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 + 1146 word(s) 1146 2021-08-08 13:34:55

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.
Liu, X.M. Targeted Liposomal. Encyclopedia. Available online: https://encyclopedia.pub/entry/13553 (accessed on 26 April 2024).
Liu XM. Targeted Liposomal. Encyclopedia. Available at: https://encyclopedia.pub/entry/13553. Accessed April 26, 2024.
Liu, X. Margaret. "Targeted Liposomal" Encyclopedia, https://encyclopedia.pub/entry/13553 (accessed April 26, 2024).
Liu, X.M. (2021, August 25). Targeted Liposomal. In Encyclopedia. https://encyclopedia.pub/entry/13553
Liu, X. Margaret. "Targeted Liposomal." Encyclopedia. Web. 25 August, 2021.
Targeted Liposomal
Edit

The targeted liposomes have been developed and utilized to deliver drugs to the tumor or tumor microenvironment with minimal non-specific distribution in normal tissues or organs. Various tumor-targeting ligands, such as small molecules, oligonucleotides, peptides, monoclonal antibodies (mAbs) and antigen-binding fragments (Fabs), have been conjugated with liposomes. 

combined chemotherapies targeted liposomes triple-negative breast cancer

1. Introduction

Triple-negative breast cancers (TNBCs) are the breast cancers that lack expression of estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor (HER)-2/neu. Currently chemotherapeutic agents are the most common clinical treatment strategies employed to suppress tumor growth, but TNBC patient responses differ from case to case. For instance, drug resistance due to drug efflux [1][2][3], apoptosis dysregulation [4][5], activation of survival, growth and invasion signaling pathways [6] or others [7][8] significantly limits their clinical efficacy and also leads to tumor recurrence and progression [9]. In addition, patients usually suffer from side effects, such as fatigue, emesis, hair loss, and anemia, due to a lack of an effective tumor targeting method.
The U.S. Food and Drug Administration (FDA) has approved liposomes as a drug delivery vehicle with guidance of “Chemistry, manufacturing, and controls; human pharmacokinetics and bioavailability; and labeling documentation” (FDA-2016-D-2817). The targeted liposomes have been developed and utilized to deliver drugs to the tumor or tumor microenvironment with minimal non-specific distribution in normal tissues or organs. Various tumor-targeting ligands, such as small molecules, oligonucleotides, peptides, monoclonal antibodies (mAbs) and antigen-binding fragments (Fabs), have been conjugated with liposomes. For example, the anti-epidermal growth factor receptor (EGFR) [10][11], HER2 [12] and vascular endothelial growth factor (VEGF) [13][14] antibodies or peptides have been linked to liposomal system to deliver doxorubicin or other medicines to breast cancers and other tumors. The fibronectin-mimetic peptide-PR_b [15], estrogen receptor-antagonist Tamoxifen [16] and peptide SP90 [17] have been used as linkers in liposomal drug formulation to treat breast cancers. Moreover, GAH mAb conjugated immunoliposomes have been fabricated to targeting deliver doxorubicin to treat human gastrointestinal cancers [18].
The EGFR, which stimulates the cancer proliferation via PI3K/RAS signaling, the repair of DNA damage and metastasis [19][20][21][22][23], is overexpressed in various tumors, e.g., TNBC (52–54%) [24][25], lung cancer (40%) [26][27], glioblastoma (50%), head and neck cancer (80–90%) [24][25][28], ovarian, cervical, bladder, gastric, endometrial and colorectal cancers [29]. EGFR is more predominant in TNBCs than other breast cancers [24][30], and usually correlates with tumor invasion and poor prognosis. From this perspective, anti-EGFR mAb was utilized in this study as a ligand to target TNBC. The targeted liposomal drug formulation is expected to prolong the circulation half-life and enhance the maximum tolerated dose.
Many innovative anti-cancer drugs failed in phase II clinical trials [31] although the pre-clinical results are promising. This could be attributed to the limitation of preclinical animal models such as lacking heterogeneity and tumor microenvironment. This challenge can be partially solved by applying patient-derived xenograft (PDX) models in the in vivo evaluation of the anti-tumor efficacy of new medicines. PDX models have been established by transplanting the cancerous cells or tissues from primary patient tumors and served as a good preclinical platform to predict the possible patient responses to new cancer medicine.

2. Targeted Liposomal Chemotherapies to Treat Triple-Negative Breast Cancer

Chemotherapies are still the major strategy to treat TNBC in clinics. We identified a new formulation of combined chemotherapies and also established a targeted delivery method for TNBC treatment, which could address the challenges of drug resistance or poor clinical efficacy as well as treatment related toxicities. We have evaluated a highly potent drug and several standard chemotherapies for cancer treatment, including DM1, GC, AC, and PTX [32], and two combinations of these drugs. Combining standard GC and potent DM1 can kill over 90% TNBC cells with significantly reduced IC50 value and also effectively inhibit TNBC tumor growth in both cell line-derived xenograft models and patient-derived xenograft models. In addition to the improved cytotoxicity, GC and DM1 have different anti-cancer mechanisms so the combination could reduce the possibility of drug resistance development during long-term treatment compared to monotherapy. Therefore, the combination of GC and DM1 has great potential to treat TNBC.
We established and optimized the procedures of neutral liposomes synthesis to pack chemotherapies, surface tagging of TNBC-targeting antibody (mAb-Lipo), PEGylation, drugs packing, purification and characterization following the published guideline and protocols [33][34][35][36][37][38][39][40][41][42][43][44] with optimization. Non-targeting liposomes [45][46][47][48][49] have been used to deliver chemotherapies and other therapies, but the mAb-Lipo has multiple advantages, such as cancer-specific targeting, high packing capability with the developed all-in-one synthesis procedure, and high plasma stability and prolonged half-life with integrated PEG. Importantly, our surface tagging technology enables conjugating single or two (even multiple) antibodies to achieve dual-targeting to cover more patients with heterogeneous tumors. In addition to chemotherapies, the cationic liposomes encapsulated plasmid DNA (named as lipoplexes) have been evaluated in clinical trials for cystic fibrosis [50], non-small-cell lung cancer [51], metastatic melanoma [52][53], and epithelial ovarian, fallopian tube or primary peritoneal cancers [54] treatment.
Literature [25][55][56][57], clinical data [24][25][55] and our immunohistochemistry staining of patient tissue microarray show that EGFR is an excellent surface receptor in human [58][59][60] and mouse [61][62][63] TNBCs. For example, the anti-EGFR cetuximab and panitumumab are used in clinic to treat head and neck cancer [64][65][66] and colorectal cancer [67][68][69]. Moreover, the cetuximab mediates antibody-dependent cell cytotoxicity (ADCC) in the intratumoral space and primes adaptive and innate cellular immunity [70]. By tagging anti-EGFR mAb (cetuximab) to the surface of liposomes, we not only achieve TNBC tumor targeting but also could integrate the immunotherapy of the mAb. Of course, further investigation is needed to delineate the possible integrated anti-TNBC mechanisms of the tagged mAb and delivered GC and DM1 in future.
The TNBC xenograft models derived from various cell lines have been widely used in vivo to evaluate the tumor treatment efficacy. The PDX models are more advanced to evaluate new therapies as they have multiple advantages such as capturing TNBC heterogeneity and tumor microenvironment. For instance, PDX tumors can accurately recapitulate the phenocopy and mutation status of patient tumors, and resemble and maintain the biological behavior correlating with high metastasis, high heterogeneity and poor survival of TNBC patient tumors. Limited by the fresh patient tissues assessment and pathology analysis, many research labs have difficulty to establish in-house TNBC PDX models. We evaluated the Jackson lab commercial PDX lines and established a robust procedure to passage and maintain PDX lines in the research lab. The identified EGFR overexpressing PDX lines can be used as a good model to evaluate the therapeutic efficiency of newly developed therapies.

3. Conclusions

The combination of chemotherapies with different anti-cancer mechanisms (gemcitabine and mertansine in this study) has great potential to treat the highly aggressive TNBC. The technical challenges to apply combined chemotherapies, including circulation stability and side effects, can be overcome by the application of a targeted liposomal delivery vehicle. Importantly, different drug combinations can be easily adapted to this system for the treatment of recurrent cancer. Despite the promising results, the developed new formulation needs further evaluation in the future, such as pharmacokinetics, dosage optimization, metastatic tumor treatment and immune modulatory response.

References

  1. Sissung, T.M.; Baum, C.E.; Kirkland, C.T.; Gao, R.; Gardner, E.R.; Figg, W.D. Pharmacogenetics of membrane transporters: An update on current approaches. Mol. Biotechnol. 2009, 44, 152–167.
  2. Yamada, A.; Ishikawa, T.; Ota, I.; Kimura, M.; Shimizu, D.; Tanabe, M.; Chishima, T.; Sasaki, T.; Ichikawa, Y.; Morita, S.; et al. High expression of ATP-binding cassette transporter ABCC11 in breast tumors is associated with aggressive subtypes and low disease-free survival. Breast Cancer Res. Treat. 2013, 137, 773–782.
  3. Mahmood, N.A.; Abdulghany, Z.; Al-Sudani, I.M. Expression of Aldehyde Dehydrogenase (ALDH1) and ATP Binding Cassette Transporter G2 (ABCG2) in Iraqi Patients with Colon Cancer and the Relation with Clinicopathological Features. Int. J. Mol. Cell. Med. 2019, 7, 234–240.
  4. Inao, T.; Iida, Y.; Moritani, T.; Okimoto, T.; Tanino, R.; Kotani, H.; Harada, M. Bcl-2 inhibition sensitizes triple-negative human breast cancer cells to doxorubicin. Oncotarget 2018, 9, 25545–25556.
  5. Campbell, K.J.; Dhayade, S.; Ferrari, N.; Sims, A.; Johnson, E.; Mason, S.; Dickson, A.; Ryan, K.M.; Kalna, G.; Edwards, J.; et al. MCL-1 is a prognostic indicator and drug target in breast cancer. Cell Death Dis. 2018, 9, 1–14.
  6. Lehmann, B.D.; Jovanović, B.; Chen, X.; Estrada, M.V.; Johnson, K.N.; Shyr, Y.; Moses, H.L.; Sanders, M.E.; Pietenpol, J.A. Refinement of Triple-Negative Breast Cancer Molecular Subtypes: Implications for Neoadjuvant Chemotherapy Selection. PLoS ONE 2016, 11, e0157368.
  7. Nedeljković, M.; Damjanović, A. Mechanisms of Chemotherapy Resistance in Triple-Negative Breast Cancer—How We Can Rise to the Challenge. Cells 2019, 8, 957.
  8. Wein, L.; Loi, S. Mechanisms of resistance of chemotherapy in early-stage triple negative breast cancer (TNBC). Breast 2017, 34, S27–S30.
  9. Zhang, H.-H.; Guo, X.-L. Combinational strategies of metformin and chemotherapy in cancers. Cancer Chemother. Pharmacol. 2016, 78, 13–26.
  10. Lehtinen, J.; Raki, M.; Bergström, K.A.; Uutela, P.; Lehtinen, K.; Hiltunen, A.; Pikkarainen, J.; Liang, H.; Pitkänen, S.; Määttä, A.-M.; et al. Pre-Targeting and Direct Immunotargeting of Liposomal Drug Carriers to Ovarian Carcinoma. PLoS ONE 2012, 7, e41410.
  11. Kim, S.K.; Huang, L. Nanoparticle delivery of a peptide targeting EGFR signaling. J. Control. Release 2012, 157, 279–286.
  12. Dehkordi, N.G.; Elahian, F.; Khosravian, P.; Mirzaei, S.A. Intelligent TAT-coupled anti-HER2 immunoliposomes knock downed MDR1 to produce chemosensitize phenotype of multidrug resistant carcinoma. J. Cell. Physiol. 2019, 234, 20769–20778.
  13. Matusewicz, L.; Filip-Psurska, B.; Psurski, M.; Tabaczar, S.; Podkalicka, J.; Wietrzyk, J.; Ziółkowski, P.; Czogalla, A.; Sikorski, A.F. EGFR-targeted immunoliposomes as a selective delivery system of simvastatin, with potential use in treatment of triple-negative breast cancers. Int. J. Pharm. 2019, 569, 118605.
  14. Wöll, S.; Dickgiesser, S.; Rasche, N.; Schiller, S.; Scherließ, R. Sortagged anti-EGFR immunoliposomes exhibit increased cytotoxicity on target cells. Eur. J. Pharm. Biopharm. 2019, 136, 203–212.
  15. Shroff, K.; Kokkoli, E. PEGylated Liposomal Doxorubicin Targeted to α5β1-Expressing MDA-MB-231 Breast Cancer Cells. Langmuir 2012, 28, 4729–4736.
  16. Jain, A.S.; Goel, P.N.; Shah, S.; Dhawan, V.V.; Nikam, Y.; Gude, R.P.; Nagarsenker, M.S. Tamoxifen guided liposomes for targeting encapsulated anticancer agent to estrogen receptor positive breast cancer cells: In vitro and in vivo evaluation. Biomed. Pharmacother. 2014, 68, 429–438.
  17. Lu, R.-M.; Chen, M.-S.; Chang, D.-K.; Chiu, C.-Y.; Lin, W.-C.; Yan, S.-L.; Wang, Y.-P.; Kuo, Y.-S.; Yeh, C.-Y.; Lo, A.; et al. Targeted Drug Delivery Systems Mediated by a Novel Peptide in Breast Cancer Therapy and Imaging. PLoS ONE 2013, 8, e66128.
  18. Hosokawa, S.; Tagawa, T.; Niki, H.; Hirakawa, Y.; Nohga, K.; Nagaike, K. Efficacy of immunoliposomes on cancer models in a cell-surface-antigen-density-dependent manner. Br. J. Cancer 2003, 89, 1545–1551.
  19. Nowsheen, S.; Cooper, T.; Stanley, J.A.; Yang, E.S. Synthetic Lethal Interactions between EGFR and PARP Inhibition in Human Triple Negative Breast Cancer Cells. PLoS ONE 2012, 7, e46614.
  20. Brand, T.M.; Iida, M.; Dunn, E.F.; Luthar, N.; Kostopoulos, K.T.; Corrigan, K.L.; Wleklinski, M.J.; Yang, D.; Wisinski, K.B.; Salgia, R.; et al. Nuclear Epidermal Growth Factor Receptor Is a Functional Molecular Target in Triple-Negative Breast Cancer. Mol. Cancer Ther. 2014, 13, 1356–1368.
  21. Zakaria, Z.; Zulkifle, M.F.; Hasan, W.A.N.W.; Azhari, A.K.; Raub, S.H.A.; Eswaran, J.; Soundararajan, M.; Husain, S.N.A.S. Epidermal growth factor receptor (EGFR) gene alteration and protein overexpression in Malaysian triple-negative breast cancer (TNBC) cohort. OncoTargets Ther. 2019, 12, 7749–7756.
  22. Song, X.; Liu, Z.; Yu, Z. EGFR Promotes the Development of Triple Negative Breast Cancer Through JAK/STAT3 Signaling. Cancer Manag. Res. 2020, 12, 703–717.
  23. Ali, R.; Wendt, M.K. The paradoxical functions of EGFR during breast cancer progression. Signal Transduct. Target. Ther. 2017, 2, 16042.
  24. Masuda, H.; Zhang, D.; Bartholomeusz, C.; Doihara, H.; Hortobagyi, G.N.; Ueno, N.T. Role of epidermal growth factor receptor in breast cancer. Breast Cancer Res. Treat. 2012, 136, 331–345.
  25. Nakai, K.; Hung, M.-C.; Yamaguchi, H. A perspective on anti-EGFR therapies targeting triple-negative breast cancer. Am. J. Cancer Res. 2016, 6, 1609–1623.
  26. Bethune, G.; Bethune, E.; Ridgway, N.; Xu, Z. Epidermal growth factor receptor (EGFR) in lung cancer: An overview and update. J. Thorac. Dis. 2010, 2, 48–51.
  27. Charakidis, M.; Boyer, M. Targeting MET and EGFR in NSCLC—what can we learn from the recently reported phase III trial of onartuzumab in combination with erlotinib in advanced non-small cell lung cancer? Transl. Lung Cancer Res. 2014, 3, 395–396.
  28. Nielsen, T.O.; Hsu, F.D.; Jensen, K.; Cheang, M.; Karaca, G.; Hu, Z.; Hernandez-Boussard, T.; Livasy, C.; Cowan, D.; Dressler, L.; et al. Immunohistochemical and Clinical Characterization of the Basal-Like Subtype of Invasive Breast Carcinoma. Clin. Cancer Res. 2004, 10, 5367–5374.
  29. Nicholson, R.; Gee, J.; Harper, M. EGFR and cancer prognosis. Eur. J. Cancer 2001, 37, 9–15.
  30. A Changavi, A.; Shashikala, A.; Ramji, A.S. Epidermal Growth Factor Receptor Expression in Triple Negative and Nontriple Negative Breast Carcinomas. J. Lab. Physicians 2015, 7, 079–083.
  31. Xu, C.; Li, X.; Liu, P.; Li, M.; Luo, F. Patient-derived xenograft mouse models: A high fidelity tool for individualized medicine (Review). Oncol. Lett. 2018, 17, 3–10.
  32. Denard, B.; Jiang, S.; Peng, Y.; Ye, J. CREB3L1 as a potential biomarker predicting response of triple negative breast cancer to doxorubicin-based chemotherapy. BMC Cancer 2018, 18, 813.
  33. Nisini, R.; Poerio, N.; Mariotti, S.; De Santis, F.; Fraziano, M. The Multirole of Liposomes in Therapy and Prevention of Infectious Diseases. Front. Immunol. 2018, 9, 155.
  34. Bao, X.; Zeng, J.; Huang, H.; Ma, C.; Wang, L.; Wang, F.; Liao, X.; Song, X. Cancer-targeted PEDF-DNA therapy for metastatic colorectal cancer. Int. J. Pharm. 2020, 576, 118999.
  35. Garbuzenko, O.B.; Kuzmov, A.; Taratula, O.; Pine, S.R.; Minko, T. Strategy to enhance lung cancer treatment by five essential elements: Inhalation delivery, nanotechnology, tumor-receptor targeting, chemo- and gene therapy. Theranostics 2019, 9, 8362–8376.
  36. Samaddar, S.; Mazur, J.; Boehm, D.; Thompson, D.H. Development And In Vitro Characterization Of Bladder Tumor Cell Targeted Lipid-Coated Polyplex For Dual Delivery Of Plasmids And Small Molecules. Int. J. Nanomed. 2019, 14, 9547–9561.
  37. Fan, J.; Liu, Y.; Liu, L.; Huang, Y.; Li, X.; Huang, W. A Multifunction Lipid-Based CRISPR-Cas13a Genetic Circuit Delivery System for Bladder Cancer Gene Therapy. ACS Synth. Biol. 2019, 9, 343–355.
  38. Itani, R.; Al Faraj, A. siRNA Conjugated Nanoparticles—A Next Generation Strategy to Treat Lung Cancer. Int. J. Mol. Sci. 2019, 20, 6088.
  39. Mizuno, M.; Yoshida, J. Improvement of Transduction Efficiency of Recombinant Adeno-associated Virus Vector by Entrapment in Multilamellar Liposomes. Jpn. J. Cancer Res. 1998, 89, 352–354.
  40. Vieweg, J.; Boczkowski, D.; Roberson, K.M.; Edwards, D.W.; Philip, M.; Philip, R.; Rudoll, T.; Smith, C.; Robertson, C.; Gilboa, E. Efficient gene transfer with adeno-associated virus-based plasmids complexed to cationic liposomes for gene therapy of human prostate cancer. Cancer Res. 1995, 55, 2366–2372.
  41. Lins-Austin, B.; Patel, S.; Mietzsch, M.; Brooke, D.; Bennett, A.; Venkatakrishnan, B.; Van Vliet, K.; Smith, A.N.; Long, J.R.; McKenna, R.; et al. Adeno-Associated Virus (AAV) Capsid Stability and Liposome Remodeling During Endo/Lysosomal pH Trafficking. Viruses 2020, 12, 668.
  42. Tang, X.; Mohuczy, D.; Zhang, Y.C.; Kimura, B.; Galli, S.M.; Phillips, M.I. Intravenous angiotensinogen antisense in AAV-based vector decreases hypertension. Am. J. Physiol. Content 1999, 277, H2392–H2399.
  43. Elsana, H.; Olusanya, T.O.B.; Carr-Wilkinson, J.; Darby, S.; Faheem, A.; Elkordy, A.A. Evaluation of novel cationic gene based liposomes with cyclodextrin prepared by thin film hydration and microfluidic systems. Sci. Rep. 2019, 9, 1–12.
  44. Gjetting, T.; Andresen, T.L.; Christensen, C.L.; Cramer, F.; Poulsen, T.T.; Poulsen, H.S. A simple protocol for preparation of a liposomal vesicle with encapsulated plasmid DNA that mediate high accumulation and reporter gene activity in tumor tissue. Results Pharma Sci. 2011, 1, 49–56.
  45. Saffari, M.; Moghimi, H.R.; Dass, C.R. Barriers to Liposomal Gene Delivery: From Application Site to the Target. Iran. J. Pharm. Res. IJPR 2016, 15, 3–17.
  46. Zylberberg, C.; Gaskill, K.; Pasley, S.; Matosevic, S. Engineering liposomal nanoparticles for targeted gene therapy. Gene Ther. 2017, 24, 441–452.
  47. Simões, S.; Filipe, A.C.D.S.; Faneca, H.; Mano, M.; Penacho, N.; Düzgünes, N.; de Lima, M.P. Cationic liposomes for gene delivery. Expert Opin. Drug Deliv. 2005, 2, 237–254.
  48. Hattori, Y.; Suzuki, S.; Kawakami, S.; Yamashita, F.; Hashida, M. The role of dioleoylphosphatidylethanolamine (DOPE) in targeted gene delivery with mannosylated cationic liposomes via intravenous route. J. Control. Release 2005, 108, 484–495.
  49. Sung, Y.K.; Kim, S.W. Recent advances in the development of gene delivery systems. Biomater. Res. 2019, 23, 1–7.
  50. Alton, E.W.F.W.; Armstrong, D.K.; Ashby, D.; Bayfield, K.J.; Bilton, D.; Bloomfield, E.V.; Boyd, A.C.; Brand, J.; Buchan, R.; Calcedo, R.; et al. Repeated nebulisation of non-viral CFTR gene therapy in patients with cystic fibrosis: A randomised, double-blind, placebo-controlled, phase 2b trial. Lancet Respir. Med. 2015, 3, 684–691.
  51. Werner-Wasik, M.; Langer, C.; Movsas, B. Randomized phase II study of amifostine mucosal protection by either subcutaneous injection or rapid IV bolus for patients with inoperable stage II–IIIA/B or stage IV non-small cell lung cancer with oligometastases receiving concurrent radiochemotherapy with carboplatin and paclitaxel followed by optional consolidative chemotherapy: A follow-up study after RTOG 98-01. Semin. Oncol. 2004, 31, 47–51.
  52. Stopeck, A.T.; Jones, A.; Hersh, E.M.; A Thompson, J.; Finucane, D.M.; Gutheil, J.C.; Gonzalez, R. Phase II study of direct intralesional gene transfer of allovectin-7, an HLA-B7/beta2-microglobulin DNA-liposome complex, in patients with metastatic melanoma. Clin. Cancer Res. 2001, 7, 2285–2291.
  53. Bergen, M.; Chen, R.; Gonzalez, R. Efficacy and safety of HLA-B7/β-2 microglobulin plasmid DNA/lipid complex (Allovectin-7®) in patients with metastatic melanoma. Expert Opin. Biol. Ther. 2003, 3, 377–384.
  54. Thaker, P.H.; Brady, W.E.; Lankes, H.A.; Odunsi, K.; Bradley, W.H.; Moore, K.N.; Muller, C.Y.; Anwer, K.; Schilder, R.J.; Alvarez, R.D.; et al. A phase I trial of intraperitoneal GEN-1, an IL-12 plasmid formulated with PEG-PEI-cholesterol lipopolymer, administered with pegylated liposomal doxorubicin in patients with recurrent or persistent epithelial ovarian, fallopian tube or primary peritoneal cancers: An NRG Oncology/Gynecologic Oncology Group study. Gynecol. Oncol. 2017, 147, 283–290.
  55. Atalay, G.; Cardoso, F.; Awada, A.; Piccart, M.J. Novel therapeutic strategies targeting the epidermal growth factor receptor (EGFR) family and its downstream effectors in breast cancer. Ann. Oncol. 2003, 14, 1346–1363.
  56. Cheung, A.; Opzoomer, J.; Ilieva, K.M.; Gazinska, P.; Hoffmann, R.M.; Mirza, H.; Marlow, R.; Francesch-Domenech, E.; Fittall, M.; Rodriguez, D.D.; et al. Anti-Folate Receptor Alpha–Directed Antibody Therapies Restrict the Growth of Triple-negative Breast Cancer. Clin. Cancer Res. 2018, 24, 5098–5111.
  57. Perez, E.A. Treatment strategies for advanced hormone receptor-positive and human epidermal growth factor 2-negative breast cancer: The role of treatment order. Drug Resist. Updat. 2016, 24, 13–22.
  58. Hossein-Nejad-Ariani, H.; AlThagafi, E.; Kaur, K. Small Peptide Ligands for Targeting EGFR in Triple Negative Breast Cancer Cells. Sci. Rep. 2019, 9, 1–10.
  59. Fitzpatrick, S.L.; Lachance, M.P.; Schultz, G.S. Characterization of epidermal growth factor receptor and action on human breast cancer cells in culture. Cancer Res. 1984, 44, 3442–3447.
  60. Flynn, J.F.; Wong, C.; Wu, J.M. Anti-EGFR Therapy: Mechanism and Advances in Clinical Efficacy in Breast Cancer. J. Oncol. 2009, 2009, 1–16.
  61. Chen, J.; He, H.; Deng, C.; Yin, L.; Zhong, Z. Saporin-loaded CD44 and EGFR dual-targeted nanogels for potent inhibition of metastatic breast cancer in vivo. Int. J. Pharm. 2019, 560, 57–64.
  62. Garrido, G.; Rabasa, A.; Garrido, C.; Chao, L.; Garrido, F.; Lora, A.M.G.; Sánchez-Ramírez, B. Upregulation of HLA Class I Expression on Tumor Cells by the Anti-EGFR Antibody Nimotuzumab. Front. Pharmacol. 2017, 8, 595.
  63. Yang, J.; Liao, D.; Chen, C.; Liu, Y.; Chuang, T.-H.; Xiang, R.; Markowitz, D.; Reisfeld, R.A.; Luo, Y. Tumor-Associated Macrophages Regulate Murine Breast Cancer Stem Cells Through a Novel Paracrine EGFR/Stat3/Sox-2 Signaling Pathway. Stem Cells 2012, 31, 248–258.
  64. Bonner, J.A.; Harari, P.M.; Giralt, J.; Azarnia, N.; Shin, D.M.; Cohen, R.B.; Jones, C.U.; Sur, R.; Raben, D.; Jassem, J.; et al. Radiotherapy plus Cetuximab for Squamous-Cell Carcinoma of the Head and Neck. N. Engl. J. Med. 2006, 354, 567–578.
  65. Leung, H.W.; Lang, H.-C.; Wang, S.-Y.; Leung, J.H.; Chan, A.L. Cost-utility analysis of stereotactic body radiotherapy plus cetuximab in previously irradiated recurrent squamous cell carcinoma of the head and neck. Expert Rev. Pharmacoeconomics Outcomes Res. 2021, 21, 489–495.
  66. Vermorken, J.B.; Stöhlmacher-Williams, J.; Davidenko, I.; Licitra, L.; Winquist, E.; Villanueva, C.; Foa, P.; Rottey, S.; Składowski, K.; Tahara, M.; et al. Cisplatin and fluorouracil with or without panitumumab in patients with recurrent or metastatic squamous-cell carcinoma of the head and neck (SPECTRUM): An open-label phase 3 randomised trial. Lancet Oncol. 2013, 14, 697–710.
  67. Guren, T.K.; Thomsen, M.; Kure, E.H.; Sorbye, H.; Glimelius, B.; Pfeiffer, P.; Österlund, P.; Sigurdsson, F.; Lothe, I.M.B.; Dalsgaard, A.M.; et al. Cetuximab in treatment of metastatic colorectal cancer: Final survival analyses and extended RAS data from the NORDIC-VII study. Br. J. Cancer 2017, 116, 1271–1278.
  68. Hwang, S.-Y.; Park, S.; Kwon, Y. Recent therapeutic trends and promising targets in triple negative breast cancer. Pharmacol. Ther. 2019, 199, 30–57.
  69. Jonker, D.J.; O’Callaghan, C.J.; Karapetis, C.; Zalcberg, J.R.; Tu, D.; Au, H.-J.; Berry, S.R.; Krahn, M.; Price, T.; Simes, R.J.; et al. Cetuximab for the Treatment of Colorectal Cancer. N. Engl. J. Med. 2007, 357, 2040–2048.
  70. Ferris, R.L.; Lenz, H.-J.; Trotta, A.M.; García-Foncillas, J.; Schulten, J.; Audhuy, F.; Merlano, M.; Milano, G. Rationale for combination of therapeutic antibodies targeting tumor cells and immune checkpoint receptors: Harnessing innate and adaptive immunity through IgG1 isotype immune effector stimulation. Cancer Treat. Rev. 2018, 63, 48–60.
More
Information
Subjects: Oncology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 327
Revision: 1 time (View History)
Update Date: 25 Aug 2021
1000/1000