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 -- 2053 2022-04-13 12:21:56 |
2 Adjust the reference format -5 word(s) 2048 2022-04-15 04:53:45 | |
3 Adjust figure legend Meta information modification 2048 2022-04-15 08:40:51 |

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.
Zhu, L.; Jiang, Y.; Lin, W. Targeted Drug Delivery for Blood Cancers. Encyclopedia. Available online: https://encyclopedia.pub/entry/21710 (accessed on 27 July 2024).
Zhu L, Jiang Y, Lin W. Targeted Drug Delivery for Blood Cancers. Encyclopedia. Available at: https://encyclopedia.pub/entry/21710. Accessed July 27, 2024.
Zhu, Linyi, Yao Jiang, Weifeng Lin. "Targeted Drug Delivery for Blood Cancers" Encyclopedia, https://encyclopedia.pub/entry/21710 (accessed July 27, 2024).
Zhu, L., Jiang, Y., & Lin, W. (2022, April 13). Targeted Drug Delivery for Blood Cancers. In Encyclopedia. https://encyclopedia.pub/entry/21710
Zhu, Linyi, et al. "Targeted Drug Delivery for Blood Cancers." Encyclopedia. Web. 13 April, 2022.
Targeted Drug Delivery for Blood Cancers
Edit

Blood cancers are a type of liquid tumor which means cancer is present in the body fluid. Multiple myeloma, leukemia, and lymphoma are the three common types of blood cancers. Chemotherapy is the major therapy of blood cancers by systemic administration of anticancer agents into the blood. However, a high incidence of relapse often happens, due to the low efficiency of the anticancer agents that accumulate in the tumor site, and therefore lead to a low survival rate of patients. This indicates an urgent need for a targeted drug delivery system to improve the safety and efficacy of therapeutics for blood cancers.

blood cancers drug delivery nanomedicines

1. Introduction

Cancers are one of the leading causes of death in the world [1]. Unlike solid tumors such as those in organs, blood cancers (including multiple myeloma, leukemia, and lymphoma) form in the bone marrow or in the lymphatic system [2][3]. Although, many chemotherapeutic drugs are clinically available for the treatment of blood cancers, there are no curative treatment approaches in clinical practice for these types of cancers due to the inevitable aggravation of blood cancers and bone metastasis [4]. Furthermore, it is difficult to achieve a sufficient therapeutic dose of anticancer agents at tumor sites inside bone marrow or the lymphatic system to suppress tumor growth via systematic administration [5]. To maintain therapeutic levels in bone marrow or the lymphatic system, chemotherapeutics require high dosage and/or more frequent administration which can result in increased side effects [6]. In addition, the bone marrow microenvironment contains a huge number of hematopoietic stem/progenitor cells which are resistance to chemotherapy and mediate disease refractory/relapse [4]. Therefore, a targeted drug delivery system for blood cancers is a significant challenge for chemotherapy.

2. Targeting Delivery Strategy

2.1. Targeting Bone Marrow and Its Microenvironment

The bone marrow microenvironment plays a critical role in the maintenance of cell renewal and differentiation, especially for cancer cells. The bone marrow contains numerous blood vessels and capillaries. It is considered to be one of the most complex systems comprising various cell types including endothelia cells, stromal cells, osteocytes, fibroblasts, mesenchymal stem cells, macrophages, osteoclasts, and osteoblasts. Moreover, the non-cellular component including the extracellular matrix, oxygen tension, cytokines, and mechanical forces are also essential for cancer cell proliferation and are related to resistance. Targeting the bone marrow microenvironment can be improved by drug delivery systems and can be achieved passively or actively.

2.1.1. Passive Targeting Strategy

Potentially beneficial properties of nanotherapeutics include improved bioavailability, reduced toxicity, greater dose response, and enhanced solubility as compared with conventional medicines [7]. Passive targeting depends on accumulation of the drug delivery system at a specific end organ or tumor site, through leaky vasculatures which mainly require a delivery system for its own characteristics including the size, shape, surface zeta-potential, and other properties. In the bone marrow, the drug accumulation amounts in the bone are related to the reticulo-endothelial cells in vessels [8]. The ideal size of nanoparticles for blood cancers should be between 50 nm and 100 nm.

2.1.2. Targeting Bone Surface-Mediated Bone Marrow

Bone is rich in hydroxyapatite which has a high affinity with glutamic acid or aspartic acid [9]. It has been reported that several oligopeptides have demonstrated their particular interactions with bone tissues. Eight repetitive aspartic acids, also known as Asp8, is one of the successful examples which has been reported to bind to bone-resorption surfaces [10]. Asp8 has been demonstrated to mainly bind to the highly crystallized hydroxyapatite of bone-resorption surfaces. It was found that Asp8-icaritin-liposome enhanced bone formation in ovariectomized mice as compared with an icaritin-liposome control lacking the Asp8 moiety (Figure 1a–c) [11]. (AspSerSer)6 is another type of oligopeptide used for the bone-surface delivery that mainly binds to calcium phosphate which is mainly distributed in the mantle dentin in the bone [12]. Hu et al. used the (AspSerSer)6-cationic liposome system to deliver miRNA-132-3p in bone, resulting in prevention and treatment of osteoporosis (Figure 1d–f) [13].
/media/item_content/202204/6258dd7e25665molecules-27-01310-g001.png
Figure 1. Targeting the bone surface-mediated bone marrow: (a) Characterization of the bone-targeting delivery system in vitro and in vivo. Chemical structure of icaritin (ICT); (b) the morphology of Asp8-liposome-icaritin taken by cryo-transmission electron microscope (Cryo-TEM) (scale bar = 100 nm, magnification = 105×); (c) localization of fluorescent-labeled liposome delivery system with or without Asp8 targeting peptide in mice by optical imaging IVIS analysis, 72 h after administration; (d) a schematic diagram to illustrate the experimental design for targeted delivery of antagomir-132 to specifically decrease miRNA-132-3p levels in bone; (e) a schematic diagram to illustrate how antagomir-132 is selectively delivered to bone formation region by (AspSerSer)6; (f) analysis of miRNA-132-3p expression in the femur bone tissues of mice after hindlimb unloading for 21 days. BL—baseline group, mice were euthanatized and sampled at the beginning of experiment; CON—control group mice were raised in normal conditions during the experiment; HU—hindlimb unloading group, mice were submitted to a hindlimb unloading experiment; HU + Mock—hindlimb unloading plus (AspSerSer)6-liposome injection group, mice were injected with the (AspSerSer)6-liposome before HU; HU + antagomir-NC—hindlimb unloading plus (AspSerSer)6-liposome-antagomir-NC injection group, mice were injected with the (AspSerSer)6-liposome-antagomir-NC before HU; HU + antagomir-132—hindlimb unloading plus (AspSerSer)6-liposome-antagomir-132 injection group, mice were injected with the (AspSerSer)6-liposome-antagomir-132 before HU. Values are shown as mean ± SD, n  =  6. * p  <  0.05. NS, not significant. (ac) Adapted with permission from [11] and (df) adapted with permission from [13].

2.1.3. Active Targeting

Active targeting increases specific delivery to tumor tissues. Many blood cancer cells express specific surface biomarkers which can be specifically targeted by coupling peptides/antibodies/ligands to the surface of nanomedicines for drug delivery [14][15]. Targeting nanomedicines modified with peptides is a common strategy investigated extensively in drug delivery research. Liposomes modified with RGD peptide (Arg-Gly-Asp) have been widely used to target angiogenic endothelial cells in tumors [16]. It has also been reported that liposomes conjugated with a cyclic pentamer peptide (VLA-4, very late antigen-4) can be used to target multiple myeloma (Figure 2a–c) [17]. Targeting via antibodies is another approach for an active targeting strategy [18]. It is known that CD38 and CD138 are widely expressed on multiple myeloma cells. Liposomes modified with anti-CD38 or anti-CD138 monoclonal antibody could be a new approach for a targeted delivery system with both targeting myeloma cells and also delivery anticancer agents to cancer cells (Figure 2d,e) [19]. Similar to CD38 and CD138, CD19 is one of the markers expressed in most of the lymphoma diseases [20]; an anti-CD19 targeted liposome encapsuled rapamycin showed promising lymphoma cell-specific treatment inducing autophagic cell death [21][22].
/media/item_content/202204/6258dd95d362amolecules-27-01310-g002.png
Figure 2. Active targeting strategy by peptide or antibody mediated nanomedicines: (a) Illustration of PEGylated non-targeted liposomal carfilzomib nanoparticles (NP[Carf], left) and VLA-4 targeted liposomal carfilzomib nanoparticles (TNP[Carf], right); (b) liposomal carfilzomib nanoparticles preferentially accumulate in the tumor, inhibit tumor growth, and reduce systemic toxicities in vivo. Tumor bearing SCID mice were injected intravenously on Days 1, 2, 8, and 9 with NP[Carf], TNP[Carf], free carfilzomib, and PBS at a dose of 5 mg/kg carfilzomib equivalence. Tumor growth inhibition was measured via calipers; (c) in vivo images of near infrared dye loaded targeted nanoparticles in tumor bearing mice. Images were taken for all mice at t = 2, 6, and 24 h using non-invasive methods. The representative images show the accumulation of the nanoparticles in the tumor (white arrow) over time; (d,e) in vivo efficacy of CD38pep- and CD138pep-targeted nanoparticles loaded with prodrug doxorubicin. Nanoparticles targeted with CD38pep or CD138pep were prepared loaded with a doxorubicin prodrug and their in vivo efficacy was tested against that of free doxorubicin in a subcutaneous xenograft mouse model. Mice were injected with H929 cells and tumors were allowed to grow to a predetermined size before i.v. injection of nanoparticle formulations began on Day 1. Mice were injected with 3 mg/kg of doxorubicin or nanoparticle prodrug equivalent on Days 1, 3, 5, 7, and 9. Tumor volume (d) and survival (e) were tracked with mice being killed when tumor volume grew too large or mouse weight was too low. n = 6 for all groups and data represent means (± s.e.m.). (ac) Adapted with permission from [17] and (de) adapted with permission from [19]).

2.2. Targeting Spleen and Lymphoid Nodes

Spleen and lymph nodes provide a distinct microenvironment for tumor cells in blood cancers. The spleen is considered to be involved in many blood cancers, especially in lymphomas. It has been reported that the spleen also plays a key role in tumor immunity by recruiting monocytes and macrophages to the tumor tissues [23]. In vivo experiments have shown that siRNA encapsuled nanoparticles can reduce tumor growth [24]. Enhanced drug concentration in the spleen has also provided therapeutic benefits in spleen resident infections and hematological disorders including malaria, hairy cell leukemia, idiopathic thrombocytopenic purpura, and autoimmune hemolytic anemia [25].
Lymph nodes initiate most immune responses which can prevent malignant transformation [26]. Antitumor immune responses are still active in some malignancies, impacting progression and outcome. In addition, the cytokines in lymphoid nodes also provide a proinflammatory microenvironment which can also support proliferation of malignant cells [27].

2.3. Targeting Vascular System

Neovascularization is always associated with poor prognosis in most blood cancers including acute myeloid leukemia, multiple myeloma, acute lymphatic leukemia, chronic lymphatic leukemia, and Burkett’s lymphoma [28]. Endothelial surface receptors are highly expressed on the inner lining of blood vessels. Shamay et al. reported that vascular endothelial growth factor receptor 1 (VEGFR1)-targeted polymer drug conjugates showed efficient antitumor effect by targeting tumor vasculature [29]. Another strategy is to utilize tumor-homing immunocytokines such as interleukin-2 (IL-2) [30]. The antibody-based delivery of IL-2 to extracellular targets expressed in the easily accessible tumor-associated vasculature showed therapeutic potential for acute myeloid leukemia and other solid tumors [31].

3. Nanomedicines for Blood Cancers

3.1. Multiple Myeloma

Multiple myeloma (MM) is a B cell malignancy disease which is characterized by the accumulation of malignant plasma cells in the bone marrow. Although the new treatment and transplant has been utilized in recent decades and has prolonged the overall survival for patients, multiple myeloma is still not curable since it is difficult to remove the tumor cells from the bone marrow. Swami et al. reported that PEG-PLGA encapsuled bortezomib nanoparticles inhibited myeloma growth in a mouse model [4]. Ashley et al. reported that carfilzomib-loaded liposomal nanoparticles targeted myeloma cells [17]. A doxorubicin liposome combined with bortezomib for the treatment of relapsed or refractory multiple myeloma has already been approved by FDA for clinical use [32]. The outcome was based on a phase III clinical trial which showed that liposomal doxorubicin was superior to bortezomib monotherapy [33].

3.2. Acute Myeloid Leukemia

Acute myeloid leukemia (AML) is another common type of hematological malignancy which is characterized by high proliferation of abnormal myeloblasts in the bone marrow [34][35]. Chemotherapy is still the primary choice for AML treatment. However, the overall survival of single chemotherapy for AML patients is still very low [36]. A combination of two or more anticancer reagents is often used for AML therapy to increase the treatment outcome, but various adverse effects can happen during the treatments [37]. Recently, there are some drug delivery systems that have been investigated to increase the anti-AML effect. Roboz et al. reported on a lipid-drug conjugate encapsuled cytarabine that has been put into a phase III clinical trial [38]. Alakhova et al. reported on a pluronic-based micelle which could increase the anti-AML efficacy of doxorubicin and was also in a phase III clinical trial [39]. Tardi et al. reported on a cytarabine liposome which could increase accumulation in leukemia cells inside the bone marrow and enhance efficacy in AML xenograft model [40].

3.3. B Cell Lymphomas

Lymphoma is a type of cancer which often happens in lymph nodes. The majority arise from B cells, and therefore, are called B cell lymphomas which include both Hodgkin’s lymphomas and most non-Hodgkin lymphomas [41]. Chemotherapy and stem cell transplantation are two main treatments for B cell lymphomas; however, relapse is often inevitable [42]. Antibody conjugates provided a new way for targeting therapy for B cell lymphomas. Brentuximab vedotin (Adcetris®, Seattle Genetics, Bothell, WA, USA) and ibritumomab tiuxetan (Zevalin®, IDEC, Cambridge, MA /Spectrum, Irvine, CA, USA) are two commercially available antibody-drug conjugates for Hodgkin lymphoma and non-Hodgkin lymphoma which have already been approved by the FDA [43][44]. Furthermore, new technology provides the possibility to selectively deliver anticancer agents to malignant cells without damaging healthy cells or systemic toxicity, allowing them to reach the lymph nodes. Nevala et al. reported on a nano-antibody targeted chemotherapy delivery system that used a slight modification of existing cancer drugs with significantly improved treatment efficacy in CD20+ B-cell lymphoma [45].

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2022. CA Cancer J. Clin. 2022, 72, 7–33.
  2. Rowe, J.M. Perspectives on current survival and new developments in AML. Best Pract. Res. Clin. Haematol. 2021, 34, 101248.
  3. Tuazon, S.A.; Holmberg, L.A.; Nadeem, O.; Richardson, P.G. A clinical perspective on plasma cell leukemia; current status and future directions. Blood Cancer J. 2021, 11, 23.
  4. Swami, A.; Reagan, M.R.; Basto, P.; Mishima, Y.; Kamaly, N.; Glavey, S.; Zhang, S.; Moschetta, M.; Seevaratnam, D.; Zhang, Y.; et al. Engineered nanomedicine for myeloma and bone microenvironment targeting. Proc. Natl. Acad. Sci. USA 2014, 111, 10287–10292.
  5. Adjei, I.M.; Sharma, B.; Peetla, C.; Labhasetwar, V. Inhibition of bone loss with surface-modulated, drug-loaded nanoparticles in an intraosseous model of prostate cancer. J. Control. Release 2016, 232, 83–92.
  6. Jiang, T.; Yu, X.; Carbone, E.J.; Nelson, C.; Kan, H.M.; Lo, K.W. Poly aspartic acid peptide-linked PLGA based nanoscale particles: Potential for bone-targeting drug delivery applications. Int. J. Pharm. 2014, 475, 547–557.
  7. Iannazzo, D.; Ettari, R.; Giofrè, S.; Eid, A.H.; Bitto, A. Recent Advances in Nanotherapeutics for Multiple Myeloma. Cancers 2020, 12, 3144.
  8. Beloqui, A.; Solinís, M.A.; Delgado, A.; Evora, C.; del Pozo-Rodríguez, A.; Rodríguez-Gascón, A. Biodistribution of Nanostructured Lipid Carriers (NLCs) after intravenous administration to rats: Influence of technological factors. Eur. J. Pharm. Biopharm. 2013, 84, 309–314.
  9. Goldberg, H.A.; Warner, K.J.; Li, M.C.; Hunter, G.K. Binding of bone sialoprotein, osteopontin and synthetic polypeptides to hydroxyapatite. Connect. Tissue Res. 2001, 42, 25–37.
  10. Wang, D.; Miller, S.C.; Shlyakhtenko, L.S.; Portillo, A.M.; Liu, X.M.; Papangkorn, K.; Kopecková, P.; Lyubchenko, Y.; Higuchi, W.I.; Kopecek, J. Osteotropic Peptide that differentiates functional domains of the skeleton. Bioconjug. Chem. 2007, 18, 1375–1378.
  11. Huang, L.; Wang, X.; Cao, H.; Li, L.; Chow, D.H.; Tian, L.; Wu, H.; Zhang, J.; Wang, N.; Zheng, L.; et al. A bone-targeting delivery system carrying osteogenic phytomolecule icaritin prevents osteoporosis in mice. Biomaterials 2018, 182, 58–71.
  12. Yang, Y.S.; Xie, J.; Chaugule, S.; Wang, D.; Kim, J.M.; Kim, J.; Tai, P.W.L.; Seo, S.K.; Gravallese, E.; Gao, G.; et al. Bone-Targeting AAV-Mediated Gene Silencing in Osteoclasts for Osteoporosis Therapy. Mol. Ther. Methods Clin. Dev. 2020, 17, 922–935.
  13. Hu, Z.; Zhang, L.; Wang, H.; Wang, Y.; Tan, Y.; Dang, L.; Wang, K.; Sun, Z.; Li, G.; Cao, X.; et al. Targeted silencing of miRNA-132-3p expression rescues disuse osteopenia by promoting mesenchymal stem cell osteogenic differentiation and osteogenesis in mice. Stem Cell Res. Ther. 2020, 11, 58.
  14. Jiang, Y.; Yang, N.; Zhang, H.; Sun, B.; Hou, C.; Ji, C.; Zheng, J.; Liu, Y.; Zuo, P. Enhanced in vivo antitumor efficacy of dual-functional peptide-modified docetaxel nanoparticles through tumor targeting and Hsp90 inhibition. J. Control. Release 2016, 221, 26–36.
  15. Yang, N.; Jiang, Y.; Zhang, H.; Sun, B.; Hou, C.; Zheng, J.; Liu, Y.; Zuo, P. Active targeting docetaxel-PLA nanoparticles eradicate circulating lung cancer stem-like cells and inhibit liver metastasis. Mol. Pharm. 2015, 12, 232–239.
  16. Mahmoudi, R.; Ashraf Mirahmadi-Babaheidri, S.; Delaviz, H.; Fouani, M.H.; Alipour, M.; Jafari Barmak, M.; Christiansen, G.; Bardania, H. RGD peptide-mediated liposomal curcumin targeted delivery to breast cancer cells. J. Biomater. Appl. 2021, 35, 743–753.
  17. Ashley, J.D.; Stefanick, J.F.; Schroeder, V.A.; Suckow, M.A.; Alves, N.J.; Suzuki, R.; Kikuchi, S.; Hideshima, T.; Anderson, K.C.; Kiziltepe, T.; et al. Liposomal carfilzomib nanoparticles effectively target multiple myeloma cells and demonstrate enhanced efficacy in vivo. J. Control. Release 2014, 196, 113–121.
  18. Kim, D.; Park, C.Y.; Medeiros, B.C.; Weissman, I.L. CD19-CD45 low/-CD38 high/CD138+ plasma cells enrich for human tumorigenic myeloma cells. Leukemia 2012, 26, 2530–2537.
  19. Omstead, D.T.; Mejia, F.; Sjoerdsma, J.; Kim, B.; Shin, J.; Khan, S.; Wu, J.; Kiziltepe, T.; Littlepage, L.E.; Bilgicer, B. In vivo evaluation of CD38 and CD138 as targets for nanoparticle-based drug delivery in multiple myeloma. J. Hematol. Oncol. 2020, 13, 145.
  20. Wang, K.; Wei, G.; Liu, D. CD19: A biomarker for B cell development, lymphoma diagnosis and therapy. Exp. Hematol. Oncol. 2012, 1, 36.
  21. Ono, K.; Sato, T.; Iyama, S.; Tatekoshi, A.; Hashimoto, A.; Kamihara, Y.; Horiguchi, H.; Kikuchi, S.; Kawano, Y.; Takada, K.; et al. A novel strategy inducing autophagic cell death in Burkitt’s lymphoma cells with anti-CD19-targeted liposomal rapamycin. Blood Cancer J. 2014, 4, e180.
  22. Zhang, J.; Shen, D.; Jia, M.; Zhao, H.; Tang, Y. The targeting effect of Hm2E8b-NCTD-liposomes on B-lineage leukaemia stem cells is associated with the HLF-SLUG axis. J. Drug Target. 2018, 26, 55–65.
  23. Bronte, V.; Pittet, M.J. The spleen in local and systemic regulation of immunity. Immunity 2013, 39, 806–818.
  24. Leuschner, F.; Dutta, P.; Gorbatov, R.; Novobrantseva, T.I.; Donahoe, J.S.; Courties, G.; Lee, K.M.; Kim, J.I.; Markmann, J.F.; Marinelli, B.; et al. Therapeutic siRNA silencing in inflammatory monocytes in mice. Nat. Biotechnol. 2011, 29, 1005–1010.
  25. Jindal, A.B. Nanocarriers for spleen targeting: Anatomo-physiological considerations, formulation strategies and therapeutic potential. Drug Deliv. Transl. Res. 2016, 6, 473–485.
  26. Mu, C.F.; Shen, J.; Liang, J.; Zheng, H.S.; Xiong, Y.; Wei, Y.H.; Li, F. Targeted drug delivery for tumor therapy inside the bone marrow. Biomaterials 2018, 155, 191–202.
  27. Park, C.S.; Choi, Y.S. How do follicular dendritic cells interact intimately with B cells in the germinal centre? Immunology 2005, 114, 2–10.
  28. Lugano, R.; Ramachandran, M.; Dimberg, A. Tumor angiogenesis: Causes, consequences, challenges and opportunities. Cell. Mol. Life Sci. 2020, 77, 1745–1770.
  29. Shamay, Y.; Golan, M.; Tyomkin, D.; David, A. Assessing the therapeutic efficacy of VEGFR-1-targeted polymer drug conjugates in mouse tumor models. J. Control. Release 2016, 229, 192–199.
  30. Runbeck, E.; Crescioli, S.; Karagiannis, S.N.; Papa, S. Utilizing Immunocytokines for Cancer Therapy. Antibodies 2021, 10, 10.
  31. Schliemann, C.; Gutbrodt, K.L.; Kerkhoff, A.; Pohlen, M.; Wiebe, S.; Silling, G.; Angenendt, L.; Kessler, T.; Mesters, R.M.; Giovannoni, L.; et al. Targeting interleukin-2 to the bone marrow stroma for therapy of acute myeloid leukemia relapsing after allogeneic hematopoietic stem cell transplantation. Cancer Immunol. Res. 2015, 3, 547–556.
  32. Ning, Y.M.; He, K.; Dagher, R.; Sridhara, R.; Farrell, A.T.; Justice, R.; Pazdur, R. Liposomal doxorubicin in combination with bortezomib for relapsed or refractory multiple myeloma. Oncology 2007, 21, 1503–1508.
  33. Orlowski, R.Z.; Nagler, A.; Sonneveld, P.; Bladé, J.; Hajek, R.; Spencer, A.; Robak, T.; Dmoszynska, A.; Horvath, N.; Spicka, I.; et al. Final overall survival results of a randomized trial comparing bortezomib plus pegylated liposomal doxorubicin with bortezomib alone in patients with relapsed or refractory multiple myeloma. Cancer 2016, 122, 2050–2056.
  34. Löwenberg, B.; Downing, J.R.; Burnett, A. Acute myeloid leukemia. N. Engl. J. Med. 1999, 341, 1051–1062.
  35. Huang, X.; Lin, H.; Huang, F.; Xie, Y.; Wong, K.H.; Chen, X.; Wu, D.; Lu, A.; Yang, Z. Targeting Approaches of Nanomedicines in Acute Myeloid Leukemia. Dose Response 2019, 17, 1559325819887048.
  36. Arber, D.A.; Orazi, A.; Hasserjian, R.; Thiele, J.; Borowitz, M.J.; Le Beau, M.M.; Bloomfield, C.D.; Cazzola, M.; Vardiman, J.W. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 2016, 127, 2391–2405.
  37. Dombret, H.; Gardin, C. An update of current treatments for adult acute myeloid leukemia. Blood 2016, 127, 53–61.
  38. Roboz, G.J.; Rosenblat, T.; Arellano, M.; Gobbi, M.; Altman, J.K.; Montesinos, P.; O’Connell, C.; Solomon, S.R.; Pigneux, A.; Vey, N.; et al. International randomized phase III study of elacytarabine versus investigator choice in patients with relapsed/refractory acute myeloid leukemia. J. Clin. Oncol 2014, 32, 1919–1926.
  39. Alakhova, D.Y.; Zhao, Y.; Li, S.; Kabanov, A.V. Effect of doxorubicin/pluronic SP1049C on tumorigenicity, aggressiveness, DNA methylation and stem cell markers in murine leukemia. PLoS ONE 2013, 8, e72238.
  40. Tardi, P.; Wan, C.P.; Mayer, L. Passive and semi-active targeting of bone marrow and leukemia cells using anionic low cholesterol liposomes. J. Drug Target. 2016, 24, 797–804.
  41. Novo, M.; Castellino, A.; Nicolosi, M.; Santambrogio, E.; Vassallo, F.; Chiappella, A.; Vitolo, U. High-grade B-cell lymphoma: How to diagnose and treat. Expert Rev. Hematol. 2019, 12, 497–506.
  42. Deshantri, A.K.; Varela Moreira, A.; Ecker, V.; Mandhane, S.N.; Schiffelers, R.M.; Buchner, M.; Fens, M. Nanomedicines for the treatment of hematological malignancies. J. Control. Release 2018, 287, 194–215.
  43. Younes, A.; Gopal, A.K.; Smith, S.E.; Ansell, S.M.; Rosenblatt, J.D.; Savage, K.J.; Ramchandren, R.; Bartlett, N.L.; Cheson, B.D.; de Vos, S.; et al. Results of a pivotal phase II study of brentuximab vedotin for patients with relapsed or refractory Hodgkin’s lymphoma. J. Clin. Oncol. 2012, 30, 2183–2189.
  44. Morschhauser, F.; Radford, J.; Van Hoof, A.; Vitolo, U.; Soubeyran, P.; Tilly, H.; Huijgens, P.C.; Kolstad, A.; d’Amore, F.; Gonzalez Diaz, M.; et al. Phase III trial of consolidation therapy with yttrium-90-ibritumomab tiuxetan compared with no additional therapy after first remission in advanced follicular lymphoma. J. Clin. Oncol. 2008, 26, 5156–5164.
  45. Nevala, W.K.; Butterfield, J.T.; Sutor, S.L.; Knauer, D.J.; Markovic, S.N. Antibody-targeted paclitaxel loaded nanoparticles for the treatment of CD20(+) B-cell lymphoma. Sci. Rep. 2017, 7, 45682.
More
Information
Subjects: Hematology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 415
Revisions: 3 times (View History)
Update Date: 15 Apr 2022
1000/1000
Video Production Service