Open Access

Receptor tyrosine kinases in carcinogenesis (Review)

Retraction in: /10.3892/ol.2020.12062

  • Authors:
    • Xiao-Ying Zhang
    • Pei-Ying Zhang
  • View Affiliations

  • Published online on: September 28, 2016     https://doi.org/10.3892/ol.2016.5200
  • Pages: 3679-3682
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Receptor tyrosine kinases (RTKs) are cell surface glycoproteins with enzymatic activity involved in the regulation of various important functions. In all-important physiological functions including differentiation, cell-cell interactions, survival, proliferation, metabolism, migration and signaling these receptors are the key players of regulation. Additionally, mutations of RTKs or their overexpression have been described in many human cancers and are being explored as a novel avenue for a new therapeutic approach. Some of the deregulated RTKs observed to be significantly affected in cancers included vascular endothelial growth factor receptor, epidermal growth factor receptor, fibroblast growth factor receptor, RTK-like orphan receptor 1 (ROR1) and the platelet‑derived growth factor receptor. These deregulated RTKs offer attractive possibilities for the new anticancer therapeutic approach involving specific targeting by monoclonal antibodies as well as kinase. The present review aimed to highlight recent perspectives of RTK ROR1 in cancer.

Introduction

The receptor tyrosine kinase (RTK)-like orphan receptor 1 (ROR1) expression has been observed to be significantly elevated in various blood and solid malignancies (16). These high expressions of ROR1 during carcinogenesis have encouraged investigation of the functional advantage conferred by ROR1 and ROR1 for targeted therapy in cancer. ROR1 expression was initially identified in chronic lymphocytic leukemia and subsequently in other malignancies (79). In 2001, two independent gene-profiling studies identified high ROR1 expressions (45-fold increase) in CLL as compared to normal mature B-lymphocytes (10,11). ROR1 protein expression has been shown in CLL (12) and in other malignancies such as acute lymphocytic leukemia (ALL), breast cancer, renal cell carcinoma, melanoma, lung adenocarcinoma and other lymphoid and myeloid malignancies (1320).

The number of ROR1 receptors on the surface of CLL cells was estimated to be in the range of 10,000/cells, which are sufficient to be targeted by monoclonal antibodies (21). ROR1 expression during CLL becomes further increased during disease progression. It has been observed that STAT3 is constitutive phosphorylated in CLL and has been shown to bind to the promoter region of ROR1 in CLL (22). Thus, STAT3 may be involved as a promoting factor in the expression of ROR1. Previous findings showed that ROR1 expression was also influenced by IL-6 in a STAT3 dose-dependent manner (23). Wnt5a is suggested to be one of the ligands proving favorable for ROR1 expression. Additionally, the above observation was confirmed by co-culturing of Wnt5a that resulted in increased survival of CLL cells in comparison to those without Wnt5a (24). On the other hand, in a similar study the Wnt5a-dependent survival of CLL cells was inhibited by ROR1 antisera (25). Previously, it was suggested that Wnt5a maintained phosphorylation of ROR1, thus favors its expression (15). ROR1 phosphorylation varied from patient to patient in CLL. High ROR1 phosphorylation intensity was found in progressive compared to non-progressive CLL. The same pattern was observed in other malignancies including breast, lung and ovarian cancer cells with an aggressive course (21). Furthermore, ROR1 expression at the protein level was significantly higher in aggressive tumors (15). Collectively these data suggested that the expression pattern of ROR1 is related to aggressiveness. ROR1 also showed an elevated expression during marginal zone lymphoma (MZL), multiple myeloma, follicular lymphoma (FL) and mantle cell lymphoma (MCL) at the gene and protein level. A high ROR1 expression was also observed in the cases of ALL (25). Consequently, ROR1 expression has been observed to be altered in various types of cancer. Thus, further studies are required to add to the present results and clarify the future perspectives of this factor.

Expression of ROR1 and associated factors in solid tumors

ROR1 expression was observed by staining in a recent study in various types of cancer including prostate, testicular, uterine, ovarian, lymphoma, adrenal and melanoma cancers (15). Strong ROR1 staining was found in 30% or greater of primary samples in the lung, colon and pancreatic cancers (22). In lung adenocarcinoma, TITF1 has been shown to regulate ROR1 expression. ROR1 upregulation was linked with the potentiation of epidermal growth factor (EGF) ligand-induced EGF receptor (EGFR) signaling, phosphorylation and activation of c-SRC (26).

ROR1 expression was also detected in all melanoma cell lines at mRNA level as well as protein level as assessed by western blotting and RT-PCR in addition to surface ROR1 staining by flow cytometry. Furthermore, knockdown of ROR1 in melanoma cell lines resulted in induction of apoptosis (21). ROR1 was shown to be phosphorylated at tyrosine and serine residues. Furthermore, monoclonal antibodies raised against ROR1 induced apoptosis. Another study on similar grounds observed ROR1 and ROR2 expression to be inversely expressed in melanoma cells and concluded that both ROR1 and ROR2 negatively regulate each other (17). Hypoxia also transfers ROR1 expression from moderate to aggressive in melanoma cells (17). Additionally, breast cancer cell lines with a strong ROR1 expression were more aggressive and invasive, but decreased in non-migrating cells. siRNA gene silencing therapy using specific ROR1 siRNA resulted in the downregulation of ROR1 expression in human breast cancer cell lines, where ROR1 was shown to activate PI3K-mediated AKT and CREB signaling pathways by interacting with casein kinase 1 (CK1). Wnt5a increased the survival of ROR1 expressing breast cancer cells, confirming the hypothesis of Wnt5a as a ligand for ROR1 (27).

ROR1 and EMT transition

High levels of ROR1 expression in patients and cell lines were linked to genes contributing to epithelial-mesenchymal transition (EMT) such as ZEB1 and vimentin and inversely related to adherent junction proteins. ROR1 expression was high in breast adenocarcinomas with a high level of EMT-associated genes and with a high capacity to metastasize. Silencing of ROR1 in the triple-negative breast cancer cell line MDA-MB321 by small hairpin (sh) RNA reduced in vitro cell migration as well as bone and lung foci xenografts (17). Knockdown of ROR1 in triple-negative breast cancer cells also reduced the EMT genes such as SNAI1, SNAI2, ZEB1 and vimentin (17). Similarly, ROR1-transfected MCF-7 cell lines showed a low level expression of adherent junction proteins E-cadherin and CK-19, which contribute to homing of cells at proliferation sites. However, ROR1 transfection did not change the expression levels of SNAI1, SNAI2 or vimentin.

ROR1 as therapeutic targets in cancer

ROR1, similar to other oncogenic RTKs, may be targeted in cancer. There are two main strategies; targeting the extracellular part of the receptor by monoclonal antibodies or by tyrosine kinase inhibitors directed against the intracellular kinase domain. Targeting the extracellular region of RTK by mAbs may disrupt the cytoplasmic kinase signaling by neutralization of the ligand, hampering the ligand binding, internalization of the receptor or by interacting with immune effectors targeting the tumor cells. Different RTKs HER-2, EGFR, vascular endothelial growth factor receptor (VEGFR) and VEGF and their ligands have been targeted in various types of cancer by mAbs. Trastumuzab was the first approved antibody against HER-2 for the treatment of breast cancer patients (28). Pertuzumab is another antibody approved against HER-2 to prevent dimerization of HER-2 with members of EGFR family. Similarly, tyrosine kinase inhibitors against the intracellular kinase domain in various type of cancer have been designed and approved for clinical use; gefitinib (29) and erlotinib (30) against EGFR and lapatinib (31) against HER2.

In a novel combination approach, the extracellular part of RTK is usually targeted by mAbs and the intracellular kinase domain by tyrosine kinase inhibitors. This combination has been shown to be synergistic in preventing the tumor growth and proliferation of cancer cells. Treatment of xenograft mice expressing HER-2 with trastuzumab and lapatinib resulted in significant tumor growth inhibition (21). Similarly, treatment of colon cancer cell lines expressing EGFR with cetuximab and gefitinib prevented proliferation and induced apoptosis (33). Targeting of HER-2 by trastuzumab and lapatinib has shown better clinical activity than either alone in HER-2 positive breast cancer patients (34).

Monoclonal antibodies against ROR1

The extracellular region of ROR1 contains Ig-like, CRD and KNG domains, which could be targeted by monoclonal antibodies. Monoclonal antibodies directed against these extracellular domains have been developed as potential therapeutic agents (35). The anti-ROR1 mAbs may kill cells by direct apoptosis or by activation of complement or immune effector cells. Most effective anti-ROR1 mAbs to induce significant apoptosis in CLL cells were those against the CRD and KNG domains. Anti-ROR1 CRD mAb also induced apoptosis in pancreatic cancer cell lines. De-phosphorylation of ROR1, the PI3Kδ, AKT and mTOR was also observed prior to apoptosis by the treatment of anti-ROR1 CRD mAb suggesting inhibition of downstream signaling but there was no effect on ERK and PKC proteins. These findings suggested that ROR1 signaling occurs via PI3K/AKT/mTOR axis in pancreatic cancer cell lines.

The anti-ROR1 mAbs also resulted in killing of melanoma cell lines by direct apoptosis as well as complement-dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC) (18). ESTDAB081 and ESTDAB094 melanoma cell lines were resistant to direct apoptosis by anti-ROR1 mAbs alone but sensitive to apoptosis by anti-ROR mAbs mediated through CDC and ADCC. Knockdown of ROR1 through specific siRNA resulted in apoptosis of melanoma cell lines. The effects of anti-ROR mAbs in TCL1 transgenic mice expressing ROR1+/CD5+/B220low leukemic B cells as a model for in vivo studies were analyzed (36). Two anti-ROR1 antibodies, D10 and 4A5 against different epitopes produced different effects in vivo. D10 anti-ROR1 mAb reduced the phosphorylation of AKT, but 4A5 mAb did not. Leukemic cells were cleared from the blood and spleen of transgenic mice following intravenous injections of D10 mAb, but not by 4A5 (37). Cirmtuzumab, a humanized anti-ROR1 mAb killed tumor cells and was internalized by malignant cells. Cirmtuzumab with antibody-drug conjugates (ADCs) cleared ROR1 expressing CLL cells in xenografted mice as well as induced apoptosis of breast and pancreas cell lines in vitro.

Anti-ROR1 antibodies have also been utilized to deliver toxins. Immunotoxin (BT-1) from Pseudomonas exotoxin (PE38) conjugated with the variable fragments of an anti-ROR1 mAb showed a dose-dependent and selective binding to leukemic cells from CLL and MCL patients, followed by internalization of the immunotoxin and subsequent apoptosis in vitro (38). ROR1 expression has been shown to be associated with EMT of tumor cells as indicated above (27). Treatment with anti-ROR1 mAbs prevented metastasis by downregulation of proteins involved in cell motility and metastasis of breast cancer cell lines.

Immunotherapeutic strategies targeting ROR1

ROR1 is highly expressed in many malignancies, but not in healthy adult tissues. ROR1 as an oncofetal antigen may thus be recognized by the immune system. A humoral immune response against ROR1 in CLL patients was observed after vaccination with Ad-CD154 transduced CLL cells (24). Antibodies induced against ROR1 blocked the interaction of Wnt5a and ROR1 and reduced proliferation of CLL cells. Lenalidomide, an immune modulating drug treatment induced ROR1 antibodies in CLL patients indicating that ROR1 may be an immunodominant epitope (39). CLL patients may also mount a type 1 T cell response against ROR1 and a humoral response against ROR1 (40). Transgenic leukemic ROR1+ mice immunized with a ROR1 peptide produced high titers of anti-ROR1 antibodies that inhibited the engraftment of ROR1+ CLL cells (41). ROR1 may therefore act as a tumor antigen for vaccination in a similar way as HER2 derived vaccine in breast cancer patients (42).

T cells from healthy donors or CLL patients have also been genetically modified to express ROR1-CART targeting ROR1+ tumor cells. CD8+ T cells engineered to express specific ROR1-CART lysed CLL and MCL cells but not mature normal cells. ROR1-CARTs were shown be as potent as CD19-CARTs in an immune deficient mouse model of human MCL (43). Clinical trials on ROR1-CART are expected to start.

Tyrosine kinase inhibitors against ROR1

Most oncogenic RTKs are highly upregulated and activated in malignant cells but have no or low expression and activity in normal tissues (44). RTKs in tumors may also be targeted by TKIs against the intracellular kinase domain (45). Axl is an RTK constitutively expressed in CLL and by targeting the intracellular kinase domain by a specific inhibitor, R428, a robust apoptosis of CLL cells was induced in a dose- and time-dependent manner (46). The VEGFR inhibitors vatalanib and pazopanib decreased phosphorylation of the VEGF receptor and induced apoptosis of CLL cells in clinically achievable concentrations with a mild cytotoxic effect on healthy B cells (47). Tyrosine kinase inhibitors may also target the intracellular kinase domain of ROR1. Treatment of CLL cells with ROR1 TKI dephosphorylated ROR1. ROR1 TKI induced apoptosis in CLL cells via caspase activation, PARP cleavage and downregulation of Mcl-1 and Bcl-2. Oral administration of ROR1 TKI in xenografted transplanted NOD SCID mice reduced leukemic cells.

Conclusions

It can be concluded from the above that RTK ROR1 may be a promising therapeutic target for ROR1 TKI and mAbs in CLL and other malignancies.

References

1 

Daneshmanesh AH, Hojjat-Farsangi M, Khan AS, Jeddi- Tehrani M, Akhondi MM, Bayat AA, Ghods R, Mahmoudi AR, Hadavi R, Österborg A, et al: Monoclonal antibodies against ROR1 induce apoptosis of chronic lymphocytic leukemia (CLL) cells. Leukemia. 26:1348–1355. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Shabani M, Naseri J and Shokri F: Receptor tyrosine kinase-like orphan receptor 1: a novel target for cancer immunotherapy. Expert Opin Ther Targets. 19:941–955. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Baskar S, Kwong KY, Hofer T, Levy JM, Kennedy MG, Lee E, Staudt LM, Wilson WH, Wiestner A and Rader C: Unique cell surface expression of receptor tyrosine kinase ROR1 in human B-cell chronic lymphocytic leukemia. Clin Cancer Res. 14:396–404. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Barna G, Mihalik R, Timár B, Tömböl J, Csende Z, Sebestyén A, Bödör C, Csernus B, Reiniger L, Peták I, et al: ROR1 expression is not a unique marker of CLL. Hematol Oncol. 29:17–21. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Zhang S, Chen L, Wang-Rodriguez J, Zhang L, Cui B, Frankel W, Wu R and Kipps TJ: The onco-embryonic antigen ROR1 is expressed by a variety of human cancers. Am J Pathol. 181:1903–1910. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Wurz GT, Kao CJ and DeGregorio MW: Novel cancer antigens for personalized immunotherapies: latest evidence and clinical potential. Ther Adv Med Oncol. 8:4–31. 2016.PubMed/NCBI

7 

Uhrmacher S, Schmidt C, Erdfelder F, Poll-Wolbeck SJ, Gehrke I, Hallek M and Kreuzer KA: Use of the receptor tyrosine kinase-like orphan receptor 1 (ROR1) as a diagnostic tool in chronic lymphocytic leukemia (CLL). Leuk Res. 35:1360–1366. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Ida L, Yamaguchi T, Yanagisawa K, Kajino T, Shimada Y, Suzuki M and Takahashi T: Receptor tyrosine kinase-like orphan receptor 1, a target of NKX2-1/TTF-1 lineage-survival oncogene, inhibits apoptosis signal-regulating kinase 1-mediated pro-apoptotic signaling in lung adenocarcinoma. Cancer Sci. 107:155–161. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Zhang H, Qiu J, Ye C, Yang D, Gao L, Su Y, Tang X, Xu N, Zhang D, Xiong L, Mao Y, Li F and Zhu J: ROR1 expression correlated with poor clinical outcome in human ovarian cancer. Sci Rep. 4:58112014.PubMed/NCBI

10 

Rosenwald A, Alizadeh AA, Widhopf G, Simon R, Davis RE, Yu X, Yang L, Pickeral OK, Rassenti LZ, Powell J, et al: Relation of gene expression phenotype to immunoglobulin mutation genotype in B cell chronic lymphocytic leukemia. J Exp Med. 194:1639–1647. 2001. View Article : Google Scholar : PubMed/NCBI

11 

Klein U, Tu Y, Stolovitzky GA, Mattioli M, Cattoretti G, Husson H, Freedman A, Inghirami G, Cro L, Baldini L, et al: Gene expression profiling of B cell chronic lymphocytic leukemia reveals a homogeneous phenotype related to memory B cells. J Exp Med. 194:1625–1638. 2001. View Article : Google Scholar : PubMed/NCBI

12 

Daneshmanesh AH, Mikaelsson E, Jeddi-Tehrani M, Bayat AA, Ghods R, Ostadkarampour M, Akhondi M, Lagercrantz S, Larsson C, Osterborg A, et al: Ror1, a cell surface receptor tyrosine kinase is expressed in chronic lymphocytic leukemia and may serve as a putative target for therapy. Int J Cancer. 123:1190–1195. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Shabani M, Asgarian-Omran H, Vossough P, Sharifian RA, Faranoush M, Ghragozlou S, Khoshnoodi J, Roohi A, Jeddi-Tehrani M, Mellstedt H, et al: Expression profile of orphan receptor tyrosine kinase (ROR1) and Wilms' tumor gene 1 (WT1) in different subsets of B-cell acute lymphoblastic leukemia. Leuk Lymphoma. 49:1360–1367. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Shabani M, Asgarian-Omran H, Jeddi-Tehrani M, Vossough P, Faranoush M, Sharifian RA, Toughe GR, Kordmahin M, Khoshnoodi J, Roohi A, et al: Overexpression of orphan receptor tyrosine kinase Ror1 as a putative tumor-associated antigen in Iranian patients with acute lymphoblastic leukemia. Tumour Biol. 28:318–326. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Zhang S, Chen L, Cui B, Chuang HY, Yu J, Wang-Rodriguez J, Tang L, Chen G, Basak GW and Kipps TJ: ROR1 is expressed in human breast cancer and associated with enhanced tumor-cell growth. PLoS One. 7:e311272012. View Article : Google Scholar : PubMed/NCBI

16 

Rabbani H, Ostadkarampour M, Manesh AH Danesh, Basiri A, Jeddi-Tehrani M and Forouzesh F: Expression of ROR1 in patients with renal cancer - a potential diagnostic marker. Iran Biomed J. 14:77–82. 2010.PubMed/NCBI

17 

O'Connell MP, Marchbank K, Webster MR, Valiga AA, Kaur A, Vultur A, Li L, Herlyn M, Villanueva J, Liu Q, et al: Hypoxia induces phenotypic plasticity and therapy resistance in melanoma via the tyrosine kinase receptors ROR1 and ROR2. Cancer Discov. 3:1378–1393. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Hojjat-Farsangi M, Ghaemimanesh F, Daneshmanesh AH, Bayat AA, Mahmoudian J, Jeddi-Tehrani M, Rabbani H and Mellstedt H: Inhibition of the receptor tyrosine kinase ROR1 by anti-ROR1 monoclonal antibodies and siRNA induced apoptosis of melanoma cells. PLoS One. 8:e611672013. View Article : Google Scholar : PubMed/NCBI

19 

Shabani M, Omran H Asgarian, Farsangi MH, Vossough P, Sharifian RA, Toughe GR, Razavi SM, Khoshnoodi J, Jeddi-Tehrani M, Rabbani H, et al: Comparative expression profile of orphan receptor tyrosine kinase ROR1 in Iranian patients with lymphoid and myeloid leukemias. Avicenna J Med Biotechnol. 3:119–125. 2011.PubMed/NCBI

20 

Daneshmanesh AH, Porwit A, Hojjat-Farsangi M, Jeddi-Tehrani M, Tamm KP, Grandér D, Lehmann S, Norin S, Shokri F, Rabbani H, et al: Orphan receptor tyrosine kinases ROR1 and ROR2 in hematological malignancies. Leuk Lymphoma. 54:843–850. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Hojjat-Farsangi M, Khan AS, Daneshmanesh AH, Moshfegh A, Sandin A, Mansouri L, Palma M, Lundin J, Österborg A and Mellstedt H: The tyrosine kinase receptor ROR1 is constitutively phosphorylated in chronic lymphocytic leukemia (CLL) cells. PLoS One. 8:e783392013. View Article : Google Scholar : PubMed/NCBI

22 

Borcherding N, Kusner D, Liu GH and Zhang W: ROR1, an embryonic protein with an emerging role in cancer biology. Protein Cell. 5:496–502. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Li P, Harris D, Liu Z, Liu J, Keating M and Estrov Z: Stat3 activates the receptor tyrosine kinase like orphan receptor-1 gene in chronic lymphocytic leukemia cells. PLoS One. 5:e118592010. View Article : Google Scholar : PubMed/NCBI

24 

Fukuda T, Chen L, Endo T, Tang L, Lu D, Castro JE, Widhopf GF II, Rassenti LZ, Cantwell MJ, Prussak CE, et al: Antisera induced by infusions of autologous Ad-CD154-leukemia B cells identify ROR1 as an oncofetal antigen and receptor for Wnt5a. Proc Natl Acad Sci USA. 105:3047–3052. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Bicocca VT, Chang BH, Masouleh BK, Muschen M, Loriaux MM, Druker BJ and Tyner JW: Crosstalk between ROR1 and the Pre-B cell receptor promotes survival of t(1;19) acute lymphoblastic leukemia. Cancer Cell. 22:656–667. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Yamaguchi T, Yanagisawa K, Sugiyama R, Hosono Y, Shimada Y, Arima C, Kato S, Tomida S, Suzuki M, Osada H, et al: NKX2-1/TITF1/TTF-1-Induced ROR1 is required to sustain EGFR survival signaling in lung adenocarcinoma. Cancer Cell. 21:348–361. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Cui B, Zhang S, Chen L, Yu J, Widhopf GF II, Fecteau JF, Rassenti LZ and Kipps TJ: Targeting ROR1 inhibits epithelial-mesenchymal transition and metastasis. Cancer Res. 73:3649–3660. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Graziano C: HER-2 breast assay, linked to Herceptin, wins FDA's okay. CAP Today. 12(1): 14–16. 1998.

29 

Herbst RS, Fukuoka M and Baselga J: Gefitinib - a novel targeted approach to treating cancer. Nat Rev Cancer. 4:956–965. 2004. View Article : Google Scholar : PubMed/NCBI

30 

Dowell J, Minna JD and Kirkpatrick P: Erlotinib hydrochloride. Nat Rev Drug Discov. 4:13–14. 2005. View Article : Google Scholar : PubMed/NCBI

31 

Xia W, Mullin RJ, Keith BR, Liu LH, Ma H, Rusnak DW, Owens G, Alligood KJ and Spector NL: Anti-tumor activity of GW572016: A dual tyrosine kinase inhibitor blocks EGF activation of EGFR/erbB2 and downstream Erk1/2 and AKT pathways. Oncogene. 21:6255–6263. 2002. View Article : Google Scholar : PubMed/NCBI

32 

Rimawi MF, Wiechmann LS, Wang YC, Huang C, Migliaccio I, Wu MF, Gutierrez C, Hilsenbeck SG, Arpino G, Massarweh S, et al: Reduced dose and intermittent treatment with lapatinib and trastuzumab for potent blockade of the HER pathway in HER2/neu-overexpressing breast tumor xenografts. Clin Cancer Res. 17:1351–1361. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Yuan HH, Han Y, Bian WX, Liu L and Bai YX: The effect of monoclonal antibody cetuximab (C225) in combination with tyrosine kinase inhibitor gefitinib (ZD1839) on colon cancer cell lines. Pathology. 44:547–551. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Guarneri V, Frassoldati A, Bottini A, Cagossi K, Bisagni G, Sarti S, Ravaioli A, Cavanna L, Giardina G, Musolino A, et al: Preoperative chemotherapy plus trastuzumab, lapatinib, or both in human epidermal growth factor receptor 2-positive operable breast cancer: results of the randomized phase II CHER-LOB study. J Clin Oncol. 30:1989–1995. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Yang J, Baskar S, Kwong KY, Kennedy MG, Wiestner A and Rader C: Therapeutic potential and challenges of targeting receptor tyrosine kinase ROR1 with monoclonal antibodies in B-cell malignancies. PLoS One. 6:e210182011. View Article : Google Scholar : PubMed/NCBI

36 

Widhopf GF II, Cui B, Ghia EM, Chen L, Messer K, Shen Z, Briggs SP, Croce CM and Kipps TJ: ROR1 can interact with TCL1 and enhance leukemogenesis in Eµ-TCL1 transgenic mice. Proc Natl Acad Sci USA. 111:793–798. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Cui B, Widhopf GF II, Prussak CE, Wu CCN, Sadarangani A, Zhang S, Lao F, Jamieson CHM, Carson DA and Kipps TJ: Cirmtuzumab vedotin (UC-961ADC3), an anti-ROR1-monomethyl auristatin E antibody-drug conjugate, is a potential treatment for ROR1-positive leukemia and solid tumors. Presented at 55th ASH Annual Meeting. (abstract 1637). 2013; https://ash.confex.com/ash/2013/webprogram/Paper65533.html

38 

Baskar S, Wiestner A, Wilson WH, Pastan I and Rader C: Targeting malignant B cells with an immunotoxin against ROR1. MAbs. 4:349–361. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Gilbert JA: Lenalidomide as first-line therapy for elderly CLL patients. Lancet Oncol. 14:e3452013. View Article : Google Scholar

40 

Hojjat-Farsangi M, Daneshmanesh AH, Osterborg A, Shokri F and Mellstedt H: Patients with chronic lymphocyte leukemia (CLL) have naturally occurring antibodies against the receptor tyrosine kinase (ROR1). Blood (ASH Annual Meeting abstracts). 118:17712011.

41 

Yu J, Cui B, Widhopf GF II, Chen L, Rassenti L, Wang Z, Ma S, Hayashi T, Carson DA and Kipps TJ: Preclinical development of ROR1 peptide based vaccine with activity against chroniclymphocytic leukemia in ROR1 transgenic mice. Blood (ASH Annual Meeting abstracts). 122:41742013.

42 

Milani A, Sangiolo D, Montemurro F, Aglietta M and Valabrega G: Active immunotherapy in HER2 overexpressing breast cancer: current status and future perspectives. Ann Oncol. 24:1740–1748. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Hudecek M, Lupo-Stanghellini MT, Kosasih PL, Sommermeyer D, Jensen MC, Rader C and Riddell SR: Receptor affinity and extracellular domain modifications affect tumor recognition by ROR1-specific chimeric antigen receptor T cells. Clin Cancer Res. 19:3153–3164. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Robertson SC, Tynan J and Donoghue DJ: RTK mutations and human syndromes: when good receptors turn bad. Trends Genet. 16:3682000. View Article : Google Scholar : PubMed/NCBI

45 

Zwick E, Bange J and Ullrich A: Receptor tyrosine kinase signalling as a target for cancer intervention strategies. Endocr Relat Cancer. 8:161–173. 2001. View Article : Google Scholar : PubMed/NCBI

46 

Ghosh AK, Secreto C, Boysen J, Sassoon T, Shanafelt TD, Mukhopadhyay D and Kay NE: The novel receptor tyrosine kinase Axl is constitutively active in B-cell chronic lymphocytic leukemia and acts as a docking site of nonreceptor kinases: implications for therapy. Blood. 117:1928–1937. 2011. View Article : Google Scholar : PubMed/NCBI

47 

Paesler J, Gehrke I, Gandhirajan RK, Filipovich A, Hertweck M, Erdfelder F, Uhrmacher S, Poll-Wolbeck SJ, Hallek M and Kreuzer KA: The vascular endothelial growth factor receptor tyrosine kinase inhibitors vatalanib and pazopanib potently induce apoptosis in chronic lymphocytic leukemia cells in vitro and in vivo. Clin Cancer Res. 16:3390–3398. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2016
Volume 12 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang X and Zhang X: Receptor tyrosine kinases in carcinogenesis (Review) Retraction in /10.3892/ol.2020.12062. Oncol Lett 12: 3679-3682, 2016
APA
Zhang, X., & Zhang, X. (2016). Receptor tyrosine kinases in carcinogenesis (Review) Retraction in /10.3892/ol.2020.12062. Oncology Letters, 12, 3679-3682. https://doi.org/10.3892/ol.2016.5200
MLA
Zhang, X., Zhang, P."Receptor tyrosine kinases in carcinogenesis (Review) Retraction in /10.3892/ol.2020.12062". Oncology Letters 12.5 (2016): 3679-3682.
Chicago
Zhang, X., Zhang, P."Receptor tyrosine kinases in carcinogenesis (Review) Retraction in /10.3892/ol.2020.12062". Oncology Letters 12, no. 5 (2016): 3679-3682. https://doi.org/10.3892/ol.2016.5200