We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Functionalized nano-targeted moieties in management of prostate cancer

    Taher A Salaheldin

    The Pharmaceutical Research Institute, Albany College of Pharmacy & Health Sciences, 1 Discovery Drive, Rensselaer, NY 12144 USA

    ,
    Dhruba J Bharali

    The Pharmaceutical Research Institute, Albany College of Pharmacy & Health Sciences, 1 Discovery Drive, Rensselaer, NY 12144 USA

    &
    Shaker A Mousa

    *Author for correspondence: Tel.: +1 518 694 7397; Fax: +1 518 694 7567;

    E-mail Address: shaker.mousa@acphs.edu

    The Pharmaceutical Research Institute, Albany College of Pharmacy & Health Sciences, 1 Discovery Drive, Rensselaer, NY 12144 USA

    Published Online:https://doi.org/10.2217/fon-2019-0635

    Multimodal properties of nanoparticles, such as simultaneously carrying drugs and/or diagnostic probes for site-specific delivery, make them excellent carriers for diagnosis and treatment of prostate cancer. Advantages are high permeability and selectivity to malignant cells to reduce systemic toxicity of chemotherapeutic drugs. Based on a review of current literature, the lack of efficient and highly specific prostate cancer cell targeting moieties is hindering successful in vivo prostate cancer-targeted drug delivery systems. Highly specific nano-targeting moieties as drug delivery vehicles might improve chemotherapeutic delivery via targeting to specific receptors expressed on the surface of prostate cancer cells. This review describes nano-targeting moieties for management of prostate cancer and its cancer stem cells. Descriptions of targeting moieties using anti-prostate-specific membrane antigen, aptamer, anti-cluster of differentiation 24/44, folic acid and other targeting strategies are highlighted. Current research results are promising and may yield development of next-generation nanoscale theragnostic targeted modalities for prostate cancer treatment.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Crawford ED, Higano CS, Shore ND, Hussain M, Petrylak DP. Treating patients with metastatic castration resistant prostate cancer: a comprehensive review of available therapies. J. Urol. 194(6), 1537–1547 (2015).
    • 2. Hamdy FC, Donovan JL, Lane JA et al. 10-year outcomes after monitoring, surgery, or radiotherapy for localized prostate cancer. N. Engl. J. Med. 375(15), 1415–1424 (2016).
    • 3. Barocas DA, Salem S, Kordan Y et al. Robotic assisted laparoscopic prostatectomy versus radical retropubic prostatectomy for clinically localized prostate cancer: comparison of short-term biochemical recurrence-free survival. J. Urol. 183(3), 990–996 (2010).
    • 4. Eandi JA, Link BA, Nelson RA et al. Robotic assisted laparoscopic salvage prostatectomy for radiation resistant prostate cancer. J. Urol. 183(1), 133–137 (2010).
    • 5. Rashid HH, Leung YY, Rashid MJ, Oleyourryk G, Valvo JR, Eichel L. Robotic surgical education: a systematic approach to training urology residents to perform robotic-assisted laparoscopic radical prostatectomy. Urology 68(1), 75–79 (2006).
    • 6. Abu-Gheida I, Reddy CA, Kotecha R et al. Ten-year outcomes of moderately hypofractionated (70 Gy in 28 fractions) intensity modulated radiation therapy for localized prostate cancer. Int. J. Radiat. Oncol. Biol. Phys. 104(2), 325–333 (2019).
    • 7. Tanaka H, Nakashima Y, Ito M et al. Intensity-modulated radiation therapy for elderly patients (aged ≥75 years) with localized prostate cancer: comparison with younger patients (aged <75 years). Mol. Clin. Oncol. 10(4), 476–480 (2019). • Clinical study for nanotargeted radiation therapy on patients with prostate cancer taking age into considration.
    • 8. Corn PG, Zhang M, Nogueras-Gonzalez GM et al. A Phase II study of cabozantinib and androgen ablation in patients with hormone-naïve metastatic prostate cancer. Clin. Cancer Res. 26(5), 990–999 (2020).
    • 9. Brawley OW. Trends in prostate cancer in the United States. J. Natl Cancer Inst. Monogr. 2012(45), 152–156 (2012).
    • 10. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J. Clin. 69(1), 7–34 (2019).
    • 11. Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ. Cancer statistics, 2009. CA Cancer J. Clin. 59(4), 225–249 (2009).
    • 12. Antonarakis ES, Lu C, Luber B et al. Androgen receptor splice variant 7 and efficacy of taxane chemotherapy in patients with metastatic castration-resistant prostate cancer. JAMA Oncol. 1(5), 582–591 (2015).
    • 13. Azad AA, Eigl BJ, Murray RN, Kollmannsberger C, Chi KN. Efficacy of enzalutamide following abiraterone acetate in chemotherapy-naive metastatic castration-resistant prostate cancer patients. Eur. Urol. 67(1), 23–29 (2015).
    • 14. Wang Q, Zhang X, Sun Y et al. Gold-caged copolymer nanoparticles as multimodal synergistic photodynamic/photothermal/chemotherapy platform against lethality androgen-resistant prostate cancer. Biomaterials 212, 73–86 (2019).
    • 15. Vandghanooni S, Eskandani M, Barar J, Omidi Y. Recent advances in aptamer-armed multimodal theranostic nanosystems for imaging and targeted therapy of cancer. Eur. J. Pharm. Sci. 117, 301–312 (2018).
    • 16. Du D, Fu HJ, Ren WW, Li XL, Guo LH. PSA targeted dual-modality manganese oxide-mesoporous silica nanoparticles for prostate cancer imaging. Biomed. Pharmacother. 121, 109614 (2020). • Mesoporous silica nanoparticles used as vehical for dual functional nano-targeting system for diagnosis and treatment of prostate cancer.
    • 17. Jang B, Kwon H, Katila P, Lee SJ, Lee H. Dual delivery of biological therapeutics for multimodal and synergistic cancer therapies. Adv. Drug Deliv. Rev. 98, 113–133 (2016).
    • 18. Eiber M, Fendler WP, Rowe SP et al. Prostate-specific membrane antigen ligands for imaging and therapy. J. Nucl. Med. 58(Suppl. 2), S67–S76 (2017).
    • 19. Maurer T, Eiber M, Schwaiger M, Gschwend JE. Current use of PSMA-PET in prostate cancer management. Nat. Rev. Urol. 13(4), 226–235 (2016).
    • 20. Junghans RP, Ma Q, Rathore R et al. Phase I trial of anti-PSMA designer CAR-T cells in prostate cancer: possible role for interacting interleukin 2-T cell pharmacodynamics as a determinant of clinical response. Prostate 76(14), 1257–1270 (2016). •• Phase I trial demonstrating significant bioactive targeting of anti-PSMA and its role for interacting interleukin 2-T cell.
    • 21. Pandit-Taskar N, O’Donoghue JA, Ruan S et al. First-in-human imaging with 89Zr-Df-IAB2M anti-PSMA minibody in patients with metastatic prostate cancer: pharmacokinetics, biodistribution, dosimetry, and lesion uptake. J. Nucl. Med. 57(12), 1858–1864 (2016).
    • 22. Fan XZ, Guo YL, Wang LF, Xiong XY, Zhu LH, Fang KJ. Diagnosis of prostate cancer using anti-PSMA aptamer A10-3.2-oriented lipid nanobubbles. Int. J. Nanomed. 11, 3939–3950 (2016).
    • 23. Barve A, Jin W, Cheng K. Prostate cancer relevant antigens and enzymes for targeted drug delivery. J. Control. Rel. 187, 118–132 (2014).
    • 24. Haberkorn U, Eder M, Kopka K, Babich JW, Eisenhut M. New strategies in prostate cancer: prostate-specific membrane antigen (PSMA) ligands for diagnosis and therapy. Clin. Cancer Res. 22(1), 9–15 (2016).
    • 25. Perera M, Papa N, Roberts M et al. Gallium-68 prostate-specific membrane antigen positron emission tomography in advanced prostate cancer-updated diagnostic utility, sensitivity, specificity, and distribution of prostate-specific membrane antigen-avid lesions: a systematic review and meta-analysis. Eur. Urol. 77(4), 403–417 (2019).
    • 26. Mease RC, Foss CA, Pomper MG. PET imaging in prostate cancer: focus on prostate-specific membrane antigen. Curr. Top. Med. Chem. 13(8), 951–962 (2013).
    • 27. Derks YHW, Lowik D, Sedelaar JPM et al. PSMA-targeting agents for radio- and fluorescence-guided prostate cancer surgery. Theranostics. 9(23), 6824–6839 (2019).
    • 28. Tse BW, Cowin GJ, Soekmadji C et al. PSMA-targeting iron oxide magnetic nanoparticles enhance MRI of preclinical prostate cancer. Nanomedicine 10(3), 375–386 (2015).
    • 29. Mukherjee A, Darlington T, Baldwin R et al. Development and screening of a series of antibody-conjugated and silica-coated iron oxide nanoparticles for targeting the prostate-specific membrane antigen. ChemMedChem 9(7), 1356–1360 (2014).
    • 30. Hariri W, Sudha T, Bharali DJ, Cui H, Mousa SA. Nano-targeted delivery of toremifene, an estrogen receptor-α blocker in prostate cancer. Pharm. Res. 32(8), 2764–2774 (2015). •• The use of estrogen receptor α blocker targeting moiety for delivery of toremifene-loaded poly(lactide-co-glycolide)-PSMA nano delivery system in prostate cancer model.
    • 31. Wong P, Li L, Chea J et al. PET imaging of 64Cu-DOTA-scFv-anti-PSMA lipid nanoparticles (LNPs): enhanced tumor targeting over anti-PSMA scFv or untargeted LNPs. Nucl. Med. Biol. 47, 62–68 (2017).
    • 32. Mangadlao JD, Wang X, McCleese C et al. Prostate-specific membrane antigen targeted gold nanoparticles for theranostics of prostate cancer. ACS Nano 12(4), 3714–3725 (2018). •• Multifuctional nanotheranostic agent based on PSMA-gold nanoparticles for prostate cancer imaging and photodynamic therapy.
    • 33. Barrio M, Fendler WP, Czernin J, Herrmann K. Prostate specific membrane antigen (PSMA) ligands for diagnosis and therapy of prostate cancer. Expert Rev. Mol. Diagn. 16(11), 1177–1188 (2016).
    • 34. Rahbar K, Afshar-Oromieh A, Jadvar H, Ahmadzadehfar H. PSMA theranostics: current status and future directions. Mol. Imaging 17, 1536012118776068 (2018).
    • 35. Sanna V, Singh CK, Jashari R et al. Targeted nanoparticles encapsulating (-)-epigallocatechin-3-gallate for prostate cancer prevention and therapy. Sci. Rep. 7, 41573 (2017). • Comprehensive in vitro evaluation of the antiproliferative activity in prostate carcinoma of nano-encapsulated epigallocathechin-3-gallate conjugated with anti-PSMA antibody.
    • 36. Sun SB, Liu P, Shao FM, Miao QL. Formulation and evaluation of PLGA nanoparticles loaded capecitabine for prostate cancer. Int. J. Clin. Exp. Med. 8(10), 19670–19681 (2015).
    • 37. Cao LB, Zeng S, Zhao W. Highly stable PEGylated poly(lactic-co-glycolic acid) (PLGA) nanoparticles for the effective delivery of docetaxel in prostate cancers. Nanoscale Res. Lett. 11(1), 305 (2016).
    • 38. Yu H, Tang Z, Li M et al. Cisplatin loaded loly(L-glutamic acid)-g-methoxy poly(ethylene glycol) complex nanoparticles for potential cancer therapy: preparation, in vitro and in vivo evaluation. J. Biomed. Nanotechnol. 12(1), 69–78 (2016).
    • 39. Alam N, Koul M, Mintoo MJ et al. Development and characterization of hyaluronic acid modified PLGA based nanoparticles for improved efficacy of cisplatin in solid tumor. Biomed. Pharmacother. 95, 856–864 (2017).
    • 40. Azandeh SS, Abbaspour M, Khodadadi A, Khorsandi L, Orazizadeh M, Heidari-Moghadam A. Anticancer activity of curcumin-loaded PLGA nanoparticles on PC3 prostate cancer cells. Iran. J. Pharm. Res. 16(3), 868–879 (2017).
    • 41. Pan M, Li W, Yang J et al. Plumbagin-loaded aptamer-targeted poly D,L-lactic-co-glycolic acid-b-polyethylene glycol nanoparticles for prostate cancer therapy. Medicine (Baltimore) 96(30), e7405 (2017).
    • 42. Sharma S, Parmar A, Kori S, Sandhir R. PLGA-based nanoparticles: a new paradigm in biomedical applications. Trends Anal. Chem. 80, 30–40 (2016).
    • 43. Cho HS, Dong Z, Pauletti GM et al. Fluorescent, superparamagnetic nanospheres for drug storage, targeting, and imaging: a multifunctional nanocarrier system for cancer diagnosis and treatment. ACS Nano 4(9), 5398–5404 (2010).
    • 44. Gao X, Luo Y, Wang Y et al. Prostate stem cell antigen-targeted nanoparticles with dual functional properties: in vivo imaging and cancer chemotherapy. Int. J. Nanomedicine 7, 4037–4051 (2012).
    • 45. Yallapu MM, Khan S, Maher DM et al. Anti-cancer activity of curcumin loaded nanoparticles in prostate cancer. Biomaterials 35(30), 8635–8648 (2014). • Natural curcumin polymeric nanoparticles as a biocompatible nano-targeted drug delivery for in vivo anticancer model.
    • 46. Mi J, Liu Y, Rabbani ZN et al. In vivo selection of tumor-targeting RNA motifs. Nat. Chem. Biol. 6(1), 22–24 (2010).
    • 47. Xuan WJ, Peng YB, Deng ZY et al. A basic insight into aptamer-drug conjugates (ApDCs). Biomaterials 182, 216–226 (2018).
    • 48. Bruno JG. A review of therapeutic aptamer conjugates with emphasis on new approaches. Pharmaceuticals 6(3), 340–357 (2013).
    • 49. Chen Z, Tai Z, Gu F, Hu C, Zhu Q, Gao S. Aptamer-mediated delivery of docetaxel to prostate cancer through polymeric nanoparticles for enhancement of antitumor efficacy. Eur. J. Pharm. Biopharm. 107, 130–141 (2016).
    • 50. Jiao J, Zou Q, Zou MH, Guo RM, Zhu S, Zhang Y. Aptamer-modified PLGA nanoparticle delivery of triplex forming oligonucleotide for targeted prostate cancer therapy. Neoplasma 63(4), 569–575 (2016).
    • 51. Leach JC, Wang A, Ye K, Jin S. A RNA-DNA hybrid aptamer for nanoparticle-based prostate tumor targeted drug delivery. Int. J. Mol. Sci. 17(3), 380 (2016).
    • 52. Tang L, Tong R, Coyle VJ et al. Targeting tumor vasculature with aptamer-functionalized doxorubicin-polylactide nanoconjugates for enhanced cancer therapy. ACS Nano 9(5), 5072–5081 (2015).
    • 53. Lin Z, Ma Q, Fei X, Zhang H, Su X. A novel aptamer functionalized CuInS2 quantum dots probe for daunorubicin sensing and near infrared imaging of prostate cancer cells. Anal. Chim. Acta 818, 54–60 (2014).
    • 54. Zhao Y, Duan S, Zeng X et al. Prodrug strategy for PSMA-targeted delivery of TGX-221 to prostate cancer cells. Mol. Pharm. 9(6), 1705–1716 (2012).
    • 55. Yu MK, Kim D, Lee IH, So JS, Jeong YY, Jon S. Image-guided prostate cancer therapy using aptamer-functionalized thermally cross-linked superparamagnetic iron oxide nanoparticles. Small 7(15), 2241–2249 (2011).
    • 56. Wu X, Tai Z, Zhu Q et al. Study on the prostate cancer-targeting mechanism of aptamer-modified nanoparticles and their potential anticancer effect in vivo. Int. J. Nanomedicine 9, 5431 (2014).
    • 57. Baek SE, Lee KH, Park YS et al. RNA aptamer-conjugated liposome as an efficient anticancer drug delivery vehicle targeting cancer cells in vivo. J. Control. Rel. 196, 234–242 (2014).
    • 58. Yang J, Xie SX, Huang Y et al. Prostate-targeted biodegradable nanoparticles loaded with androgen receptor silencing constructs eradicate xenograft tumors in mice. Nanomedicine 7(9), 1297–1309 (2012).
    • 59. Gu FF, Hu CL, Xia QM, Gong CA, Gao S, Chen ZJ. Aptamer-conjugated multi-walled carbon nanotubes as a new targeted ultrasound contrast agent for the diagnosis of prostate cancer. J. Nanopart. Res. 20(11), 303–312 (2018). • Multiwalled carbon nanotubes increase the contrast of ultrasound imaging of localized prostate cancer.
    • 60. Zhen S, Takahashi Y, Narita S, Yang YC, Li X. Targeted delivery of CRISPR/Cas9 to prostate cancer by modified gRNA using a flexible aptamer-cationic liposome. Oncotarget 8(6), 9375–9387 (2017).
    • 61. Fazel-Ghaziyani M, Shahbazi-Gahrouei D, Pourhassan-Moghaddam M, Baradaran B, Ghavami M. Targeted detection of the cancer cells using the anti-CD24 bio modified PEGylated gold nanoparticles: the application of CD24 as a vital cancer biomarker. Nanomed. J. 5(3), 172–179 (2018).
    • 62. Zununi Vahed S, Fathi N, Samiei M, Maleki Dizaj S, Sharifi S. Targeted cancer drug delivery with aptamer-functionalized polymeric nanoparticles. J. Drug Target. 27(3), 292–299 (2019).
    • 63. Zhang YF, Li BJ, Zhang XY et al. CD24 is a genetic modifier for risk and progression of prostate cancer. Mol. Carcinogen. 56(2), 641–650 (2017).
    • 64. Bharali DJ, Sudha T, Cui H, Mian BM, Mousa SA. Anti-CD24 nano-targeted delivery of docetaxel for the treatment of prostate cancer. Nanomedicine 13(1), 263–273 (2017).
    • 65. Huang WY, Lin JN, Hsieh JT et al. Nanoparticle targeting CD44-positive cancer cells for site-specific drug delivery in prostate cancer therapy. ACS Appl. Mater. Interfaces 8(45), 30722–30734 (2016).
    • 66. Spadea A, Rios de la Rosa JM, Tirella A et al. Evaluating the efficiency of hyaluronic acid for tumor targeting via CD44. Mol. Pharm. 16(6), 2481–2493 (2019).
    • 67. Mahira S, Kommineni N, Husain GM, Khan W. Cabazitaxel and silibinin co-encapsulated cationic liposomes for CD44 targeted delivery: a new insight into nanomedicine based combinational chemotherapy for prostate cancer. Biomed. Pharmacother. 110, 803–817 (2019).
    • 68. Wei J, Sun J, Liu Y. Enhanced targeting of prostate cancer-initiating cells by salinomycin-encapsulated lipid-PLGA nanoparticles linked with CD44 antibodies. Oncol. Lett. 17(4), 4024–4033 (2019).
    • 69. Cheung A, Bax HJ, Josephs DH et al. Targeting folate receptor alpha for cancer treatment. Oncotarget 7(32), 52553–52574 (2016).
    • 70. Assaraf YG, Leamon CP, Reddy JA. The folate receptor as a rational therapeutic target for personalized cancer treatment. Drug Resist. Updates 17(4–6), 89–95 (2014).
    • 71. Evans JC, Malhotra M, Guo J et al. Folate-targeted amphiphilic cyclodextrin.siRNA nanoparticles for prostate cancer therapy exhibit PSMA mediated uptake, therapeutic gene silencing in vitro and prolonged circulation in vivo. Nanomedicine 12(8), 2341–2351 (2016).
    • 72. Khoeeniha MK, Esfandyari-Manesh M, Behrouz H et al. Targeted delivery of cabazitaxel by conjugation to albumin-PEG-folate nanoparticles using a cysteine-acrylate linker and simple synthesis conditions. Curr. Drug Deliv. 14(8), 1120–1129 (2017).
    • 73. Choi KH, Nam KC, Malkinski L, Choi EH, Jung JS, Park BJ. Size-dependent photodynamic anticancer activity of biocompatible multifunctional magnetic submicron particles in prostate cancer cells. Molecules 21(9), (2016). doi.org/10.3390/molecules21091187
    • 74. Guo J, O’Driscoll CM, Holmes JD, Rahme K. Bioconjugated gold nanoparticles enhance cellular uptake: A proof of concept study for siRNA delivery in prostate cancer cells. Int. J. Pharm. 509(1–2), 16–27 (2016).
    • 75. de Oliveira LF, Bouchmella K, Goncalves Kde A, Bettini J, Kobarg J, Cardoso MB. Functionalized silica nanoparticles as an alternative platform for targeted drug-delivery of water insoluble drugs. Langmuir 32(13), 3217–3225 (2016).
    • 76. Au KM, Satterlee A, Min Y et al. Folate-targeted pH-responsive calcium zoledronate nanoscale metal-organic frameworks: turning a bone antiresorptive agent into an anticancer therapeutic. Biomaterials 82, 178–193 (2016).
    • 77. Shen Y, Ma Z, Chen F et al. Effective photothermal chemotherapy with docetaxel-loaded gold nanospheres in advanced prostate cancer. J. Drug Target. 23(6), 568–576 (2015).
    • 78. El-Gogary RI, Rubio N, Wang JT et al. Polyethylene glycol conjugated polymeric nanocapsules for targeted delivery of quercetin to folate-expressing cancer cells in vitro and in vivo. ACS Nano 8(2), 1384–1401 (2014).
    • 79. Wittig R, Rosenholm JM, von Haartman E et al. Active targeting of mesoporous silica drug carriers enhances gamma-secretase inhibitor efficacy in an in vivo model for breast cancer. Nanomedicine 9(7), 971–987 (2014).
    • 80. Evans JC, Malhotra M, Sweeney K et al. Folate-targeted amphiphilic cyclodextrin nanoparticles incorporating a fusogenic peptide deliver therapeutic siRNA and inhibit the invasive capacity of 3D prostate cancer tumours. Int. J. Pharm. 532(1), 511–518 (2017).
    • 81. Singh R, Kesharwani P, Mehra NK, Singh S, Banerjee S, Jain NK. Development and characterization of folate anchored Saquinavir entrapped PLGA nanoparticles for anti-tumor activity. Drug Dev. Ind. Pharm. 41(11), 1888–1901 (2015).
    • 82. Barile M, Giancaspero TA, Leone P, Galluccio M, Indiveri C. Riboflavin transport and metabolism in humans. J. Inherit. Metab. Dis. 39(4), 545–557 (2016).
    • 83. Jayapaul J, Arns S, Bunker M et al. In vivo evaluation of riboflavin receptor targeted fluorescent USPIO in mice with prostate cancer xenografts. Nano Res. 9(5), 1319–1333 (2016).
    • 84. Wu C-Y, Chen Y-C. Riboflavin immobilized Fe3O4 magnetic nanoparticles carried with n-butylidenephthalide as targeting-based anticancer agents. Artif. Cells Nanomed. Biotechnol. 47(1), 210–220 (2019).
    • 85. Tsvetkova Y, Beztsinna N, Baues M et al. Balancing passive and active targeting to different tumor compartments using riboflavin-functionalized polymeric nanocarriers. Nano Lett. 17(8), 4665–4674 (2017).
    • 86. Martinez-Bosch N, Rodriguez-Vida A, Juanpere N et al. Galectins in prostate and bladder cancer: tumorigenic roles and clinical opportunities. Nat. Rev. Urol. 16(7), 433–445 (2019).
    • 87. Thiemann S, Baum LG. Galectins and immune responses – just how do they do those things they do? Ann. Rev. Immunol. 34(1), 243–264 (2016).
    • 88. D’Haene N, Sauvage S, Maris C et al. VEGFR1 and VEGFR2 involvement in extracellular galectin-1- and galectin-3-induced angiogenesis. PLOS ONE 8(6), e67029 (2013).
    • 89. Yasinska IM, Sumbayev VV. Alliance of galectin-3 and CD74 biochemical networks as a crucial component of survival machinery operated by human acute myeloid leukaemia cells. Ebiomedicine 44, 2–3 (2019).
    • 90. Fettis MM, Farhadi SA, Hudalla GA. A chimeric, multivalent assembly of galectin-1 and galectin-3 with enhanced extracellular activity. Biomater. Sci. 7(5), 1852–1862 (2019).
    • 91. Cousin JM, Cloninger MJ. The role of galectin-1 in cancer progression, and synthetic multivalent systems for the study of galectin-1. Int. J. Mol. Sci. 17(9), 1566 (2016).
    • 92. Jenkins SV, Nedosekin DA, Miller EK et al. Galectin-1-based tumour-targeting for gold nanostructure-mediated photothermal therapy. Int. J. Hyperthermia 34(1), 19–29 (2018).
    • 93. Wang L, Guo XL. Molecular regulation of galectin-3 expression and therapeutic implication in cancer progression. Biomed. Pharmacother. 78, 165–171 (2016).
    • 94. Laderach DJ, Gentilini LD, Giribaldi L et al. A unique galectin signature in human prostate cancer progression suggests galectin-1 as a key target for treatment of advanced disease. Cancer Res. 73(1), 86–96 (2013).