Advancements in Nanotechnology for Targeted and Controlled Drug Delivery in Hematologic Malignancies: Shaping the Future of Targeted Therapeutics
Abstract
:1. Introduction
1.1. Precision Drug Delivery: Transforming Hematological Malignancies
1.2. Nanotechnology-Driven Advancements in Hematologic Malignancioes Treatment
1.3. Current Landscape of Nanotechnology in Drug Delivery
1.4. Nanocarriers: Types and Mechanisms
1.5. Advantages over Conventional Drug Delivery Methods
2. Applications in Hematologic Malignancies
2.1. Nanocarriers in Leukemia Treatment
2.2. Nanocarriers in Lymphoma
2.3. Nanocarriers in Multiple Myeloma
2.4. Emerging Clinical Trials and Future Directions
3. Mechanisms of Precision Delivery via Nanotechnology
3.1. Controlled Drug Release and Passive Targeting Mechanisms
3.2. Active Targeting Mechanisms
3.3. Stimuli-Responsive Nanocarriers
3.4. Clinical Advances and Challenges in Nanotechnology for Hematologic Malignancies
3.5. Clinical Trials and Real-World Applications
3.6. Safety, Toxicity, and Efficacy Concerns
3.7. Scalability, Manufacturing, and Regulatory Challenges
3.8. Future Directions and Prospects in Nanotechnology for Hematologic Malignancies
3.9. Emerging Trends and Personalized Medicine
3.10. Research Gaps and Challenges
3.11. Potential Clinical Impact and Future Directions
4. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Siegel, R.L.; Miller, K.D.; Wagle, N.S.; Jemal, A. Cancer statistics, 2023. CA Cancer J. Clin. 2023, 73, 17–48. [Google Scholar] [CrossRef] [PubMed]
- Swerdlow, S.H.; Campo, E.; Pileri, S.A.; Harris, N.L.; Stein, H.; Siebert, R.; Advani, R.; Ghielmini, M.; Salles, G.A.; Zelenetz, A.D.; et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood 2016, 127, 2375–2390. [Google Scholar] [CrossRef]
- Campo, E.; Harris, N.L.; Jaffe, E.S.; Pileri, S.A.; Stein, H.; Thiele, J. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues; International Agency for Research on Cancer: Lyon, France, 2017. [Google Scholar]
- Malard, F.; Neri, P.; Bahlis, N.J.; Terpos, E.; Moukalled, N.; Hungria, V.T.M.; Manier, S.; Mohty, M. Multiple myeloma. Nat. Rev. Dis. Primers 2024, 10, 45. [Google Scholar] [CrossRef] [PubMed]
- Nogami, A.; Sasaki, K. Therapeutic Advances in Immunotherapies for Hematological Malignancies. Int. J. Mol. Sci. 2022, 23, 11526. [Google Scholar] [CrossRef] [PubMed]
- Imai, Y. Novel treatment strategies for hematological malignancies in the immunotherapy era. Int. J. Hematol. 2024, 120, 3–5. [Google Scholar] [CrossRef]
- DiNardo, C.D.; Jonas, B.A.; Pullarkat, V.; Thirman, M.J.; Garcia, J.S.; Wei, A.H.; Konopleva, M.; Döhner, H.; Letai, A.; Fenaux, P.; et al. Azacitidine and Venetoclax in Previously Untreated Acute Myeloid Leukemia. N. Engl. J. Med. 2020, 383, 617–629. [Google Scholar] [CrossRef]
- Tang, L.; Huang, Z.; Mei, H.; Hu, Y. Immunotherapy in hematologic malignancies: Achievements, challenges and future prospects. Signal Transduct. Target. Ther. 2023, 8, 306. [Google Scholar] [CrossRef]
- Lica, J.J.; Pradhan, B.; Safi, K.; Jakóbkiewicz-Banecka, J.; Hellmann, A. Promising Therapeutic Strategies for Hematologic Malignancies: Innovations and Potential. Molecules 2024, 29, 4280. [Google Scholar] [CrossRef]
- Garg, A.; Nair, K.; Chavan, S.; Mukundan, M.; Kumar, P. Quality of life in adult patients with hematological malignancy- treading a road less travelled. Indian J. Hematol. Blood Transfus. 2024. [Google Scholar] [CrossRef]
- Rosenblum, D.; Joshi, N.; Tao, W.; Karp, J.M.; Peer, D. Progress and challenges towards targeted delivery of cancer therapeutics. Nat. Commun. 2018, 9, 1410. [Google Scholar] [CrossRef]
- Megías-Vericat, J.E.; Martínez-Cuadrón, D.; Solana-Altabella, A.; Montesinos, P. Precision medicine in acute myeloid leukemia: Where are we now and what does the future hold? Expert. Rev. Hematol. 2020, 13, 1057–1065. [Google Scholar] [CrossRef] [PubMed]
- Pui, C.H. Precision medicine in acute lymphoblastic leukemia. Front. Med. 2020, 14, 689–700. [Google Scholar] [CrossRef] [PubMed]
- Ferrari, M. Cancer nanotechnology: Opportunities and challenges. Nat. Rev. Cancer 2005, 5, 161–171. [Google Scholar] [CrossRef] [PubMed]
- Allegra, A.; Gioacchino, M.D.; Tonacci, A.; Petrarca, C.; Gangemi, S. Nanomedicine for Immunotherapy Targeting Hematological Malignancies: Current Approaches and Perspective. Nanomaterials 2021, 11, 2792. [Google Scholar] [CrossRef]
- Wang, B.; Hu, S.; Teng, Y.; Chen, J.; Wang, H.; Xu, Y.; Wang, K.; Xu, J.; Cheng, Y.; Gao, X. Current advance of nanotechnology in diagnosis and treatment for malignant tumors. Signal Transduct. Target. Ther. 2024, 9, 200. [Google Scholar] [CrossRef]
- Amin, M.; Seynhaeve, A.L.B.; Sharifi, M.; Falahati, M.; Ten Hagen, T.L.M. Liposomal Drug Delivery Systems for Cancer Therapy: The Rotterdam Experience. Pharmaceutics 2022, 14, 2165. [Google Scholar] [CrossRef]
- Olusanya, T.O.B.; Haj Ahmad, R.R.; Ibegbu, D.M.; Smith, J.R.; Elkordy, A.A. Liposomal Drug Delivery Systems and Anticancer Drugs. Molecules 2018, 23, 907. [Google Scholar] [CrossRef]
- Chen, J.; Hu, S.; Sun, M.; Shi, J.; Zhang, H.; Yu, H.; Yang, Z. Recent advances and clinical translation of liposomal delivery systems in cancer therapy. Eur. J. Pharm. Sci. 2024, 193, 106688. [Google Scholar] [CrossRef]
- Maeda, H.; Nakamura, H.; Fang, J. The EPR effect for macromolecular drug delivery to solid tumors: Improvement of tumor uptake, lowering of systemic toxicity, and distinct tumor imaging in vivo. Adv. Drug Deliv. Rev. 2013, 65, 71–79. [Google Scholar] [CrossRef]
- Subhan, M.A.; Parveen, F.; Filipczak, N.; Yalamarty, S.S.K.; Torchilin, V.P. Approaches to Improve EPR-Based Drug Delivery for Cancer Therapy and Diagnosis. J. Pers. Med. 2023, 13, 389. [Google Scholar] [CrossRef]
- Cho, K.; Wang, X.; Nie, S.; Chen, Z.G.; Shin, D.M. Therapeutic nanoparticles for drug delivery in cancer. Clin. Cancer Res. 2008, 14, 1310–1316. [Google Scholar] [CrossRef] [PubMed]
- Yao, Y.; Zhou, Y.; Liu, L.; Xu, Y.; Chen, Q.; Wang, Y.; Wu, S.; Deng, Y.; Zhang, J.; Shao, A. Nanoparticle-Based Drug Delivery in Cancer Therapy and Its Role in Overcoming Drug Resistance. Front. Mol. Biosci. 2020, 7, 193. [Google Scholar] [CrossRef] [PubMed]
- Khawar, I.A.; Kim, J.H.; Kuh, H.J. Improving drug delivery to solid tumors: Priming the tumor microenvironment. J. Control Release 2015, 201, 78–89. [Google Scholar] [CrossRef] [PubMed]
- Khan, N.; Ruchika; Dhritlahre, R.K.; Saneja, A. Recent advances in dual-ligand targeted nanocarriers for cancer therapy. Drug Discov. Today 2022, 27, 2288–2299. [Google Scholar] [CrossRef]
- Yan, S.; Na, J.; Liu, X.; Wu, P. Different Targeting Ligands-Mediated Drug Delivery Systems for Tumor Therapy. Pharmaceutics 2024, 16, 248. [Google Scholar] [CrossRef]
- Sharifi, M.; Cho, W.C.; Ansariesfahani, A.; Tarharoudi, R.; Malekisarvar, H.; Sari, S.; Bloukh, S.H.; Edis, Z.; Amin, M.; Gleghorn, J.P.; et al. An Updated Review on EPR-Based Solid Tumor Targeting Nanocarriers for Cancer Treatment. Cancers 2022, 14, 2868. [Google Scholar] [CrossRef]
- Chen, H.-J.; Cheng, Y.-A.; Chen, Y.-T.; Li, C.-C.; Huang, B.-C.; Hong, S.-T.; Chen, I.J.; Ho, K.-W.; Chen, C.-Y.; Chen, F.-M.; et al. Targeting and internalizing PEGylated nanodrugs to enhance the therapeutic efficacy of hematologic malignancies by anti-PEG bispecific antibody (mPEG × CD20). Cancer Nanotechnol. 2023, 14, 78. [Google Scholar] [CrossRef]
- Vasir, J.K.; Labhasetwar, V. Targeted drug delivery in cancer therapy. Technol. Cancer Res. Treat. 2005, 4, 363–374. [Google Scholar] [CrossRef]
- Wang, X.; Li, C.; Wang, Y.; Chen, H.; Zhang, X.; Luo, C.; Zhou, W.; Li, L.; Teng, L.; Yu, H.; et al. Smart drug delivery systems for precise cancer therapy. Acta Pharm. Sin. B 2022, 12, 4098–4121. [Google Scholar] [CrossRef]
- AlSawaftah, N.M.; Awad, N.S.; Pitt, W.G.; Husseini, G.A. pH-Responsive Nanocarriers in Cancer Therapy. Polymers 2022, 14, 936. [Google Scholar] [CrossRef]
- Palanikumar, L.; Al-Hosani, S.; Kalmouni, M.; Nguyen, V.P.; Ali, L.; Pasricha, R.; Barrera, F.N.; Magzoub, M. pH-responsive high stability polymeric nanoparticles for targeted delivery of anticancer therapeutics. Commun. Biol. 2020, 3, 95. [Google Scholar] [CrossRef] [PubMed]
- Sun, L.; Liu, H.; Ye, Y.; Lei, Y.; Islam, R.; Tan, S.; Tong, R.; Miao, Y.-B.; Cai, L. Smart nanoparticles for cancer therapy. Signal Transduct. Target. Ther. 2023, 8, 418. [Google Scholar] [CrossRef] [PubMed]
- Nteli, P.; Bajwa, D.E.; Politakis, D.; Michalopoulos, C.; Kefala-Narin, A.; Efstathopoulos, E.P.; Gazouli, M. Nanomedicine approaches for treatment of hematologic and oncologic malignancies. World J. Clin. Oncol. 2022, 13, 553–566. [Google Scholar] [CrossRef] [PubMed]
- Tian, H.; Zhang, T.; Qin, S.; Huang, Z.; Zhou, L.; Shi, J.; Nice, E.C.; Xie, N.; Huang, C.; Shen, Z. Enhancing the therapeutic efficacy of nanoparticles for cancer treatment using versatile targeted strategies. J. Hematol. Oncol. 2022, 15, 132. [Google Scholar] [CrossRef]
- Qian, S.; Zheng, C.; Wu, Y.; Huang, H.; Wu, G.; Zhang, J. Targeted therapy for leukemia based on nanomaterials. Heliyon 2024, 10, e34951. [Google Scholar] [CrossRef]
- Salama, M.M.; Aborehab, N.M.; El Mahdy, N.M.; Zayed, A.; Ezzat, S.M. Nanotechnology in leukemia: Diagnosis, efficient-targeted drug delivery, and clinical trials. Eur. J. Med. Res. 2023, 28, 566. [Google Scholar] [CrossRef]
- Fumoto, S.; Nishida, K. Co-delivery Systems of Multiple Drugs Using Nanotechnology for Future Cancer Therapy. Chem. Pharm. Bull. 2020, 68, 603–612. [Google Scholar] [CrossRef]
- Han, Y.; Zhang, P.; Chen, Y.; Sun, J.; Kong, F. Co-delivery of plasmid DNA and doxorubicin by solid lipid nanoparticles for lung cancer therapy. Int. J. Mol. Med. 2014, 34, 191–196. [Google Scholar] [CrossRef]
- Aloss, K.; Hamar, P. Recent Preclinical and Clinical Progress in Liposomal Doxorubicin. Pharmaceutics 2023, 15, 893. [Google Scholar] [CrossRef]
- Vinhas, R.; Mendes, R.; Fernandes, A.R.; Baptista, P.V. Nanoparticles-Emerging Potential for Managing Leukemia and Lymphoma. Front. Bioeng. Biotechnol. 2017, 5, 79. [Google Scholar] [CrossRef]
- Gao, Y.; Wang, K.; Zhang, J.; Duan, X.; Sun, Q.; Men, K. Multifunctional nanoparticle for cancer therapy. MedComm 2023, 4, e187. [Google Scholar] [CrossRef] [PubMed]
- Fan, D.; Cao, Y.; Cao, M.; Wang, Y.; Cao, Y.; Gong, T. Nanomedicine in cancer therapy. Signal Transduct. Target. Ther. 2023, 8, 293. [Google Scholar] [CrossRef] [PubMed]
- Dhiman, R.; Bazad, N.; Mukherjee, R.; Himanshu; Gunjan; Leal, E.; Ahmad, S.; Kaur, K.; Raj, V.S.; Chang, C.-M.; et al. Enhanced drug delivery with nanocarriers: A comprehensive review of recent advances in breast cancer detection and treatment. Discov. Nano 2024, 19, 143. [Google Scholar] [CrossRef] [PubMed]
- Zhuo, Y.; Zhao, Y.-G.; Zhang, Y. Enhancing Drug Solubility, Bioavailability, and Targeted Therapeutic Applications through Magnetic Nanoparticles. Molecules 2024, 29, 4854. [Google Scholar] [CrossRef]
- Crintea, A.; Dutu, A.G.; Sovrea, A.; Constantin, A.-M.; Samasca, G.; Masalar, A.L.; Ifju, B.; Linga, E.; Neamti, L.; Tranca, R.A.; et al. Nanocarriers for Drug Delivery: An Overview with Emphasis on Vitamin D and K Transportation. Nanomaterials 2022, 12, 1376. [Google Scholar] [CrossRef]
- Cong, X.; Zhang, Z.; Li, H.; Yang, Y.-G.; Zhang, Y.; Sun, T. Nanocarriers for targeted drug delivery in the vascular system: Focus on endothelium. J. Nanobiotechnol. 2024, 22, 620. [Google Scholar] [CrossRef]
- Hu, Q.; Katti, P.S.; Gu, Z. Enzyme-responsive nanomaterials for controlled drug delivery. Nanoscale 2014, 6, 12273–12286. [Google Scholar] [CrossRef]
- Kapalatiya, H.; Madav, Y.; Tambe, V.S.; Wairkar, S. Enzyme-responsive smart nanocarriers for targeted chemotherapy: An overview. Drug Deliv. Transl. Res. 2022, 12, 1293–1305. [Google Scholar] [CrossRef]
- Raikwar, S.; Vyas, S.; Sharma, R.; Mody, N.; Dubey, S.; Vyas, S.P. Nanocarrier-Based Combination Chemotherapy for Resistant Tumor: Development, Characterization, and Ex Vivo Cytotoxicity Assessment. AAPS PharmSciTech 2018, 19, 3839–3849. [Google Scholar] [CrossRef]
- Kumar, A.; Lunawat, A.K.; Kumar, A.; Sharma, T.; Islam, M.M.; Kahlon, M.S.; Mukherjee, D.; Narang, R.K.; Raikwar, S. Recent Trends in Nanocarrier-Based Drug Delivery System for Prostate Cancer. AAPS PharmSciTech 2024, 25, 55. [Google Scholar] [CrossRef]
- Dang, Y.; Guan, J. Nanoparticle-based drug delivery systems for cancer therapy. Smart Mater. Med. 2020, 1, 10–19. [Google Scholar] [CrossRef] [PubMed]
- Gabizon, A.; Shmeeda, H.; Barenholz, Y. Pharmacokinetics of pegylated liposomal Doxorubicin: Review of animal and human studies. Clin. Pharmacokinet. 2003, 42, 419–436. [Google Scholar] [CrossRef] [PubMed]
- Cheng, Z.; Fobian, S.-F.; Gurrieri, E.; Amin, M.; D’Agostino, V.G.; Falahati, M.; Zalba, S.; Debets, R.; Garrido, M.J.; Saeed, M.; et al. Lipid-based nanosystems: The next generation of cancer immune therapy. J. Hematol. Oncol. 2024, 17, 53. [Google Scholar] [CrossRef] [PubMed]
- Allen, T.M.; Cullis, P.R. Liposomal drug delivery systems: From concept to clinical applications. Adv. Drug Deliv. Rev. 2013, 65, 36–48. [Google Scholar] [CrossRef]
- Sercombe, L.; Veerati, T.; Moheimani, F.; Wu, S.Y.; Sood, A.K.; Hua, S. Advances and Challenges of Liposome Assisted Drug Delivery. Front. Pharmacol. 2015, 6, 286. [Google Scholar] [CrossRef]
- Barenholz, Y. Doxil®—The first FDA-approved nano-drug: Lessons learned. J. Control Release 2012, 160, 117–134. [Google Scholar] [CrossRef]
- Bhardwaj, V.; Kaushik, A.; Khatib, Z.M.; Nair, M.; McGoron, A.J. Recalcitrant Issues and New Frontiers in Nano-Pharmacology. Front. Pharmacol. 2019, 10, 1369. [Google Scholar] [CrossRef]
- Kamaly, N.; Yameen, B.; Wu, J.; Farokhzad, O.C. Degradable Controlled-Release Polymers and Polymeric Nanoparticles: Mechanisms of Controlling Drug Release. Chem. Rev. 2016, 116, 2602–2663. [Google Scholar] [CrossRef]
- Wicki, A.; Witzigmann, D.; Balasubramanian, V.; Huwyler, J. Nanomedicine in cancer therapy: Challenges, opportunities, and clinical applications. J. Control Release 2015, 200, 138–157. [Google Scholar] [CrossRef]
- Norouzi, M.; Amerian, M.; Amerian, M.; Atyabi, F. Clinical applications of nanomedicine in cancer therapy. Drug Discov. Today 2020, 25, 107–125. [Google Scholar] [CrossRef]
- Prajapati, S.; Maurya, S.; Das, M.; Tilak, V.; Verma, K.; Dhakar, R.C. Dendrimers in Drug Delivery, Diagnosis and Therapy: Basics and Potential Applications. J. Drug Deliv. Ther. 2016, 6, 67–92. [Google Scholar] [CrossRef]
- Jain, R.K.; Stylianopoulos, T. Delivering nanomedicine to solid tumors. Nat. Rev. Clin. Oncol. 2010, 7, 653–664. [Google Scholar] [CrossRef] [PubMed]
- Sharma, N.; Bhatt, G.; Kothiyal, P. Gold Nanoparticles synthesis, properties, and forthcoming applications: A review. Indian J. Pharm. Biol. Res. 2015, 3. [Google Scholar] [CrossRef]
- Alex, S.; Tiwari, A. Functionalized Gold Nanoparticles: Synthesis, Properties and Applications—A Review. J. Nanosci. Nanotechnol. 2015, 15, 1869–1894. [Google Scholar] [CrossRef]
- Pisitsak, P.; Chamchoy, K.; Chinprateep, V.; Khobthong, W.; Chitichotpanya, P.; Ummartyotin, S. Synthesis of Gold Nanoparticles Using Tannin-Rich Extract and Coating onto Cotton Textiles for Catalytic Degradation of Congo Red. J. Nanotechnol. 2021, 2021, 6380283. [Google Scholar] [CrossRef]
- Maeda, H. The enhanced permeability and retention (EPR) effect in tumor vasculature: The key role of tumor-selective macromolecular drug targeting. Adv. Enzym. Regul. 2001, 41, 189–207. [Google Scholar] [CrossRef]
- Wu, J. The Enhanced Permeability and Retention (EPR) Effect: The Significance of the Concept and Methods to Enhance Its Application. J. Pers. Med. 2021, 11, 771. [Google Scholar] [CrossRef]
- Allen, T.M.; Cullis, P.R. Drug delivery systems: Entering the mainstream. Science 2004, 303, 1818–1822. [Google Scholar] [CrossRef]
- Blanco, M.D.; Teijón, C.; Olmo, R.M.; Teijón, J.M. Targeted Nanoparticles for Cancer Therapy. In Recent Advances in Novel Drug Carrier Systems; Ali Demir, S., Ed.; IntechOpen: Rijeka, Croatia, 2012; Chapter 9. [Google Scholar]
- Bhirde, A.A.; Chikkaveeraiah, B.V.; Srivatsan, A.; Niu, G.; Jin, A.J.; Kapoor, A.; Wang, Z.; Patel, S.; Patel, V.; Gorbach, A.M.; et al. Targeted therapeutic nanotubes influence the viscoelasticity of cancer cells to overcome drug resistance. ACS Nano 2014, 8, 4177–4189. [Google Scholar] [CrossRef]
- Aborode, A.T.; Oluwajoba, A.S.; Ibrahim, A.M.; Ahmad, S.; Mehta, A.; Osayawe, O.J.-K.; Oyebode, D.; Akinsola, O.; Osinuga, A.; Onifade, I.A.; et al. Nanomedicine in cancer therapy: Advancing precision treatments. Adv. Biomark. Sci. Technol. 2024, 6, 105–119. [Google Scholar] [CrossRef]
- Wu, X.; Xin, Y.; Zhang, H.; Quan, L.; Ao, Q. Biopolymer-Based Nanomedicine for Cancer Therapy: Opportunities and Challenges. Int. J. Nanomed. 2024, 19, 7415–7471. [Google Scholar] [CrossRef] [PubMed]
- Bertrand, N.; Wu, J.; Xu, X.; Kamaly, N.; Farokhzad, O.C. Cancer nanotechnology: The impact of passive and active targeting in the era of modern cancer biology. Adv. Drug Deliv. Rev. 2014, 66, 2–25. [Google Scholar] [CrossRef] [PubMed]
- Patnaik, S. Nanomedicine Magic Bullet for Human Cancer; IGI Global: Hershey, PA, USA, 2017. [Google Scholar] [CrossRef]
- Minru, G.; Venkatraman, K.; Venkatraman, S. The magic bullet as cancer therapeutic- has nanotechnology failed to find its mark? Prog. Biomed. Eng. 2020, 2, 042004. [Google Scholar] [CrossRef]
- Hani, U.; Gowda, B.H.J.; Haider, N.; Ramesh, K.; Paul, K.; Ashique, S.; Ahmed, M.G.; Narayana, S.; Mohanto, S.; Kesharwani, P. Nanoparticle-Based Approaches for Treatment of Hematological Malignancies: A Comprehensive Review. AAPS PharmSciTech 2023, 24, 233. [Google Scholar] [CrossRef]
- Tiwari, H.; Rai, N.; Singh, S.; Gupta, P.; Verma, A.; Singh, A.K.; Kajal; Salvi, P.; Singh, S.K.; Gautam, V. Recent Advances in Nanomaterials-Based Targeted Drug Delivery for Preclinical Cancer Diagnosis and Therapeutics. Bioengineering 2023, 10, 760. [Google Scholar] [CrossRef]
- Samir, A.; Elgamal, B.M.; Gabr, H.; Sabaawy, H.E. Nanotechnology applications in hematological malignancies (Review). Oncol. Rep. 2015, 34, 1097–1105. [Google Scholar] [CrossRef]
- Xiao, X.; Teng, F.; Shi, C.; Chen, J.; Wu, S.; Wang, B.; Meng, X.; Essiet Imeh, A.; Li, W. Polymeric nanoparticles-Promising carriers for cancer therapy. Front. Bioeng. Biotechnol. 2022, 10, 1024143. [Google Scholar] [CrossRef]
- Yousefi Rizi, H.A.; Hoon Shin, D.; Yousefi Rizi, S. Polymeric Nanoparticles in Cancer Chemotherapy: A Narrative Review. Iran. J. Public Health 2022, 51, 226–239. [Google Scholar] [CrossRef]
- Avramović, N.; Mandić, B.; Savić-Radojević, A.; Simić, T. Polymeric Nanocarriers of Drug Delivery Systems in Cancer Therapy. Pharmaceutics 2020, 12, 298. [Google Scholar] [CrossRef]
- Pourmadadi, M.; Dehaghi, H.M.; Ghaemi, A.; Maleki, H.; Yazdian, F.; Rahdar, A.; Pandey, S. Polymeric nanoparticles as delivery vehicles for targeted delivery of chemotherapy drug fludarabine to treat hematological cancers. Inorg. Chem. Commun. 2024, 167, 112819. [Google Scholar] [CrossRef]
- Li, J.; Wang, Q.; Han, Y.; Jiang, L.; Lu, S.; Wang, B.; Qian, W.; Zhu, M.; Huang, H.; Qian, P. Development and application of nanomaterials, nanotechnology and nanomedicine for treating hematological malignancies. J. Hematol. Oncol. 2023, 16, 65. [Google Scholar] [CrossRef] [PubMed]
- Rahim, M.A.; Jan, N.; Khan, S.; Shah, H.; Madni, A.; Khan, A.; Jabar, A.; Khan, S.; Elhissi, A.; Hussain, Z.; et al. Recent Advancements in Stimuli Responsive Drug Delivery Platforms for Active and Passive Cancer Targeting. Cancers 2021, 13, 670. [Google Scholar] [CrossRef] [PubMed]
- Mi, P. Stimuli-responsive nanocarriers for drug delivery, tumor imaging, therapy and theranostics. Theranostics 2020, 10, 4557–4588. [Google Scholar] [CrossRef]
- Alwattar, J.K.; Mneimneh, A.T.; Abla, K.K.; Mehanna, M.M.; Allam, A.N. Smart Stimuli-Responsive Liposomal Nanohybrid Systems: A Critical Review of Theranostic Behavior in Cancer. Pharmaceutics 2021, 13, 355. [Google Scholar] [CrossRef] [PubMed]
- Ashrafizadeh, M.; Delfi, M.; Zarrabi, A.; Bigham, A.; Sharifi, E.; Rabiee, N.; Paiva-Santos, A.C.; Kumar, A.P.; Tan, S.C.; Hushmandi, K.; et al. Stimuli-responsive liposomal nanoformulations in cancer therapy: Pre-clinical & clinical approaches. J. Control. Release 2022, 351, 50–80. [Google Scholar] [CrossRef]
- Lee, S.-M.; Nguyen, S.T. Smart Nanoscale Drug Delivery Platforms from Stimuli-Responsive Polymers and Liposomes. Macromolecules 2013, 46, 9169–9180. [Google Scholar] [CrossRef]
- Shi, J.; Kantoff, P.W.; Wooster, R.; Farokhzad, O.C. Cancer nanomedicine: Progress, challenges and opportunities. Nat. Rev. Cancer 2017, 17, 20–37. [Google Scholar] [CrossRef]
- Davis, M.E.; Chen, Z.; Shin, D.M. Nanoparticle therapeutics: An emerging treatment modality for cancer. Nat. Rev. Drug Discov. 2008, 7, 771–782. [Google Scholar] [CrossRef]
- Chauhan, V.P.; Jain, R.K. Strategies for advancing cancer nanomedicine. Nat. Mater. 2013, 12, 958–962. [Google Scholar] [CrossRef]
- Salvioni, L.; Rizzuto, M.A.; Bertolini, J.A.; Pandolfi, L.; Colombo, M.; Prosperi, D. Thirty Years of Cancer Nanomedicine: Success, Frustration, and Hope. Cancers 2019, 11, 1855. [Google Scholar] [CrossRef]
- Xu, M.; Han, X.; Xiong, H.; Gao, Y.; Xu, B.; Zhu, G.; Li, J. Cancer Nanomedicine: Emerging Strategies and Therapeutic Potentials. Molecules 2023, 28, 5145. [Google Scholar] [CrossRef] [PubMed]
- Feldman, E.J.; Lancet, J.E.; Kolitz, J.E.; Ritchie, E.K.; Roboz, G.J.; List, A.F.; Allen, S.L.; Asatiani, E.; Mayer, L.D.; Swenson, C.; et al. First-in-man study of CPX-351: A liposomal carrier containing cytarabine and daunorubicin in a fixed 5:1 molar ratio for the treatment of relapsed and refractory acute myeloid leukemia. J. Clin. Oncol. 2011, 29, 979–985. [Google Scholar] [CrossRef] [PubMed]
- Kobayashi, T.; Tsukagoshi, S.; Sakurai, Y. Enhancement of the cancer chemotherapeutic effect of cytosine arabinoside entrapped in liposomes on mouse leukemia L-1210. Gan 1975, 66, 719–720. [Google Scholar] [PubMed]
- Mayhew, E.; Papahadjopoulos, D.; Rustum, Y.M.; Dave, C. Inhibition of tumor cell growth in vitro and in vivo by 1-beta-D-arabinofuranosylcytosine entrapped within phospholipid vesicles. Cancer Res. 1976, 36, 4406–4411. [Google Scholar]
- Lancet, J.E.; Uy, G.L.; Newell, L.F.; Lin, T.L.; Ritchie, E.K.; Stuart, R.K.; Strickland, S.A.; Hogge, D.; Solomon, S.R.; Bixby, D.L.; et al. CPX-351 versus 7+3 cytarabine and daunorubicin chemotherapy in older adults with newly diagnosed high-risk or secondary acute myeloid leukaemia: 5-year results of a randomised, open-label, multicentre, phase 3 trial. Lancet Haematol. 2021, 8, e481–e491. [Google Scholar] [CrossRef]
- Drinković, N.; Beus, M.; Barbir, R.; Debeljak, Ž.; Tariba Lovaković, B.; Kalčec, N.; Ćurlin, M.; Bekavac, A.; Gorup, D.; Mamić, I.; et al. Novel PLGA-based nanoformulation decreases doxorubicin-induced cardiotoxicity. Nanoscale 2024, 16, 9412–9425. [Google Scholar] [CrossRef]
- Hans, M.L.; Lowman, A.M. Biodegradable nanoparticles for drug delivery and targeting. Curr. Opin. Solid State Mater. Sci. 2002, 6, 319–327. [Google Scholar] [CrossRef]
- Gagliardi, A.; Giuliano, E.; Venkateswararao, E.; Fresta, M.; Bulotta, S.; Awasthi, V.; Cosco, D. Biodegradable Polymeric Nanoparticles for Drug Delivery to Solid Tumors. Front. Pharmacol. 2021, 12, 601626. [Google Scholar] [CrossRef]
- Wiwanitkit, V. Biodegradable Nanoparticles for Drug Delivery and Targeting. In Surface Modification of Nanoparticles for Targeted Drug Delivery; Springer: Cham, Switzerland, 2019; pp. 167–181. [Google Scholar] [CrossRef]
- Alvi, M.; Yaqoob, A.; Rehman, K.; Shoaib, S.M.; Akash, M.S.H. PLGA-based nanoparticles for the treatment of cancer: Current strategies and perspectives. AAPS Open 2022, 8, 12. [Google Scholar] [CrossRef]
- Sönksen, M.; Kerl, K.; Bunzen, H. Current status and future prospects of nanomedicine for arsenic trioxide delivery to solid tumors. Med. Res. Rev. 2022, 42, 374–398. [Google Scholar] [CrossRef]
- Houshmand, M.; Garello, F.; Circosta, P.; Stefania, R.; Aime, S.; Saglio, G.; Giachino, C. Nanocarriers as Magic Bullets in the Treatment of Leukemia. Nanomaterials 2020, 10, 276. [Google Scholar] [CrossRef] [PubMed]
- Alimoghaddam, K. A review of arsenic trioxide and acute promyelocytic leukemia. Int. J. Hematol. Oncol. Stem Cell Res. 2014, 8, 44–54. [Google Scholar] [PubMed]
- Jiang, Y.; Shen, X.; Zhi, F.; Wen, Z.; Gao, Y.; Xu, J.; Yang, B.; Bai, Y. An overview of arsenic trioxide-involved combined treatment algorithms for leukemia: Basic concepts and clinical implications. Cell Death Discov. 2023, 9, 266. [Google Scholar] [CrossRef] [PubMed]
- Fonseca-Santos, B.; da Silva, P.B.; Eloy, J.O.; Chorilli, M. Nanocarriers for the Diagnosis and Treatment of Cancer. In Nanocarriers for Drug Delivery: Concepts and Applications; Eloy, J.O., Abriata, J.P., Marchetti, J.M., Eds.; Springer International Publishing: Cham, Switzerland, 2021; pp. 223–252. [Google Scholar] [CrossRef]
- Acuña Cruz, E.; Cannata Ortiz, J.; García-Noblejas, A.; Alegre, A.; Arranz Sáez, R. Liposomal Doxorubicin in Aggressive B Cell Lymphoma Has Similar Efficacy to the Conventional Formulation: Results from a Retrospective Cohort Study. Blood 2015, 126, 5106. [Google Scholar] [CrossRef]
- Matsumura, Y.; Kataoka, K. Preclinical and clinical studies of anticancer agent-incorporating polymer micelles. Cancer Sci. 2009, 100, 572–579. [Google Scholar] [CrossRef]
- Zhang, Y.; Huang, Y.; Li, S. Polymeric Micelles: Nanocarriers for Cancer-Targeted Drug Delivery. AAPS PharmSciTech 2014, 15, 862–871. [Google Scholar] [CrossRef]
- Hari, S.K.; Gauba, A.; Shrivastava, N.; Tripathi, R.M.; Jain, S.K.; Pandey, A.K. Polymeric micelles and cancer therapy: An ingenious multimodal tumor-targeted drug delivery system. Drug Deliv. Transl. Res. 2023, 13, 135–163. [Google Scholar] [CrossRef]
- Waheed, I.; Ali, A.; Tabassum, H.; Khatoon, N.; Lai, W.F.; Zhou, X. Lipid-based nanoparticles as drug delivery carriers for cancer therapy. Front. Oncol. 2024, 14, 1296091. [Google Scholar] [CrossRef]
- Adamo, F.M.; De Falco, F.; Dorillo, E.; Sorcini, D.; Stella, A.; Esposito, A.; Arcaleni, R.; Rosati, E.; Sportoletti, P. Nanotechnology Advances in the Detection and Treatment of Lymphoid Malignancies. Int. J. Mol. Sci. 2024, 25, 9253. [Google Scholar] [CrossRef]
- Minai, L.; Yeheskely-Hayon, D.; Yelin, D. High levels of reactive oxygen species in gold nanoparticle-targeted cancer cells following femtosecond pulse irradiation. Sci. Rep. 2013, 3, 2146. [Google Scholar] [CrossRef]
- Jain, S.; Hirst, D.G.; O’Sullivan, J.M. Gold nanoparticles as novel agents for cancer therapy. Br. J. Radiol. 2012, 85, 101–113. [Google Scholar] [CrossRef] [PubMed]
- Huang, X.; El-Sayed, I.H.; Qian, W.; El-Sayed, M.A. Cancer cell imaging and photothermal therapy in the near-infrared region by using gold nanorods. J. Am. Chem. Soc. 2006, 128, 2115–2120. [Google Scholar] [CrossRef] [PubMed]
- Zhang, C.; Kuo, J.C.; Huang, Y.; Hu, Y.; Deng, L.; Yung, B.C.; Zhao, X.; Zhang, Z.; Pan, J.; Ma, Y.; et al. Optimized Liposomal Delivery of Bortezomib for Advancing Treatment of Multiple Myeloma. Pharmaceutics 2023, 15, 2674. [Google Scholar] [CrossRef] [PubMed]
- Ashley, J.D.; Stefanick, J.F.; Schroeder, V.A.; Suckow, M.A.; Kiziltepe, T.; Bilgicer, B. Liposomal Bortezomib Nanoparticles via Boronic Ester Prodrug Formulation for Improved Therapeutic Efficacy in Vivo. J. Med. Chem. 2014, 57, 5282–5292. [Google Scholar] [CrossRef]
- Liu, J.; Zhao, R.; Jiang, X.; Li, Z.; Zhang, B. Progress on the Application of Bortezomib and Bortezomib-Based Nanoformulations. Biomolecules 2021, 12, 51. [Google Scholar] [CrossRef]
- Federico, C.; Alhallak, K.; Sun, J.; Duncan, K.; Azab, F.; Sudlow, G.P.; de la Puente, P.; Muz, B.; Kapoor, V.; Zhang, L.; et al. Tumor microenvironment-targeted nanoparticles loaded with bortezomib and ROCK inhibitor improve efficacy in multiple myeloma. Nat. Commun. 2020, 11, 6037. [Google Scholar] [CrossRef]
- Cholujova, D.; Koklesova, L.; Lukacova Bujnakova, Z.; Dutkova, E.; Valuskova, Z.; Beblava, P.; Matisova, A.; Sedlak, J.; Jakubikova, J. In vitro and ex vivo anti-myeloma effects of nanocomposite As4S4/ZnS/Fe3O4. Sci. Rep. 2022, 12, 17961. [Google Scholar] [CrossRef]
- Dickerson, E.B.; Dreaden, E.C.; Huang, X.; El-Sayed, I.H.; Chu, H.; Pushpanketh, S.; McDonald, J.F.; El-Sayed, M.A. Gold nanorod assisted near-infrared plasmonic photothermal therapy (PPTT) of squamous cell carcinoma in mice. Cancer Lett. 2008, 269, 57–66. [Google Scholar] [CrossRef]
- Argyriou, A.A.; Iconomou, G.; Kalofonos, H.P. Bortezomib-induced peripheral neuropathy in multiple myeloma: A comprehensive review of the literature. Blood 2008, 112, 1593–1599. [Google Scholar] [CrossRef]
- Boccadoro, M.; Morgan, G.; Cavenagh, J. Preclinical evaluation of the proteasome inhibitor bortezomib in cancer therapy. Cancer Cell Int. 2005, 5, 18. [Google Scholar] [CrossRef]
- Liu, Z.; Shen, H.; Liu, H.; Ding, K.; Song, J.; Zhang, J.; Ding, D.; Fu, R. Advancements in drugs restructured with nanomedicines for multiple myeloma treatment. Sci. China Mater. 2024, 67, 3780–3795. [Google Scholar] [CrossRef]
- Zheleznyak, A.; Shokeen, M.; Achilefu, S. Nanotherapeutics for multiple myeloma. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol 2018, 10, e1526. [Google Scholar] [CrossRef] [PubMed]
- Li, S.; Wang, H.; Xiong, S.; Liu, J.; Sun, S. Targeted Delivery Strategies for Multiple Myeloma and Their Adverse Drug Reactions. Pharmaceuticals 2024, 17, 832. [Google Scholar] [CrossRef] [PubMed]
- Yang, M.; Chen, Y.; Zhu, L.; You, L.; Tong, H.; Meng, H.; Sheng, J.; Jin, J. Harnessing Nanotechnology: Emerging Strategies for Multiple Myeloma Therapy. Biomolecules 2024, 14, 83. [Google Scholar] [CrossRef]
- Chauhan, A.S. Dendrimer nanotechnology for enhanced formulation and controlled delivery of resveratrol. Ann. N. Y. Acad. Sci. 2015, 1348, 134–140. [Google Scholar] [CrossRef]
- Silverman, J.A.; Deitcher, S.R. Marqibo® (vincristine sulfate liposome injection) improves the pharmacokinetics and pharmacodynamics of vincristine. Cancer Chemother. Pharmacol. 2013, 71, 555–564. [Google Scholar] [CrossRef]
- Silverman, J.A.; Reynolds, L.; Deitcher, S.R. Pharmacokinetics and pharmacodynamics of vincristine sulfate liposome injection (VSLI) in adults with acute lymphoblastic leukemia. J. Clin. Pharmacol. 2013, 53, 1139–1145. [Google Scholar] [CrossRef]
- Sasaki, K.; Jabbour, E.J.; Khouri, M.; Thomas, D.A.; Garcia-Manero, G.; Ravandi, F.; Borthakur, G.; Short, N.J.; Issa, G.C.; Kadia, T.; et al. Phase II Study of Hyper-Cmad with Liposomal Vincristine (Marqibo) for Patients with Newly Diagnosed Acute Lymphoblastic Leukemia (ALL). Blood 2017, 130, 2554. [Google Scholar] [CrossRef]
- Goli, N.; Bolla, P.K.; Talla, V. Antibody-drug conjugates (ADCs): Potent biopharmaceuticals to target solid and hematological cancers- an overview. J. Drug Deliv. Sci. Technol. 2018, 48, 106–117. [Google Scholar] [CrossRef]
- Gébleux, R.; Casi, G. Antibody-drug conjugates: Current status and future perspectives. Pharmacol. Ther. 2016, 167, 48–59. [Google Scholar] [CrossRef]
- Younes, A.; Bartlett, N.L.; Leonard, J.P.; Kennedy, D.A.; Lynch, C.M.; Sievers, E.L.; Forero-Torres, A. Brentuximab vedotin (SGN-35) for relapsed CD30-positive lymphomas. N. Engl. J. Med. 2010, 363, 1812–1821. [Google Scholar] [CrossRef] [PubMed]
- Mura, S.; Couvreur, P. Nanotheranostics for personalized medicine. Adv. Drug Deliv. Rev. 2012, 64, 1394–1416. [Google Scholar] [CrossRef] [PubMed]
- Hristova-Panusheva, K.; Xenodochidis, C.; Georgieva, M.; Krasteva, N. Nanoparticle-Mediated Drug Delivery Systems for Precision Targeting in Oncology. Pharmaceuticals 2024, 17, 677. [Google Scholar] [CrossRef] [PubMed]
- Torchilin, V.P. Passive and Active Drug Targeting: Drug Delivery to Tumors as an Example. In Drug Delivery; Schäfer-Korting, M., Ed.; Springer: Berlin/Heidelberg, Germany, 2010; pp. 3–53. [Google Scholar] [CrossRef]
- Etrych, T.; Braunova, A.; Zogala, D.; Lambert, L.; Renesova, N.; Klener, P. Targeted Drug Delivery and Theranostic Strategies in Malignant Lymphomas. Cancers 2022, 14, 626. [Google Scholar] [CrossRef]
- Jiang, Y.; Lin, W.; Zhu, L. Targeted Drug Delivery for the Treatment of Blood Cancers. Molecules 2022, 27, 1310. [Google Scholar] [CrossRef]
- Bandyopadhyay, A.; Das, T.; Nandy, S.; Sahib, S.; Preetam, S.; Gopalakrishnan, A.V.; Dey, A. Ligand-based active targeting strategies for cancer theranostics. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2023, 396, 3417–3441. [Google Scholar] [CrossRef]
- Wang, K.; Wei, G.; Liu, D. CD19: A biomarker for B cell development, lymphoma diagnosis and therapy. Exp. Hematol. Oncol. 2012, 1, 36. [Google Scholar] [CrossRef]
- Horna, P.; Nowakowski, G.; Endell, J.; Boxhammer, R. Comparative Assessment of Surface CD19 and CD20 Expression on B-Cell Lymphomas from Clinical Biopsies: Implications for Targeted Therapies. Blood 2019, 134, 5345. [Google Scholar] [CrossRef]
- Morandi, F.; Horenstein, A.L.; Costa, F.; Giuliani, N.; Pistoia, V.; Malavasi, F. CD38: A Target for Immunotherapeutic Approaches in Multiple Myeloma. Front. Immunol. 2018, 9, 2722. [Google Scholar] [CrossRef]
- Sun, H.; Zhu, X.; Lu, P.Y.; Rosato, R.R.; Tan, W.; Zu, Y. Oligonucleotide aptamers: New tools for targeted cancer therapy. Mol. Ther. Nucleic Acids 2014, 3, e182. [Google Scholar] [CrossRef]
- Zhu, G.; Chen, X. Aptamer-based targeted therapy. Adv. Drug Deliv. Rev. 2018, 134, 65–78. [Google Scholar] [CrossRef] [PubMed]
- Mahmoudian, F.; Ahmari, A.; Shabani, S.; Sadeghi, B.; Fahimirad, S.; Fattahi, F. Aptamers as an approach to targeted cancer therapy. Cancer Cell Int. 2024, 24, 108. [Google Scholar] [CrossRef] [PubMed]
- Agiba, A.M.; Arreola-Ramírez, J.L.; Carbajal, V.; Segura-Medina, P. Light-Responsive and Dual-Targeting Liposomes: From Mechanisms to Targeting Strategies. Molecules 2024, 29, 636. [Google Scholar] [CrossRef]
- McCallion, C.; Peters, A.D.; Booth, A.; Rees-Unwin, K.; Adams, J.; Rahi, R.; Pluen, A.; Hutchinson, C.V.; Webb, S.J.; Burthem, J. Dual-action CXCR4-targeting liposomes in leukemia: Function blocking and drug delivery. Blood Adv. 2019, 3, 2069–2081. [Google Scholar] [CrossRef]
- Murugan, B.; Sagadevan, S.; Fatimah, I.; Oh, W.-C.; Motalib Hossain, M.A.; Johan, M.R. Smart stimuli-responsive nanocarriers for the cancer therapy—Nanomedicine. Nanotechnol. Rev. 2021, 10, 933–953. [Google Scholar] [CrossRef]
- Zhang, Q.; Kuang, G.; Li, W.; Wang, J.; Ren, H.; Zhao, Y. Stimuli-Responsive Gene Delivery Nanocarriers for Cancer Therapy. Nanomicro Lett. 2023, 15, 44. [Google Scholar] [CrossRef]
- Ganta, S.; Devalapally, H.; Shahiwala, A.; Amiji, M. A review of stimuli-responsive nanocarriers for drug and gene delivery. J. Control. Release 2008, 126, 187–204. [Google Scholar] [CrossRef]
- Majumder, J.; Minko, T. Multifunctional and stimuli-responsive nanocarriers for targeted therapeutic delivery. Expert Opin. Drug Deliv. 2021, 18, 205–227. [Google Scholar] [CrossRef]
- Mura, S.; Nicolas, J.; Couvreur, P. Stimuli-responsive nanocarriers for drug delivery. Nat. Mater. 2013, 12, 991–1003. [Google Scholar] [CrossRef]
- Sullivan, H.L.; Liang, Y.; Worthington, K.; Luo, C.; Gianneschi, N.C.; Christman, K.L. Enzyme-Responsive Nanoparticles for the Targeted Delivery of an MMP Inhibitor to Acute Myocardial Infarction. Biomacromolecules 2023, 24, 4695–4704. [Google Scholar] [CrossRef]
- Abed, H.F.; Abuwatfa, W.H.; Husseini, G.A. Redox-Responsive Drug Delivery Systems: A Chemical Perspective. Nanomaterials 2022, 12, 3183. [Google Scholar] [CrossRef]
- Meng, X.; Shen, Y.; Zhao, H.; Lu, X.; Wang, Z.; Zhao, Y. Redox-manipulating nanocarriers for anticancer drug delivery: A systematic review. J. Nanobiotechnol. 2024, 22, 587. [Google Scholar] [CrossRef]
- Jordan, A.; Scholz, R.; Wust, P.; Fähling, H.; Roland, F. Magnetic fluid hyperthermia (MFH): Cancer treatment with AC magnetic field induced excitation of biocompatible superparamagnetic nanoparticles. J. Magn. Magn. Mater. 1999, 201, 413–419. [Google Scholar] [CrossRef]
- Fatima, H.; Charinpanitkul, T.; Kim, K.S. Fundamentals to Apply Magnetic Nanoparticles for Hyperthermia Therapy. Nanomaterials 2021, 11, 1203. [Google Scholar] [CrossRef]
- Dutz, S.; Hergt, R. Magnetic nanoparticle heating and heat transfer on a microscale: Basic principles, realities and physical limitations of hyperthermia for tumour therapy. Int. J. Hyperth. 2013, 29, 790–800. [Google Scholar] [CrossRef]
- Li, Y.; Li, X.; Zhou, F.; Doughty, A.; Hoover, A.R.; Nordquist, R.E.; Chen, W.R. Nanotechnology-based photoimmunological therapies for cancer. Cancer Lett. 2019, 442, 429–438. [Google Scholar] [CrossRef]
- Zhou, F.; Hasan, T.; Frochot, C.; Chen, W.R. Nanotechnology, photonics, and immunotherapy for cancer diagnostics and therapeutics. Nanophotonics 2021, 10, 2969–2971. [Google Scholar] [CrossRef]
- Li, G.; Wang, C.; Jin, B.; Sun, T.; Sun, K.; Wang, S.; Fan, Z. Advances in smart nanotechnology-supported photodynamic therapy for cancer. Cell Death Discov. 2024, 10, 466. [Google Scholar] [CrossRef]
- Shakil, M.S.; Niloy, M.S.; Mahmud, K.M.; Kamal, M.A.; Islam, M.A. Theranostic Potentials of Gold Nanomaterials in Hematological Malignancies. Cancers 2022, 14, 3047. [Google Scholar] [CrossRef]
- Chauhan, A.S. Dendrimers for Drug Delivery. Molecules 2018, 23, 938. [Google Scholar] [CrossRef]
- Zhu, W.; Guo, J.; Agola, J.; Croissant, J.; Wang, Z.; Shang, J.; Coker, E.; Motevalli, B.; Zimpel, A.; Wuttke, S.; et al. Metal-Organic Framework Nanoparticle-Assisted Cryopreservation of Red Blood Cells. J. Am. Chem. Soc. 2019, 141, 7789–7796. [Google Scholar] [CrossRef]
- Wang, R.; Gao, J.; Vijayalakshmi, M.; Tang, H.; Chen, K.; Reddy, C.V.; Kakarla, R.R.; Anjana, P.M.; Rezakazemi, M.; Cheolho, B.; et al. Metal–organic frameworks and their composites: Design, synthesis, properties, and energy storage applications. Chem. Eng. J. 2024, 496, 154294. [Google Scholar] [CrossRef]
- Vavassori, V.; Ferrari, S.; Beretta, S.; Asperti, C.; Albano, L.; Annoni, A.; Gaddoni, C.; Varesi, A.; Soldi, M.; Cuomo, A.; et al. Lipid nanoparticles allow efficient and harmless ex vivo gene editing of human hematopoietic cells. Blood 2023, 142, 812–826. [Google Scholar] [CrossRef]
- Su, K.; Shi, L.; Sheng, T.; Yan, X.; Lin, L.; Meng, C.; Wu, S.; Chen, Y.; Zhang, Y.; Wang, C.; et al. Reformulating lipid nanoparticles for organ-targeted mRNA accumulation and translation. Nat. Commun. 2024, 15, 5659. [Google Scholar] [CrossRef]
- Djayanti, K.; Maharjan, P.; Cho, K.H.; Jeong, S.; Kim, M.S.; Shin, M.C.; Min, K.A. Mesoporous Silica Nanoparticles as a Potential Nanoplatform: Therapeutic Applications and Considerations. Int. J. Mol. Sci. 2023, 24, 6349. [Google Scholar] [CrossRef]
- Houshmand, F.; Schofield, J.; Moafi, Z. Electronic and structural properties of functionalized Silica nanoparticles: DFT and SCC DFTB calculation. Res. Sq. 2023. [Google Scholar] [CrossRef]
- Aljabali, A.A.; Rezigue, M.; Alsharedeh, R.H.; Obeid, M.A.; Mishra, V.; Serrano-Aroca, Á.; El-Tanani, M.; Tambuwala, M.M. Protein-based nanomaterials: A new tool for targeted drug delivery. Ther. Deliv. 2022, 13, 321–338. [Google Scholar] [CrossRef]
- Kaltbeitzel, J.; Wich, P.R. Protein-based Nanoparticles: From Drug Delivery to Imaging, Nanocatalysis and Protein Therapy. Angew. Chem. Int. Ed. 2023, 62, e202216097. [Google Scholar] [CrossRef]
- Thakare, V.S.; Das, M.; Jain, A.K.; Patil, S.; Jain, S. Carbon nanotubes in cancer theragnosis. Nanomedicine 2010, 5, 1277–1301. [Google Scholar] [CrossRef]
- Huang, J.Y.; Chen, S.; Wang, Z.Q.; Kempa, K.; Wang, Y.M.; Jo, S.H.; Chen, G.; Dresselhaus, M.S.; Ren, Z.F. Superplastic carbon nanotubes. Nature 2006, 439, 281. [Google Scholar] [CrossRef]
- Jain, T.K.; Reddy, M.K.; Morales, M.A.; Leslie-Pelecky, D.L.; Labhasetwar, V. Biodistribution, clearance, and biocompatibility of iron oxide magnetic nanoparticles in rats. Mol. Pharm. 2008, 5, 316–327. [Google Scholar] [CrossRef]
- Ahmed, M.; Douek, M. The role of magnetic nanoparticles in the localization and treatment of breast cancer. Biomed. Res. Int. 2013, 2013, 281230. [Google Scholar] [CrossRef]
- Patel, A.; Patel, K.; Patel, V.; Rajput, M.S.; Patel, R.; Rajput, A. Nanocrystals: An emerging paradigm for cancer therapeutics. Future J. Pharm. Sci. 2024, 10, 4. [Google Scholar] [CrossRef]
- Straus, D.J.; Długosz-Danecka, M.; Connors, J.M.; Alekseev, S.; Illés, Á.; Picardi, M.; Lech-Maranda, E.; Feldman, T.; Smolewski, P.; Savage, K.J.; et al. Brentuximab vedotin with chemotherapy for stage III or IV classical Hodgkin lymphoma (ECHELON-1): 5-year update of an international, open-label, randomised, phase 3 trial. Lancet Haematol. 2021, 8, e410–e421. [Google Scholar] [CrossRef]
- Borchmann, P.; Ferdinandus, J.; Schneider, G.; Moccia, A.; Greil, R.; Hertzberg, M.; Schaub, V.; Hüttmann, A.; Keil, F.; Dierlamm, J.; et al. Assessing the efficacy and tolerability of PET-guided BrECADD versus eBEACOPP in advanced-stage, classical Hodgkin lymphoma (HD21): A randomised, multicentre, parallel, open-label, phase 3 trial. Lancet 2024, 404, 341–352. [Google Scholar] [CrossRef]
- Cucinotto, I.; Fiorillo, L.; Gualtieri, S.; Arbitrio, M.; Ciliberto, D.; Staropoli, N.; Grimaldi, A.; Luce, A.; Tassone, P.; Caraglia, M.; et al. Nanoparticle albumin bound Paclitaxel in the treatment of human cancer: Nanodelivery reaches prime-time? J. Drug Deliv. 2013, 2013, 905091. [Google Scholar] [CrossRef]
- Giordano, G.; Pancione, M.; Olivieri, N.; Parcesepe, P.; Velocci, M.; Di Raimo, T.; Coppola, L.; Toffoli, G.; D’Andrea, M.R. Nano albumin bound-paclitaxel in pancreatic cancer: Current evidences and future directions. World J. Gastroenterol. 2017, 23, 5875–5886. [Google Scholar] [CrossRef]
- Hassan, M.S.; Awasthi, N.; Ponna, S.; von Holzen, U. Nab-Paclitaxel in the Treatment of Gastrointestinal Cancers—Improvements in Clinical Efficacy and Safety. Biomedicines 2023, 11, 2000. [Google Scholar] [CrossRef]
- Rosi, N.L.; Giljohann, D.A.; Thaxton, C.S.; Lytton-Jean, A.K.R.; Han, M.S.; Mirkin, C.A. Oligonucleotide-Modified Gold Nanoparticles for Intracellular Gene Regulation. Science 2006, 312, 1027–1030. [Google Scholar] [CrossRef]
- Bavelaar, B.M.; Song, L.; Jackson, M.R.; Able, S.; Tietz, O.; Skaripa-Koukelli, I.; Waghorn, P.A.; Gill, M.R.; Carlisle, R.C.; Tarsounas, M.; et al. Oligonucleotide-Functionalized Gold Nanoparticles for Synchronous Telomerase Inhibition, Radiosensitization, and Delivery of Theranostic Radionuclides. Mol. Pharm. 2021, 18, 3820–3831. [Google Scholar] [CrossRef]
- Kaplan, L.D.; Deitcher, S.R.; Silverman, J.A.; Morgan, G. Phase II Study of Vincristine Sulfate Liposome Injection (Marqibo) and Rituximab for Patients With Relapsed and Refractory Diffuse Large B-Cell Lymphoma or Mantle Cell Lymphoma in Need of Palliative Therapy. Clin. Lymphoma Myeloma Leuk. 2014, 14, 37–42. [Google Scholar] [CrossRef]
- Silverman, J.A.; Aulitzky, W.E.; Lister, J.; Maness, L.; Schiller, G.J.; Seiter, K.; Smith, S.; Stock, W.; Yehuda, D.B.; Deitcher, S.R. Marqibo® (vincristine sulfate liposomes injection; VSLI) Optimizes the Dosing, Delivery, and Pharmacokinetic (PK) Profile of Vincristine Sulfate (VCR) In Adults with Relapsed and Refractory Acute Lymphoblastic Leukemia (ALL). Blood 2010, 116, 2142. [Google Scholar] [CrossRef]
- Hammami, I.; Alabdallah, N.M.; Jomaa, A.A.; Kamoun, M. Gold nanoparticles: Synthesis properties and applications. J. King Saud Univ.-Sci. 2021, 33, 101560. [Google Scholar] [CrossRef]
- Mikhailova, E.O. Gold Nanoparticles: Biosynthesis and Potential of Biomedical Application. J. Funct. Biomater. 2021, 12, 70. [Google Scholar] [CrossRef]
- Szebeni, J. Complement activation-related pseudoallergy: A new class of drug-induced acute immune toxicity. Toxicology 2005, 216, 106–121. [Google Scholar] [CrossRef]
- Etheridge, M.L.; Campbell, S.A.; Erdman, A.G.; Haynes, C.L.; Wolf, S.M.; McCullough, J. The big picture on nanomedicine: The state of investigational and approved nanomedicine products. Nanomedicine 2013, 9, 1–14. [Google Scholar] [CrossRef]
- Hertig, J.B.; Shah, V.P.; Flühmann, B.; Mühlebach, S.; Stemer, G.; Surugue, J.; Moss, R.; Di Francesco, T. Tackling the challenges of nanomedicines: Are we ready? Am. J. Health Syst. Pharm. 2021, 78, 1047–1056. [Google Scholar] [CrossRef]
- Mühlebach, S. Regulatory challenges of nanomedicines and their follow-on versions: A generic or similar approach? Adv. Drug Deliv. Rev. 2018, 131, 122–131. [Google Scholar] [CrossRef]
- Md Anwar Nawaz, R.; Darul Raiyaan, G.I.; Sivakumar, K.; Arunachalam, K.D. Nanomedicine Regulation and Future Prospects. In Advances in Novel Formulations for Drug Delivery; Scrivener Publishing: Beverly, MA, USA, 2023; pp. 67–80. [Google Scholar] [CrossRef]
- Liu, P.; Chen, G.; Zhang, J. A Review of Liposomes as a Drug Delivery System: Current Status of Approved Products, Regulatory Environments, and Future Perspectives. Molecules 2022, 27, 1372. [Google Scholar] [CrossRef]
- Beltrán-Gracia, E.; López-Camacho, A.; Higuera-Ciapara, I.; Velázquez-Fernández, J.B.; Vallejo-Cardona, A.A. Nanomedicine review: Clinical developments in liposomal applications. Cancer Nanotechnol. 2019, 10, 11. [Google Scholar] [CrossRef]
- Yu, J.; Dan, N.; Eslami, S.M.; Lu, X. State of the Art of Silica Nanoparticles: An Overview on Biodistribution and Preclinical Toxicity Studies. AAPS J. 2024, 26, 35. [Google Scholar] [CrossRef] [PubMed]
- Zhang, R.; Kiessling, F.; Lammers, T.; Pallares, R.M. Clinical translation of gold nanoparticles. Drug Deliv. Transl. Res. 2023, 13, 378–385. [Google Scholar] [CrossRef] [PubMed]
- Cheng, Z.; Li, Y.; Zhao, D.; Zhao, W.; Wu, M.; Zhang, W.; Cui, Y.; Zhang, P.; Zhang, Z. Nanocarriers for intracellular co-delivery of proteins and small-molecule drugs for cancer therapy. Front. Bioeng. Biotechnol. 2022, 10, 994655. [Google Scholar] [CrossRef] [PubMed]
- Rana, I.; Oh, J.; Baig, J.; Moon, J.H.; Son, S.; Nam, J. Nanocarriers for cancer nano-immunotherapy. Drug Deliv. Transl. Res. 2023, 13, 1936–1954. [Google Scholar] [CrossRef]
- Fadilah, N.I.; Isa, I.L.; Zaman, W.S.; Tabata, Y.; Fauzi, M.B. The Effect of Nanoparticle-Incorporated Natural-Based Biomaterials towards Cells on Activated Pathways: A Systematic Review. Polymers 2022, 14, 476. [Google Scholar] [CrossRef]
- Saikia, N. Inorganic-Based Nanoparticles and Biomaterials as Biocompatible Scaffolds for Regenerative Medicine and Tissue Engineering: Current Advances and Trends of Development. Inorganics 2024, 12, 292. [Google Scholar] [CrossRef]
- Wei, Z.; Zhou, Y.; Wang, R.; Wang, J.; Chen, Z. Aptamers as Smart Ligands for Targeted Drug Delivery in Cancer Therapy. Pharmaceutics 2022, 14, 2561. [Google Scholar] [CrossRef]
- Liu, Q.; Jin, C.; Wang, Y.; Fang, X.; Zhang, X.; Chen, Z.; Tan, W. Aptamer-conjugated nanomaterials for specific cancer cell recognition and targeted cancer therapy. NPG Asia Mater. 2014, 6, e95. [Google Scholar] [CrossRef]
- Fu, Z.; Xiang, J. Aptamer-Functionalized Nanoparticles in Targeted Delivery and Cancer Therapy. Int. J. Mol. Sci. 2020, 21, 9123. [Google Scholar] [CrossRef]
- Tiwari, R.; Tiwari, G.; Parashar, P. Theranostics Applications of Functionalized Magnetic Nanoparticles. In Multifunctional And Targeted Theranostic Nanomedicines: Formulation, Design And Applications; Jain, K., Jain, N.K., Eds.; Springer Nature: Singapore, 2023; pp. 361–382. [Google Scholar] [CrossRef]
- Sermer, D.; Elavalakanar, P.; Abramson, J.S.; Palomba, M.L.; Salles, G.; Arnason, J. Targeting CD19 for diffuse large B cell lymphoma in the era of CARs: Other modes of transportation. Blood Rev. 2023, 57, 101002. [Google Scholar] [CrossRef]
- Marvin-Peek, J.; Savani, B.N.; Olalekan, O.O.; Dholaria, B. Challenges and Advances in Chimeric Antigen Receptor Therapy for Acute Myeloid Leukemia. Cancers 2022, 14, 497. [Google Scholar] [CrossRef] [PubMed]
- Qu, Y.; Chu, B.; Wei, X.; Chen, Y.; Yang, Y.; Hu, D.; Huang, J.; Wang, F.; Chen, M.; Zheng, Y.; et al. Cancer-Cell-Biomimetic Nanoparticles for Targeted Therapy of Multiple Myeloma Based on Bone Marrow Homing. Adv. Mater. 2022, 34, e2107883. [Google Scholar] [CrossRef] [PubMed]
- Kazemian, P.; Yu, S.Y.; Thomson, S.B.; Birkenshaw, A.; Leavitt, B.R.; Ross, C.J.D. Lipid-Nanoparticle-Based Delivery of CRISPR/Cas9 Genome-Editing Components. Mol. Pharm. 2022, 19, 1669–1686. [Google Scholar] [CrossRef] [PubMed]
- Ni, H.; Hatit, M.Z.C.; Zhao, K.; Loughrey, D.; Lokugamage, M.P.; Peck, H.E.; Cid, A.D.; Muralidharan, A.; Kim, Y.; Santangelo, P.J.; et al. Piperazine-derived lipid nanoparticles deliver mRNA to immune cells in vivo. Nat. Commun. 2022, 13, 4766. [Google Scholar] [CrossRef]
- Duan, L.; Ouyang, K.; Xu, X.; Xu, L.; Wen, C.; Zhou, X.; Qin, Z.; Xu, Z.; Sun, W.; Liang, Y. Nanoparticle Delivery of CRISPR/Cas9 for Genome Editing. Front. Genet. 2021, 12, 673286. [Google Scholar] [CrossRef]
- Das, K.P.; J, C. Nanoparticles and convergence of artificial intelligence for targeted drug delivery for cancer therapy: Current progress and challenges. Front. Med. Technol. 2022, 4, 1067144. [Google Scholar] [CrossRef]
- Rodríguez-Criado, J.; Quiñonero, F.; Prados, J.; Melguizo, C. Nanoparticles Combining Gene Therapy and Chemotherapy as a Treatment for Gastrointestinal Tumors: A Systematic Review. Appl. Sci. 2024, 14, 7872. [Google Scholar] [CrossRef]
- Wang, A.X.; Ong, X.J.; D’Souza, C.; Neeson, P.J.; Zhu, J.J. Combining chemotherapy with CAR-T cell therapy in treating solid tumors. Front. Immunol. 2023, 14, 1140541. [Google Scholar] [CrossRef]
- Hua, S.; de Matos, M.B.C.; Metselaar, J.M.; Storm, G. Current Trends and Challenges in the Clinical Translation of Nanoparticulate Nanomedicines: Pathways for Translational Development and Commercialization. Front. Pharmacol. 2018, 9, 790. [Google Scholar] [CrossRef]
- Rodríguez, F.; Caruana, P.; De la Fuente, N.; Español, P.; Gámez, M.; Balart, J.; Llurba, E.; Rovira, R.; Ruiz, R.; Martín-Lorente, C.; et al. Nano-Based Approved Pharmaceuticals for Cancer Treatment: Present and Future Challenges. Biomolecules 2022, 12, 784. [Google Scholar] [CrossRef]
- Tinkle, S.; McNeil, S.E.; Mühlebach, S.; Bawa, R.; Borchard, G.; Barenholz, Y.C.; Tamarkin, L.; Desai, N. Nanomedicines: Addressing the scientific and regulatory gap. Ann. N. Y. Acad. Sci. 2014, 1313, 35–56. [Google Scholar] [CrossRef]
Nanoformulation Type | Mechanism of Action | Target Malignancy | Clinical Phase | Key References |
---|---|---|---|---|
Liposomes (e.g., Doxil®) | Passive targeting via EPR | Multiple myeloma, lymphoma | FDA Approved | [53] |
Polymeric Nanoparticles | Controlled release of drugs | Leukemia, multiple myeloma | Preclinical | [99,100] |
Gold Nanoparticles | Active targeting using conjugated antibodies | Lymphoma | Preclinical | [116,165] |
Dendrimers | Multifunctional targeting and co-delivery of drugs | Leukemia, multiple myeloma | Preclinical | [130,166] |
Metal–Organic Frameworks | Enzyme-triggered release in tumor microenvironment | Lymphoma | Preclinical | [167,168] |
pH-Responsive Nanocarriers | pH-sensitive release in acidic tumor environments | Multiple myeloma | Preclinical | [32,86] |
Lipid Nanoparticles (LNPs) | mRNA delivery for gene modulation | Leukemia | Preclinical | [169,170] |
Silica Nanoparticles | Drug encapsulation with high stability and release | Acute lymphoblastic leukemia | Preclinical | [171,172] |
Protein-Based Nanoparticles | Biodegradable carriers for targeted delivery | Lymphoma, leukemia | Preclinical | [173,174] |
Carbon Nanotubes | Dual delivery (chemotherapy + photothermal therapy) | Lymphoma | Preclinical | [175,176] |
Magnetic Nanoparticles | Magnetic field-guided targeting and hyperthermia | Multiple myeloma, lymphoma | Preclinical | [177,178] |
Polymeric Micelles | Solubilization of hydrophobic drugs | Lymphoma | Preclinical | [110,111] |
Nanocrystals | High drug-loading capacity for chemotherapy | Acute myeloid leukemia | Preclinical | [41,179] |
Antibody-Drug Conjugates (ADCs) | Targeted drug delivery through conjugated antibodies | Hodgkin lymphoma | Clinical Trials | [180,181] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Mahmoud, A.M.; Deambrogi, C. Advancements in Nanotechnology for Targeted and Controlled Drug Delivery in Hematologic Malignancies: Shaping the Future of Targeted Therapeutics. Appl. Biosci. 2025, 4, 16. https://doi.org/10.3390/applbiosci4010016
Mahmoud AM, Deambrogi C. Advancements in Nanotechnology for Targeted and Controlled Drug Delivery in Hematologic Malignancies: Shaping the Future of Targeted Therapeutics. Applied Biosciences. 2025; 4(1):16. https://doi.org/10.3390/applbiosci4010016
Chicago/Turabian StyleMahmoud, Abdurraouf Mokhtar, and Clara Deambrogi. 2025. "Advancements in Nanotechnology for Targeted and Controlled Drug Delivery in Hematologic Malignancies: Shaping the Future of Targeted Therapeutics" Applied Biosciences 4, no. 1: 16. https://doi.org/10.3390/applbiosci4010016
APA StyleMahmoud, A. M., & Deambrogi, C. (2025). Advancements in Nanotechnology for Targeted and Controlled Drug Delivery in Hematologic Malignancies: Shaping the Future of Targeted Therapeutics. Applied Biosciences, 4(1), 16. https://doi.org/10.3390/applbiosci4010016