-144 and inhibits colorectal cancer tumor growth via a Notch-1 dependent mechanism. J. Nanobiotechnol. 2011, 9, 40. [Google Scholar] [CrossRef]
- Sureban, S.M.; May, R.; Qu, D.; Weygant, N.; Chandrakesan, P.; Ali, N.; Lightfoot, S.A.; Pantazis, P.; Rao, C.V.; Postier, R.G.; et al. DCLK1 regulates pluripotency and angiogenic factors via microRNA-dependent mechanisms in pancreatic cancer. PLoS ONE 2013, 8, e73940. [Google Scholar] [CrossRef] [PubMed]
- Sureban, S.M.; May, R.; Ramalingam, S.; Subramaniam, D.; Natarajan, G.; Anant, S.; Houchen, C.W. Selective blockade of DCAMKL-1 results in tumor growth arrest by a Let-7a MicroRNA-dependent mechanism. Gastroenterology 2009, 137, 649–659. [Google Scholar] [CrossRef] [PubMed]
- Sureban, S.M.; May, R.; Weygant, N.; Qu, D.; Chandrakesan, P.; Bannerman-Menson, E.; Ali, N.; Pantazis, P.; Westphalen, C.B.; Wang, T.C.; et al. XMD8-92 inhibits pancreatic tumor xenograft growth via a DCLK1-dependent mechanism. Cancer Lett. 2014, 351, 151–161. [Google Scholar (VSports在线直播)] [CrossRef] [PubMed]
- Nakanishi, Y.; Seno, H.; Fukuoka, A.; Ueo, T.; Yamaga, Y.; Maruno, T.; Nakanishi, N.; Kanda, K.; Komekado, H.; Kawada, M.; et al. Dclk1 distinguishes between tumor and normal stem cells in the intestine. Nat. Genet. 2013, 45, 98. [Google Scholar] [CrossRef] [PubMed]
- Westphalen, C.B.; Asfaha, S.; Hayakawa, Y.; Takemoto, Y.; Lukin, D.J.; Nuber, A.H.; Brandtner, A.; Setlik, W.; Remotti, H.; Muley, A.; et al. Long-lived intestinal tuft cells serve as colon cancer-initiating cells. J. Clin. Invest. 2014, 124, 1283–1295. [Google Scholar] [CrossRef] [PubMed]
- Weygant, N.; Ge, Y.; Qu, D.; Kaddis, J.S.; Berry, W.L.; May, R.; Chandrakesan, P.; Bannerman-Menson, E.; Vega, K.J.; Tomasek, J.J.; et al. Survival of patients with gastrointestinal cancers can be predicted by a surrogate microRNA signature for cancer stem-like cells marked by dclk1 kinase. Cancer Res. 2016, 76, 4090–4099. [Google Scholar] [CrossRef] [PubMed]
- Maus, M.V. Designing CAR T cells for glioblastoma. Oncoimmunology 2015, 4, e1048956. [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]
- Gross, G.; Eshhar, Z. Therapeutic potential of T cell chimeric antigen receptors (CARs) in cancer treatment: Counteracting off-tumor toxicities for safe CAR T cell therapy. Annu. Rev. Pharmacol. Toxicol. 2016, 56, 59–83. [Google Scholar] [CrossRef]
- Abken, H. Adoptive therapy with CAR redirected T cells: The challenges in targeting solid tumors. Immunotherapy 2015, 7, 535–544. [Google Scholar] [CrossRef]
- Lanitis, E.; Dangaj, D.; Irving, M.; Coukos, G. Mechanisms regulating T-cell infiltration and activity in solid tumors. Ann. Oncol. 2017, 28, xii18–xii32. [Google Scholar] [CrossRef] [PubMed]
- Maimela, N.R.; Liu, S.; Zhang, Y. Fates of CD8+ T cells in tumor microenvironment. Comput. Struct. Biotechnol. J. 2019, 17, 1–13. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Yan, L.; Liu, B. Critical factors in chimeric antigen receptor-modified T-cell (CAR-T) therapy for solid tumors. Onco Targets Ther. 2019, 12, 193–204. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Brown, C.E.; Mackall, C.L. CAR T cell therapy: Inroads to response and resistance. Nat. Rev. Immunol. 2019, 19, 73–74. [Google Scholar] [CrossRef]
- Cheadle, E.J.; Gornall, H.; Baldan, V.; Hanson, V.; Hawkins, R.E.; Gilham, D.E. CAR T cells: Driving the road from the laboratory to the clinic. Immunol. Rev. 2014, 257, 91–106. [Google Scholar] [CrossRef]
- Berahovich, R.; Zhou, H.; Xu, S.; Wei, Y.; Guan, J.; Guan, J.; Harto, H.; Fu, S.; Yang, K.; Zhu, S.; et al. CAR-T cells based on novel BCMA monoclonal antibody block multiple myeloma cell growth. Cancers 2018, 10, 323. ["V体育2025版" Google Scholar] [CrossRef]
- Golubovskaya, V.M.; Berahovich, R.; Xu, Q.; Zhou, H.; Xu, S.; Guan, J.; Harto, H.; Li, L.; Wu, L. GITR domain inside CAR co-stimulates activity of CAR-T cells against cancer. Front. Biosci. 2018, 23, 2245–2254. [VSports手机版 - Google Scholar] [CrossRef]
- Golubovskaya, V. CAR-T cell therapy: From the bench to the bedside. Cancers 2017, 9, 150. [Google Scholar] [CrossRef]
- Golubovskaya, V.; Berahovich, R.; Zhou, H.; Xu, S.; Harto, H.; Li, L.; Chao, C.C.; Mao, M.M.; Wu, L. CD47-CAR-T Cells Effectively Kill Target Cancer Cells and Block Pancreatic Tumor Growth. Cancers 2017, 9, 139. [Google Scholar] [CrossRef] [PubMed]
- Berahovich, R.; Xu, S.; Zhou, H.; Harto, H.; Xu, Q.; Garcia, A.; Liu, F.; Golubovskaya, V.M.; Wu, L. FLAG-tagged CD19-specific CAR-T cells eliminate CD19-bearing solid tumor cells in vitro and in vivo. Front. Biosci. 2017, 22, 1644–1654. [Google Scholar]
- Berahovich, R.; Liu, X.; Zhou, H.; Tsadik, E.; Xu, S.; Golubovskaya, V.; Wu, L. Hypoxia selectively impairs CAR-T Cells in vitro. Cancers 2019, 11, 602. [Google Scholar] [CrossRef] [PubMed]
- Golubovskaya, V.; Wu, L. Different subsets of T cells, memory, effector functions, and CAR-T immunotherapy. Cancers 2016, 8, 36. [Google Scholar] [CrossRef]
- Xu, Q.; Harto, H.; Berahovich, R.; Xu, S.; Zhou, H.; Golubovskaya, V.; Wu, L. Generation of CAR-T cells for cancer immunotherapy. Methods Mol. Biol. 2019, 1884, 349–360. [Google Scholar] [CrossRef]
- Ge, Y.; Weygant, N.; Qu, D.; May, R.; Berry, W.L.; Yao, J.; Chandrakesan, P.; Zheng, W.; Zhao, L.; Zhao, K.L.; et al. Alternative splice variants of DCLK1 mark cancer stem cells, promote self-renewal and drug-resistance, and can be targeted to inhibit tumorigenesis in kidney cancer. Int. J. Cancer 2018, 143, 1162–1175. [Google Scholar] [CrossRef]
- Qu, D.; Weygant, N.; Yao, J.; Chandrakesan, P.; Berry, W.L.; May, R.; Pitts, K.; Husain, S.; Lightfoot, S.; Li, M.; et al. Overexpression of DCLK1-AL increases tumor cell invasion, drug resistance, and KRAS activation and can be targeted to inhibit tumorigenesis in pancreatic cancer. J. Oncol. 2019, 2019, 6402925. [Google Scholar (V体育官网入口)] [CrossRef]
- Sarkar, S.; Popov, V.L.; O’Connell, M.R.; Stevenson, H.L.; Lee, B.S.; Obeid, R.A.; Luthra, G.K.; Singh, P. A novel antibody against cancer stem cell biomarker, DCLK1-S, is potentially useful for assessing colon cancer risk after screening colonoscopy. Lab. Investig. 2017, 97, 1245–1261. [Google Scholar] [CrossRef]
- Mu, L.; Huang, K.; Hu, Y.; Yan, C.; Li, X.; Tao, D.; Gong, J.; Qin, J. Small-sized colorectal cancer cells harbor metastatic tumor-initiating cells. Oncotarget 2017, 8, 107907–107919. [Google Scholar] [CrossRef]
- Ahmed, D.; Eide, P.W.; Eilertsen, I.A.; Danielsen, S.A.; Eknaes, M.; Hektoen, M.; Lind, G.E.; Lothe, R.A. Epigenetic and genetic features of 24 colon cancer cell lines. Oncogenesis 2013, 2, e71. [Google Scholar] [CrossRef]
- Ji, D.; Zhan, T.; Li, M.; Yao, Y.; Jia, J.; Yi, H.; Qiao, M.; Xia, J.; Zhang, Z.; Ding, H.; et al. Enhancement of sensitivity to chemo/radiation therapy by using miR-15b against DCLK1 in colorectal cancer. Stem Cell Rep. 2018, 11, 1506–1522. [V体育平台登录 - Google Scholar] [CrossRef] [PubMed]
- Turano, M.; Costabile, V.; Cerasuolo, A.; Duraturo, F.; Liccardo, R.; Delrio, P.; Pace, U.; Rega, D.; Dodaro, C.A.; Milone, M.; et al. Characterisation of mesenchymal colon tumour-derived cells in tumourspheres as a model for colorectal cancer progression. Int. J. Oncol. 2018, 53, 2379–2396. [Google Scholar] [CrossRef] [PubMed]
- Bao, Y.; Lu, Y.; Wang, X.; Feng, W.; Sun, X.; Guo, H.; Tang, C.; Zhang, X.; Shi, Q.; Yu, H. Eukaryotic translation initiation factor 5A2 (eIF5A2) regulates chemoresistance in colorectal cancer through epithelial mesenchymal transition. Cancer Cell Int. 2015, 15, 109. [Google Scholar (V体育官网入口)] [CrossRef] [PubMed]
- Larson, S.M.; Truscott, L.C.; Chiou, T.T.; Patel, A.; Kao, R.; Tu, A.; Tyagi, T.; Lu, X.; Elashoff, D.; De Oliveira, S.N. Pre-clinical development of gene modification of haematopoietic stem cells with chimeric antigen receptors for cancer immunotherapy. Hum. Vaccines Immunother. 2017, 13, 1094–1104. [Google Scholar] [CrossRef]
- Maliar, A.; Servais, C.; Waks, T.; Chmielewski, M.; Lavy, R.; Altevogt, P.; Abken, H.; Eshhar, Z. Redirected T cells that target pancreatic adenocarcinoma antigens eliminate tumors and metastases in mice. Gastroenterology 2012, 143, 1375–1384. [Google Scholar (VSports最新版本)] [CrossRef]
- Weygant, N.; Qu, D.; May, R.; Chandrakesan, P.; Ge, Y.; Ryan, C.D.; An, G.; Schlosser, M.J.; Bannerman-Menson, E.; Houchen, C.W. Systemic delivery of CBT-15G DCLK1-targeted monoclonal antibody dramatically decreases tumorigenesis in a xenograft model of pancreatic cancer. Cancer Res. 2016, 76, Am2016–Am2577. [Google Scholar (VSports)]





© 2019 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 (http://creativecommons.org/licenses/by/4.0/).
"V体育ios版" Share and Cite
Sureban, S.M.; Berahovich, R.; Zhou, H.; Xu, S.; Wu, L.; Ding, K.; May, R.; Qu, D.; Bannerman-Menson, E.; Golubovskaya, V.; et al. DCLK1 Monoclonal Antibody-Based CAR-T Cells as a Novel Treatment Strategy against Human Colorectal Cancers. Cancers 2020, 12, 54. https://doi.org/10.3390/cancers12010054
Sureban SM, Berahovich R, Zhou H, Xu S, Wu L, Ding K, May R, Qu D, Bannerman-Menson E, Golubovskaya V, et al. DCLK1 Monoclonal Antibody-Based CAR-T Cells as a Novel Treatment Strategy against Human Colorectal Cancers. Cancers. 2020; 12(1):54. https://doi.org/10.3390/cancers12010054
Chicago/Turabian StyleSureban, Sripathi M., Robert Berahovich, Hua Zhou, Shirley Xu, Lijun Wu, Kai Ding, Randal May, Dongfeng Qu, Edwin Bannerman-Menson, Vita Golubovskaya, and et al. 2020. "DCLK1 Monoclonal Antibody-Based CAR-T Cells as a Novel Treatment Strategy against Human Colorectal Cancers" Cancers 12, no. 1: 54. https://doi.org/10.3390/cancers12010054
APA StyleSureban, S. M., Berahovich, R., Zhou, H., Xu, S., Wu, L., Ding, K., May, R., Qu, D., Bannerman-Menson, E., Golubovskaya, V., & Houchen, C. W. (2020). DCLK1 Monoclonal Antibody-Based CAR-T Cells as a Novel Treatment Strategy against Human Colorectal Cancers. Cancers, 12(1), 54. https://doi.org/10.3390/cancers12010054

