All content on this site is intended for healthcare professionals only. By acknowledging this message and accessing the information on this website you are confirming that you are a Healthcare Professional. If you are a patient or carer, please visit Know AML.

The AML Hub uses cookies on this website. They help us give you the best online experience. By continuing to use our website without changing your cookie settings, you agree to our use of cookies in accordance with our updated Cookie Policy

Introducing

Now you can personalise
your AML Hub experience!

Bookmark content to read later

Select your specific areas of interest

View content recommended for you

Find out more
  TRANSLATE

The AML Hub website uses a third-party service provided by Google that dynamically translates web content. Translations are machine generated, so may not be an exact or complete translation, and the AML Hub cannot guarantee the accuracy of translated content. The AML Hub and its employees will not be liable for any direct, indirect, or consequential damages (even if foreseeable) resulting from use of the Google Translate feature. For further support with Google Translate, visit Google Translate Help.

Steering CommitteeAbout UsNewsletterContact
LOADING
You're logged in! Click here any time to manage your account or log out.
LOADING
You're logged in! Click here any time to manage your account or log out.
2024-08-12T12:17:19.000Z

Liposomal drug delivery in acute myeloid leukemia


After reading, you'll be able to:
  • discuss liposomal drug delivery and its application in the treatment of acute myeloid leukemia.
Share:
 0 minute

Bookmark this article

Test your knowledge! Take our quick quiz before and after you read this article to find out if you improved your knowledge. Results help us to improve content and continually provide open-access education.

Acute myeloid leukemia (AML) is a heterogenous, aggressive myeloid malignancy, characterized by proliferation of blast cells and disruption of normal hematopoiesis.1,2 Despite considerable advances in understanding the pathobiology of AML, outcomes remain poor at the population level.2

Challenges with current treatment

Current clinical therapies for AML include chemotherapy, immunotherapy, targeted agents, and hematopoietic stem cell transplantation (HSCT).1 The “7+3 regimen”, a continuous 7-day infusion of cytarabine and a 3-day infusion of anthracycline (idarubicin or daunorubicin), has historically been the mainstay of induction treatment over the past 40 years1,3; however, intensive chemotherapy regimens used in the induction and consolidation stages are not suitable for all patients, especially those with advanced age, secondary AML, serious underlying diseases, or poor physical fitness.1 Moreover, chemoresistance is becoming an increasing issue due to the heterogeneity and complexity of bone marrow.1 Therefore, therapeutic agents such as hypomethylating agents, B-cell lymphoma 2 (Bcl-2) inhibitor, isocitrate dehydrogenase 1 (IDH1) inhibitors, IDH2 inhibitors, and FMS-like tyrosine kinase 3 (FLT3) inhibitors have emerged as standard treatment approaches and are approved for the management of patients with AML. Despite these advancements, significant unmet needs remain, including drug toxicity, drug resistance, disease relapse, off-target adverse effects, and graft-versus-host disease.1

Liposomal drug delivery

To address current treatment gaps in AML, novel nanotechnologies—including liposomes, metallic nanoparticles, dendrimers, micelles, lipid nanoparticles, and polymeric nanoparticles—have emerged as promising approaches.3 Notably, liposomes are the first nanomedicine delivery system to progress from concept to clinical application and have been successfully adopted in clinical settings.1

Liposomes are spherical vesicles comprising phospholipids and cholesterol in a bilayer arrangement.3,4 Due to their amphiphilic nature, liposomes are capable of delivering both hydrophobic drugs (embedded within the phospholipid bilayer) and hydrophilic drugs (encased in the core).4 Figure 1 illustrates the basic structure of liposomes and their various types, and the potential benefits of liposomes are depicted in Figure 2


Figure 3 illustrates the mechanism of action of liposomal drug delivery systems. Liposomes can be functionalized using various methods, including antibodies, peptides, aptamers, proteins, small molecules, and hyaluronan, as shown in Figure 1, which can bind to specific receptors or antigens on leukemia cells and penetrate them. Liposomes can evade immune cell attacks by camouflaging plasma membranes from normal or leukemia cells, enhancing their uptake by leukemia cells through membrane fusion and increasing their targeting ability.1

Approved liposomal drug formulations for AML

CPX-3515

The first liposomal products, such as those for daunorubicin or doxorubicin hydrochloride, were formulated with high cholesterol and encapsulated only a single agent.5 While these single-agent formulations had distinct pharmacokinetic profiles, the required fixed synergistic ratio could not be retained when administration was combined and manipulation of the liposomal composition was required to maintain a synergistic molar ratio for extended periods in vivo.5 To address this, CPX-351, a dual-drug low-cholesterol liposomal encapsulation of daunorubicin and cytarabine, was rationally designed.5

The structure of CPX-351 is shown in Figure 4. The liposome membrane is composed of distearoylphosphatidylcholine (DSPC), distearoylphosphatidylglycerol (DSPG), and cholesterol (CHOL) in a 7:2:1 molar ratio, giving the liposome a high melting point. It allows the liposomes to remain in a gel phase at body temperature and maintain the synergistic drug ratio in vivo for >24 hours post intravenous administration.5 The low level of cholesterol in the membrane helps in retaining hydrophilic substances and stabilizing the lipid bilayer. Cytarabine and daunorubicin are co-encapsulated in the liposome’s aqueous layer at a molar ratio of 5:1.5 Cytarabine is passively encapsulated into the preformed liposomes using copper gluconate buffer, while daunorubicin is subsequently actively encapsulated through complexation with the intra-liposomal copper at a ratio of 1:1 to 1:2, which helps to retain both drugs effectively within the liposome.5

CPX-351 has demonstrated its efficacy and safety in clinical trials (Table 1). In 2017, the US Food and Drug Administration approved CPX-351 liposome injection for the treatment of adult patients with newly diagnosed therapy-related AML or AML with myelodysplasia-related changes.6 In 2021, the U.S. Food and Drug Administration (FDA) approved extended indication of CPX-351 to include pediatric patients (aged ≥1 year) with AML.7

Learn more about patient health-related quality of life and real-world outcomes with CPX-351 here.

To learn more about the advantages of CPX-351 over the conventional “7+3” treatment regimen, watch this expert opinion video: Does CPX-351 induce deeper responses compared to 7+3?

Liposomal formulations in development for AML

Prexigebersen (BP1001)

Growth factor receptor-bound protein-2 (GRB2) plays an essential role in cancer progression via the RAS activation pathway.13 Prexigebersen is a liposome-incorporated antisense oligodeoxynucleotide that targets GRB2.13 The liposomal formulation comprises of a P-ethoxy nucleic acid backbone and a neutral dioleoylphosphatidylcholine (DOPC) lipid delivery vehicle (Figure 5).14 The P-ethoxy backbone is formed by adding an ethyl group to the non-bridging oxygen atom of the phosphate bonds, producing a neutral, nuclease-resistant DNA antisense oligodeoxynucleotide that enhances its biodistribution and intracellular uptake.14

BP1002

BP1002 (liposomal Bcl-2 antisense oligodeoxynucleotide) is a neutral-charge antisense drug incorporated into liposomes, designed to inhibit the synthesis of Bcl-2, a protein that blocks programmed cell death.16,17 The FDA-approved Bcl-2 inhibitor venetoclax is used in frontline combination therapies for patients with AML ineligible for intensive chemotherapy; however, instances of resistance have been reported.16,17 A recent study revealed that patients with AML who relapsed following frontline venetoclax-based therapy were refractory to salvage therapy and had a very poor prognosis, with a median survival of less than 3 months.16,17 BP1002 could therefore be a potential treatment option for venetoclax-relapsed AML, as preclinical studies indicate that it works effectively with decitabine in venetoclax-resistant cells.16 A Phase 1/1b study (NCT05190471) is currently underway to evaluate BP1002 in combination with decitabine in patients with AML who have relapsed after venetoclax-based treatment.16,17

L-annamycin

Although anthracycline-based chemotherapy (e.g., “7+3”) is still a common induction approach in AML treatment, their use can be constrained due to inherent cardiotoxicity and drug resistance.18,19 L-annamycin (L-ANN) is a novel, doxorubicin congener encapsulated in multilamellar liposomes, designed to overcome multidrug resistance and avoid cardiotoxic effects.18,19 It is currently being investigated in a phase I/II MB-106 study (Table 3).

L-ANN has been granted Orphan Drug Designation by the EMA.20 L-ANN was previously granted Fast Track Designation21 and Orphan Drug Designation22 by the FDA for the treatment of patients with relapsed/refractory acute myeloid leukemia.

Other liposomal formulations in development

Table 4 provides a list of additional ongoing trials assessing liposomal drug delivery systems for AML.

Conclusion

The treatment landscape of AML is evolving, with the introduction of advanced liposomal drug delivery systems. By evading the immune response, and through enhanced cell permeability and retention, the novel liposomal formulations offer a promising approach to address the major treatment gaps in conventional therapies. Currently, CPX-351 is the only liposomal drug approved for the treatment of AML and is widely utilized in clinical practice, demonstrating improved efficacy compared with the standard “7 + 3” induction chemotherapy. Other liposomal formulations are in development, including prexigebersen and L-ANN, both of which have received Orphan Drug Designation from the FDA and EMA. Ongoing research in liposomal formulations marks a substantial step forward in management of AML, offering hope for more effective and personalized treatment options.

After reading the educational resource on liposomal drug delivery in AML, I commit to reviewing the latest data on CPX-351 and other liposomal drugs in development to guide my treatment of AML in clinical practice.
6 votes - 54 days left

This educational resource is independently supported by Jazz Pharmaceuticals. All content is developed by SES in collaboration with an expert steering committee; funders are allowed no influence on the content of this resource.

  1. Wu X, Wang F, Yang X, et al. Advances in Drug Delivery Systems for the Treatment of Acute Myeloid Leukemia. Small. 2024:2403409. Online ahead of print. DOI: 1002/smll.202403409
  2. Chen EC, Fathi AT, Brunner AM. Reformulating acute myeloid leukemia: liposomal cytarabine and daunorubicin (CPX-351) as an emerging therapy for secondary AML. OTT. 2018;11:3425-3434. DOI: 2147/OTT.S141212
  3. Briot T, Roger E, Thépot S, Lagarce F. Advances in treatment formulations for acute myeloid leukemia. Drug Discov Today. 2018;23(12):1936-1949. DOI: 1016/j.drudis.2018.05.040
  4. Benko A. Nanocarrier drug resistant tumor interactions: novel approaches to fight drug resistance in cancer. Cancer Drug Resist. 2021;4:264-297. DOI: 20517/cdr.2020.81
  5. Mayer LD, Tardi P, Louie AC. CPX-351: a nanoscale liposomal co-formulation of daunorubicin and cytarabine with unique biodistribution and tumor cell uptake properties. Int J Nanomedicine. 2019;14:3819-3830. DOI: 2147/IJN.S139450
  6. FDA News Release: FDA approves first treatment for certain types of poor-prognosis acute myeloid leukemia. https://www.fda.gov/news-events/press-announcements/fda-approves-first-treatment-certain-types-poor-prognosis-acute-myeloid-leukemia. Published Aug 3, 2017. Accessed Jul 26, 2024.
  7. PR Newswire. Jazz Pharmaceuticals Announces FDA Approval of Additional Indication for Vyxeos® (daunorubicin and cytarabine) for the Treatment of Secondary Acute Myeloid Leukemia in Pediatric Patients. https://www.prnewswire.com/news-releases/jazz-pharmaceuticals-announces-fda-approval-of-additional-indication-for-vyxeos-daunorubicin-and-cytarabine-for-the-treatment-of-secondary-acute-myeloid-leukemia-in-pediatric-patients-301259059.html. Published Mar 20, 2021. Accessed Jul 26, 2024.
  8. Feldman EJ, Lancet JE, Kolitz JE, 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(8):979- DOI: 10.1200/JCO.2010.30.5961
  9. Cortes JE, Goldberg SL, Feldman EJ, et al. Phase II, multicenter, randomized trial of CPX‐351 (cytarabine:daunorubicin) liposome injection versus intensive salvage therapy in adults with first relapse AML. Cancer. 2015;121(2):234-242. DOI: 1002/cncr.28974
  10. Lancet JE, Cortes JE, Hogge DE, et al. Phase 2 trial of CPX-351, a fixed 5:1 molar ratio of cytarabine/daunorubicin, vs cytarabine/daunorubicin in older adults with untreated AML. Blood. 2014;123(21):3239-3246. DOI: 1182/blood-2013-12-540971
  11. Lancet JE, Uy GL, Cortes JE, et al. CPX-351 (cytarabine and daunorubicin) liposome for injection versus conventional cytarabine plus daunorubicin in older patients with newly diagnosed secondary acute myeloid leukemia. J Clin Oncol. 2018;36(26):2684-2692. DOI: 1200/JCO.2017.77.6112
  12. Lancet JE, Uy GL, Newell LF, 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(7):e481-e491. DOI: 1016/S2352-3026(21)00134-4
  13. Ohanian M, Abdul-Hay M, Lin TL, et al. Interim safety and efficacy of BP1001 in a phase II acute myeloid leukemia (AML) study. Oral Abstract #6511. Presented at: 2024 American Society of Clinical Oncology (ASCO) Annual Meeting; May 31 – Jun 4, 2024; Chicago, US.
  14. Ohanian M, Tari Ashizawa A, Garcia-Manero G, et al. Liposomal Grb2 antisense oligodeoxynucleotide (BP1001) in patients with refractory or relapsed haematological malignancies: a single-centre, open-label, dose-escalation, phase 1/1b trial. Lancet Haematol. 2018;5(4):e136-e146. DOI: 1016/S2352-3026(18)30021-8
  15. Prexigebersen (Liposomal Grb2 antisense) for acute myeloid leukemia (AML). Bio-Path Holdings. https://www.biopathholdings.com/pipeline/prexigebersen/. Accessed July 24, 2024.
  16. Gagliardi M, Ashizawa AT. The combination of liposomal Bcl-2 antisense oligonucleotide (BP1002) with decitabine is efficacious in venetoclax-resistant cells. Abstract #959. Presented at: American Association for Cancer Research Annual Meeting 2021; May 17–21, 2021; Philadelphia, US.
  17. BP1002 (Liposomal BCL-2) for AML patients relapsed from Bcl-2 inhibitor treatment. https://www.biopathholdings.com/pipeline/bp1002/. Accessed July 23, 2024.
  18. Wadolowska A, Papayannidis C, Sartor C, et al. Liposomal annamycin (L-ANN) in combination with cytarabine for treatment of acute myeloid leukemia (AML) (MB-106 Study). Abstract#P537. Presented at: European Hematology Association 2024 Hybrid Congress; Jun 13–16, 2024; Madrid, ES.
  19. Gil L, Wierzbowska A, Wrobel T, et al. Results of a Phase I Study of Liposomal Annamycin for Treatment of R/R AML Patients after Induction Therapy. Br J Cancer Res. 2023;6(1). DOI: 31488/bjcr.183
  20. PR Newswire. European Medicines Agency (EMA) Grants Orphan Drug Designation for Moleculin’s Treatment of Acute Myeloid Leukemia (AML). https://www.prnewswire.com/news-releases/european-medicines-agency-ema-grants-orphan-drug-designation-for-moleculins-treatment-of-acute-myeloid-leukemia-aml-302120221.html. Published Apr 18, 2024. Accessed July 26, 2024.
  21. PR Newswire. Moleculin Receives FDA Approval of Fast Track Designation for Annamycin. https://www.prnewswire.com/news-releases/moleculin-receives-fda-approval-of-fast-track-designation-for-annamycin-300834393.html. Published Apr 18, 2019. Accessed July 26, 2024.
  22.  Moleculin Biotech Receives Orphan Drug Designation For Annamycin For The Treatment Of Acute Myeloid Leukemia. https://www.biospace.com/moleculin-biotech-receives-orphan-drug-designation-for-annamycin-for-the-treatment-of-acute-myeloid-leukemia. Published Mar 22, 2017. Accessed July 26, 2024.
  23. gov. MA+AZA regimen for the Treatment of Newly Diagnosed Acute Myeloid Leukemia (AML). https://clinicaltrials.gov/study/NCT06345365. Accessed July 26, 2024.
  24. gov. Mitoxantrone Hydrochloride Liposome Injection, Cytarabine Combined With Venetoclax in the Treatment of R/R AML. https://clinicaltrials.gov/study/NCT06434662. Updated May 30, 2024. Accessed July 26, 2024.
  25. gov. Liposome-encapsulated Daunorubicin-Cytarabine and Venetoclax in Treating Participants with Relapsed, Refractory or Untreated Acute Myeloid Leukemia. https://clinicaltrials.gov/study/NCT03629171. Updated July 30, 2024. Accessed July 30, 2024.
  26. gov. Clinical Study of Mitoxantrone Hydrochloride Liposome Injection in Subjects With Acute Myeloid Leukemia. https://clinicaltrials.gov/study/NCT05522192. Updated Aug 30, 2022. Accessed July 26, 2024.
  27. gov. VMAC+DLI Treatment of Patients With Relapse of AML After Allo-HSCT (VMAC+DLI). https://clinicaltrials.gov/study/NCT06447090. Updated June 6, 2024. Accessed July 26, 2024.
  28. gov. Comparing the Addition of an Anti-Cancer Drug, Pomalidomide, to the Usual Chemotherapy Treatment (Daunorubicin and Cytarabine Liposome) in Newly Diagnosed Acute Myeloid Leukemia With Myelodysplastic Syndrome-Related Changes. https://clinicaltrials.gov/study/NCT04802161. Updated Jul 25, 2024. Accessed July 26, 2024.
  29. gov. Liposome-encapsulated daunorubicin-cytarabine and gemtuzumab ozogamicin in treating patients with relapsed or refractory acute myeloid leukemia (AML) or high risk myelodysplastic syndrome. https://clinicaltrials.gov/study/NCT03672539. Updated Jul 30, 2024. Accessed July 26, 2024.