You are here
Home > Blog > Uncategorized > Cytokine Engineering: IL-2, IL-15, and Beyond

Cytokine Engineering: IL-2, IL-15, and Beyond

Cytokine Engineering: IL-2, IL-15, and Beyond


Cytokine Engineering


 Check out our extensive video library (see channel for our latest videos)

Abstract

This paper looks into cytokine engineering, with particular focus on interleukin-2 (IL-2) and interleukin-15 (IL-15). These proteins play essential roles in immune system regulation and have become important targets for medical treatments. The paper reviews the basic science behind cytokines, discusses engineering methods used to modify these proteins, and explores current and future medical applications. Key topics include protein structure modifications, half-life extension techniques, targeted delivery systems, and clinical outcomes in cancer and autoimmune diseases. The analysis draws from recent clinical trials and laboratory studies to provide healthcare professionals with current knowledge about engineered cytokines in medical practice. Safety considerations, dosing strategies, and combination therapies are examined. The paper also addresses challenges in cytokine engineering, including unwanted side effects and manufacturing difficulties, while highlighting promising developments in next-generation engineered cytokines.



Introduction

Cytokines are small proteins that cells use to communicate with each other. They act as messengers in the immune system, telling cells when to grow, move, or fight infections. Among the many cytokines in the human body, IL-2 and IL-15 stand out for their powerful effects on immune cells, particularly T cells and natural killer (NK) cells.

The discovery of IL-2 in 1976 opened new doors in immunology. Scientists found they could grow T cells in laboratory dishes by adding IL-2, a breakthrough that enabled studies of the immune system. IL-15 was discovered in 1994 and shares many similarities with IL-2 but has unique properties that make it interesting for various medical uses.

Natural cytokines often have problems when used as medicines. They break down quickly in the body, requiring frequent doses. They can cause severe side effects because they affect many different cell types. They might not reach the specific tissues where they are needed most. These limitations led scientists to develop engineered versions of cytokines that function more effectively as treatments.

Cytokine engineering involves altering the structure of these proteins to enhance their therapeutic utility. This can mean prolonging their residence in the body, reducing side effects, or helping them target specific organs or tumors. The field has grown rapidly in the past decade, with several engineered cytokines now approved for treating cancer and other diseases.


The Biology of IL-2 and IL-15

IL-2 Structure and Function

IL-2 is a protein made up of 133 amino acids arranged in a specific three-dimensional shape. It has four alpha helices (spiral structures) that form a compact bundle. This shape allows IL-2 to bind to receptors on immune cells.

The IL-2 receptor comes in three forms, depending on which protein chains combine:

  • Low-affinity receptor: Contains only the alpha chain (CD25)
  • Intermediate affinity receptor: Contains beta (CD122) and gamma (CD132) chains
  • High-affinity receptor: Contains all three chains (alpha, beta, and gamma)

Different immune cells have different combinations of these receptor chains. Regulatory T cells (Tregs) have high levels of the alpha chain, making them very sensitive to IL-2. Effector T cells and NK cells mainly express intermediate-affinity receptors.

When IL-2 binds its receptor, it triggers a cascade of intracellular signals. These signals tell the cell to divide, produce other proteins, and become more active. The strength and duration of these signals depend on which receptor type is engaged.

IL-15 Structure and Function

IL-15 has 114 amino acids and a similar four-helix bundle structure to IL-2. Despite sharing only about 20% sequence similarity with IL-2, IL-15 can bind to the same beta and gamma receptor chains. However, IL-15 uses a different alpha chain (IL-15Rα) than IL-2.

A unique feature of IL-15 is trans-presentation. Cells that produce IL-15 often present it to other cells while bound to IL-15Rα on their surface. It differs from IL-2, which typically functions as a free-floating protein. Trans-presentation helps IL-15 signal more effectively and last longer in tissues.

IL-15 preferentially stimulates memory CD8+ T cells and NK cells without strongly activating Tregs. This makes it attractive for cancer treatment, where activating Tregs could suppress anti-tumor responses.

Comparing IL-2 and IL-15 Effects

Interleukin-2 (IL-2) and interleukin-15 (IL-15) share receptor subunits, yet their biological actions differ significantly. IL-2 powerfully expands regulatory T cells and enhances their suppressive function, but prolonged exposure can drive activation-induced cell death in conventional T cells. It also has a notably short half-life, lasting only minutes to hours in circulation, and higher doses are associated with toxicities such as vascular leak syndrome.

In contrast, IL-15 preferentially supports the survival of memory T cells and natural killer cells, without strongly stimulating regulatory T cells. It is a key driver of long-term T-cell memory formation and is generally better tolerated clinically, with fewer adverse effects at therapeutic doses compared with IL-2.


Principles of Cytokine Engineering Top Of Page

Protein Engineering Strategies

Scientists use several approaches to modify cytokines:

Amino Acid Substitution: Changing specific amino acids can alter how a cytokine binds to its receptors. For example, replacing certain amino acids in IL-2 can reduce its binding to the alpha chain, decreasing Treg activation while maintaining effects on effector T cells.

Deletion and Addition: Removing or adding protein segments can change cytokine properties. Some engineered IL-2 variants have deletions that prevent binding to certain receptor chains.

Fusion Proteins: Joining cytokines to other proteins can improve their properties. Common fusion partners include:

  • Antibody Fc regions: help extend a cytokine’s half-life by engaging the body’s natural Fc-receptor recycling pathways, allowing the fused molecule to circulate longer before clearance.
  • Albumin: This prolongs the lifetime of cytokine in the bloodstream, as albumin naturally has a long half-life and avoids rapid renal clearance.
  • Targeting domains: These guide cytokines to specific tissues or cell types, enhancing therapeutic precision while limiting off-target effects. 

Chemical Modifications: Adding polyethylene glycol (PEG) chains or other chemical groups can protect cytokines from breakdown and reduce immune recognition.

Half-Life Extension Technologies

Natural cytokines are quickly cleared from the bloodstream. Several methods extend their duration:

Fc Fusion: Attaching the Fc portion of antibodies allows cytokines to use the same recycling system that makes antibodies last weeks in the body. This involves the neonatal Fc receptor (FcRn), which protects proteins from degradation.

PEGylation: Adding PEG chains increases protein size and shields them from kidney filtration and enzyme degradation. The size and attachment site of PEG chains must be carefully chosen to maintain cytokine activity.

Albumin Binding: Some engineered cytokines contain domains that bind to albumin, a long-lasting blood protein. This indirect approach avoids increasing the cytokine’s size while extending its half-life.

Controlled Release Formulations: Embedding cytokines in slowly dissolving materials can provide sustained release over days or weeks.

Targeting Strategies

Directing cytokines to specific locations reduces side effects and improves efficacy:

Antibody-Cytokine Fusions: Also called immunocytokines, these combine tumor-targeting antibodies with cytokines. The antibody portion guides the cytokine to cancer cells.

Cell Surface Anchoring: Engineering cytokines to stay attached to cell membranes limits their systemic effects while maintaining local activity.

Conditional Activation: Some designs include masks that block cytokine activity until removed by enzymes found in tumors or inflamed tissues.


Current Engineered IL-2 and IL-15 Variants

Modified IL-2 Variants in Development

NKTR-214 (Bempegaldesleukin): This IL-2 variant has multiple PEG chains attached at specific sites. The PEGylation pattern preferentially blocks binding to the alpha chain, reducing Treg activation. In the body, PEG chains slowly detach, creating a controlled-release effect. Clinical trials demonstrated tumour responses in patients with melanoma and renal cell carcinoma.

ALKS 4230: This engineered fusion protein combines a circularly permuted IL-2 with the extracellular domain of IL-2Rα. This design prevents interaction with the high-affinity receptor on Tregs while maintaining activity on intermediate-affinity receptors. Phase 1 and 2 studies demonstrated acceptable safety profiles and anti-tumor activity.

THOR-707: Uses specific amino acid mutations to eliminate alpha chain binding. Additional modifications extend the half-life without using PEGylation. Early clinical data showed immune activation with reduced vascular leak syndrome compared to standard IL-2.

Synthorin (STK-012): Contains precise polymer attachments that modulate receptor binding and extend half-life. The polymer placement was optimized through computational design and screening.

Engineered IL-15 Variants

ALT-803 (N-803): Consists of an IL-15 mutant (N72D) complexed with IL-15Rα-Fc fusion protein. The mutation increases biological activity, while the receptor-Fc fusion extends half-life and enables trans-presentation. Clinical studies showed NK and T cell activation in cancer patients.

SO-C101: A fusion of IL-15 with a proprietary protein domain that extends half-life while maintaining native IL-15 activity. Designed for less frequent dosing compared to native IL-15.

RLI-15: Comprises IL-15 bound to the extracellular domain of IL-15Rα, mimicking natural trans-presentation. Shows enhanced stability and activity compared to free IL-15.

NKTR-255: A polymer-conjugated IL-15 designed for monthly dosing. The polymer attachment sites were selected to maintain receptor binding while dramatically extending circulation time.


Clinical Applications Top Of Page

Cancer Immunotherapy

Engineered cytokines show promise across multiple cancer types:

Melanoma: Several engineered IL-2 variants demonstrated tumor shrinkage in patients with advanced melanoma. Response rates range from 10% to 25%, with some patients achieving long-lasting remissions. Combining checkpoint inhibitors, such as anti-PD-1 antibodies, increases response rates.

Renal Cell Carcinoma: High-dose IL-2 was one of the first immunotherapies approved for kidney cancer. Engineered variants show similar efficacy with improved safety profiles. Studies combining engineered cytokines with tyrosine kinase inhibitors are ongoing.

Lymphomas: IL-15-based therapies show particular promise due to their strong ability to activate NK cells. Early trials of ALT-803 in combination with rituximab showed encouraging results in B-cell lymphomas.

Solid Tumors: Immunocytokines targeting tumor antigens concentrate cytokine activity in tumors. Examples include anti-GD2-IL-2 for neuroblastoma and anti-EpCAM-IL-2 for epithelial cancers.

Combination Therapy Approaches

Engineered cytokines work well with other cancer treatments:

With Checkpoint Inhibitors: IL-2 and IL-15 variants increase T cell numbers and activity, potentially overcoming resistance to PD-1/PD-L1 blockade. Multiple combination trials are evaluating optimal dosing schedules.

With Adoptive Cell Therapy: Engineered cytokines support transferred T cells or NK cells. Lower doses may suffice when cytokines target transferred cells specifically.

With Cancer Vaccines: Cytokines boost immune responses to tumor antigens. Timing relative to vaccination affects outcomes, with some studies showing benefits from cytokine administration several days after vaccination.

With Targeted Therapies: Combining cytokines with antibodies or small molecules that target cancer cells creates synergistic effects. The cytokine activates immune cells while targeted therapy weakens cancer cells.

Autoimmune and Inflammatory Diseases

Low-dose IL-2 therapy explores a different application – promoting Tregs to control autoimmune diseases:

Type 1 Diabetes: Low-dose IL-2 increased Treg numbers and function in type 1 diabetes patients. Some studies showed preservation of insulin-producing cells in newly diagnosed patients.

Systemic Lupus Erythematosus: Clinical trials demonstrated that low-dose IL-2 reduced disease activity scores and allowed decreased use of immunosuppressive drugs.

Graft-versus-Host Disease: Low-dose IL-2 helps prevent or treat this complication of bone marrow transplantation by supporting T regulatory cells (Tregs).

Some engineered IL-2 variants designed to preferentially activate Tregs are in development for autoimmune applications.


Safety Considerations and Side Effect Management

Common Adverse Effects

Understanding and managing cytokine-related side effects is crucial for healthcare providers:

Flu-like Symptoms: Fever, chills, muscle aches, and fatigue occur frequently with cytokine therapy. These result from immune activation and cytokine release. Management includes acetaminophen, NSAIDs, and meperidine for severe chills.

Vascular Leak Syndrome: High-dose IL-2 can cause fluid to leak from blood vessels into tissues. This leads to swelling, low blood pressure, and organ dysfunction. Engineered variants with reduced alpha-chain binding exhibit decreased vascular leakage.

Cardiovascular Effects: Hypotension, arrhythmias, and myocarditis can occur. Careful fluid management and cardiac monitoring are essential. Some centers use prophylactic medications to prevent arrhythmias.

Laboratory Abnormalities: Cytokine therapy often causes predictable lab changes:

  • Lymphopenia followed by lymphocytosis
  • Eosinophilia
  • Elevated liver enzymes
  • Thyroid dysfunction
  • Anemia and thrombocytopenia

Dosing Strategies

Optimal dosing remains an active area of research:

High-Dose Regimens: Traditional high-dose IL-2 uses 600,000-720,000 IU/kg every 8 hours. This requires inpatient monitoring due to severe side effects.

Intermediate Doses: Some centers use lower doses (72,000-125,000 IU/kg) with improved tolerability and maintained efficacy in selected patients.

Engineered Variant Dosing: Longer-acting variants allow less frequent administration. Weekly or even monthly dosing becomes possible with some engineered cytokines.

Personalized Dosing: Biomarkers like baseline immune cell counts or early response markers may guide dose selection. Some trials adjust doses based on side effects or immune monitoring.

Monitoring Parameters

Careful monitoring ensures patient safety:

Vital Signs: Blood pressure, heart rate, temperature, and oxygen saturation should be assessed frequently during treatment.

Fluid Balance: Daily weights, intake/output monitoring, and edema assessment guide fluid management.

Laboratory Tests:

  • Complete blood counts with differential
  • Comprehensive metabolic panel
  • Liver function tests
  • Thyroid function (baseline and periodic)
  • C-reactive protein or other inflammatory markers

Imaging: Baseline and follow-up scans assess tumor response. Chest X-rays may detect pulmonary edema during treatment.


Manufacturing and Quality Control

Production Methods

Producing engineered cytokines requires sophisticated biotechnology:

Expression Systems: Most therapeutic cytokines are produced in:

  • Chinese Hamster Ovary (CHO) cells: These are the industry standard for making glycosylated proteins with human-like post-translational modifications. 
  • E. coli: Some cytokines are made in E. coli, which is suitable for producing non-glycosylated variants quickly and cost-effectively.
  • Yeast: It is used as an alternative platform when a balance between eukaryotic folding and rapid, scalable production is needed.

Purification Processes: Multiple steps ensure purity:

  • Chromatography is used to separate the desired cytokine from other cellular proteins and impurities
  • Viral inactivation steps are included to ensure patient safety by eliminating potential viral contaminants
  • Endotoxin removal is another critical step, especially for products expressed in bacteria, to prevent harmful inflammatory reactions.
  • Final formulation process optimizes the cytokine’s stability, solubility, and shelf life.

Quality Attributes: Throughout manufacturing, strict control of key quality attributes is essential. Manufacturers must ensure correct protein folding and structural integrity, maintain appropriate glycosylation patterns for glycosylated cytokines, and minimise aggregate formation, which can affect safety and efficacy. They must also verify biological activity through functional assays and ensure long-term stability so the product remains safe and effective throughout its shelf life.

Regulatory Considerations

Engineered cytokines face rigorous regulatory review:

Preclinical Requirements: Animal studies must demonstrate:

  • Mechanism of action: Research must elucidate the cytokine’s interactions with its target cells and pathways, thereby illustrating the biological processes by which it exerts its therapeutic effect.
  • Toxicity profile: Comprehensive animal studies are required to identify any harmful effects, determine dose-dependent toxicity, and assess potential organ-specific or systemic adverse reactions.
  • Pharmacokinetics: Regulators require data on how the cytokine is absorbed, distributed, metabolised, and eliminated, which helps predict its behaviour in the human body.
  • Starting dose rationale for human studies: Preclinical studies provide evidence to validate the initial human dose by ascertaining the maximal non-toxic dose in animal models, delineating dose–response relationships, and evaluating exposure levels correlated with both efficacy and toxicity. Using safety factors and modeling, these data are then used to derive a safe, conservative starting dose for human trials.

Clinical Development: Phase 1 studies focus on safety and dose finding. Phase 2 trials explore efficacy signals and optimal patient populations. Phase 3 trials provide definitive efficacy data for approval.

Post-Marketing Surveillance: Continued monitoring identifies rare side effects and long-term outcomes. Registry studies track real-world effectiveness.

Biosimilar Considerations: As patents expire, biosimilar versions of engineered cytokines enter development. These require extensive comparison to the original products.

Cytokine Engineering

 


Future Directions Top Of Page

Next-Generation Engineering Approaches

Computational Design: Machine learning predicts optimal mutations and modifications. Algorithms consider receptor binding, stability, and immunogenicity simultaneously.

Conditional Activation: Smart cytokines activate only in disease environments. Examples include:

  • pH-sensitive variants active in acidic tumor environments
  • Protease-activated cytokines
  • Temperature-sensitive designs

Multi-Specific Designs: Proteins targeting multiple receptors or combining different cytokine activities. These activate certain pathways while blocking others.

Synthetic Biology Approaches: Engineered cells producing cytokines in response to disease signals. CAR-T cells secreting cytokines represent early examples.

Emerging Cytokine Targets

Beyond IL-2 and IL-15, other cytokines undergo engineering:

IL-12 and IL-23: Important for T helper cell differentiation. Engineered versions aim to enhance anti-tumor immunity while avoiding autoimmunity.

IL-10: An anti-inflammatory cytokine with potential in inflammatory bowel disease and other conditions. PEGylated IL-10 showed promise in early trials.

IL-21: Supports B-cell and T-cell functions. Combination with other immunotherapies under investigation.

Type I Interferons: Engineered interferons with improved half-life and reduced side effects target viral infections and cancer.

Personalized Medicine Applications

Biomarker-Guided Therapy: Selecting patients likely to respond based on:

  • Tumor immune infiltration
  • Cytokine receptor expression
  • Genetic markers
  • Baseline immune status

Combination Selection: Algorithms may predict optimal cytokine combinations for individual patients.

Dose Optimization: Pharmacokinetic/pharmacodynamic modeling enables personalized dosing.

Response Monitoring: Liquid biopsies and immune monitoring guide treatment modifications.


Challenges and Limitations

Technical Challenges

Immunogenicity: Engineered proteins may trigger antibody responses that neutralize activity. Strategies to reduce immunogenicity include:

  • Humanization of sequences
  • Avoiding aggregation-prone modifications
  • PEGylation to shield immunogenic sites

Manufacturing Complexity: Producing consistent, high-quality engineered cytokines requires:

  • Sophisticated production facilities
  • Extensive quality control
  • High costs that affect drug pricing

Stability Issues: Engineered proteins may be less stable than natural versions. Formulation development addresses:

  • Storage conditions
  • Delivery methods
  • In-use stability

Biological Limitations

Redundancy in Cytokine Networks: Multiple cytokines often have overlapping functions. Blocking or activating single cytokines may have limited effects.

Context-Dependent Effects: The same cytokine can have opposite effects in different settings. IL-2 promotes both immune activation and regulation.

Resistance Mechanisms: Tumors may develop resistance through:

  • Downregulation of cytokine receptors
  • Production of inhibitory factors
  • Changes in tumor microenvironment

Individual Variability: Patient factors affecting response include:

  • Genetic polymorphisms in cytokine pathways
  • Prior treatments affecting immune function
  • Comorbidities influencing drug metabolism

Clinical Translation Challenges

Dose Finding: The optimal biological dose may differ from the maximum tolerated dose. Immune effects don’t always follow typical dose-response curves.

Endpoint Selection: Measuring immune activation doesn’t always predict clinical benefit. Survival endpoints require long follow-up periods.

Combination Complexity: Adding cytokines to other treatments creates challenges:

  • Overlapping toxicities
  • Optimal sequencing unknown
  • Increased costs
  • Regulatory hurdles

Cytokine Engineering


Conclusion Led   Top Of Page

Cytokine engineering represents a powerful approach to improving immunotherapy. By modulating IL-2, IL-15, and other cytokines, scientists develop drugs with improved safety profiles and enhanced therapeutic effects. Current engineered variants show promise in treating cancer and autoimmune diseases, with several products in late-stage clinical trials or recently approved.

Key advances include methods to extend cytokine half-life, reduce unwanted receptor interactions, and target specific cell populations. These improvements address limitations of natural cytokines, making treatment more practical and tolerable for patients. Healthcare providers now have access to engineered cytokines that can be administered less frequently, with fewer side effects than earlier generations.

However, challenges remain in optimizing these therapies. Individual patient responses vary, and predicting who will benefit most remains difficult. Manufacturing costs are high, potentially limiting access. Combination strategies require careful study to maximize benefits while minimizing risks.

Looking ahead, next-generation engineering approaches promise even more sophisticated cytokine-based therapies. Computational design, conditional activation, and personalized medicine applications may further improve outcomes. As our understanding of immune regulation deepens, cytokine engineering will likely play an expanding role in treating cancer, autoimmune diseases, and infectious diseases.

For healthcare professionals, staying informed about engineered cytokines is important as these therapies become more common in clinical practice. Understanding their mechanisms, managing side effects, and selecting appropriate patients will optimize outcomes. The field continues to evolve rapidly, making ongoing education essential.

Key Takeaways

  1. Engineered cytokines address the limitations of natural IL-2 and IL-15, including their short half-lives, severe side effects, and lack of cell-type selectivity.
  2. Major engineering strategies include PEGylation, Fc fusion, targeted mutations, and antibody conjugation to improve pharmacokinetics and reduce toxicity.
  3. Several engineered IL-2 variants (NKTR-214, ALKS 4230, THOR-707) show clinical promise with reduced Treg activation and vascular leak syndrome.
  4. IL-15-based therapies (ALT-803, NKTR-255) preferentially activate NK cells and memory T cells without strong Treg stimulation.
  5. Combination approaches with checkpoint inhibitors, adoptive cell therapy, and targeted agents enhance therapeutic effects.
  6. Low-dose IL-2 therapy shows efficacy in autoimmune diseases by promoting regulatory T cell expansion.
  7. Careful monitoring of flu-like symptoms, vascular leak, and cardiovascular effects is essential during cytokine therapy.
  8. Manufacturing complexity and quality control requirements affect the cost and accessibility of engineered cytokines.
  9. Future directions include computational design, conditional activation, and personalized medicine approaches.
  10. Success with engineered cytokines requires appropriate patient selection, dose optimization, and side-effect management by trained healthcare teams.

 

Frequently Asked Questions:    Top Of Page

Q: What are the main differences between engineered and natural cytokines?

A: Engineered cytokines have modifications that make them last longer in the body, cause fewer side effects, and target specific cell types better than natural versions. Natural cytokines break down quickly and affect many different cells, while engineered versions are designed to work more precisely.

Q: How do engineered IL-2 variants reduce side effects compared to high-dose IL-2?

A: Engineered IL-2 variants often have modifications that prevent binding to the alpha chain of the IL-2 receptor. It reduces activation of regulatory T cells and decreases vascular leak syndrome while maintaining anti-cancer effects through other receptor types.

Q: What makes IL-15 attractive for cancer therapy compared to IL-2?

A: IL-15 preferentially activates NK cells and memory T cells without strongly stimulating regulatory T cells that can suppress immune responses. IL-15 also promotes long-term immune memory and is generally better tolerated than IL-2.

Q: Can engineered cytokines be used in outpatient settings?

A: Many newer engineered cytokines can be given in outpatient settings due to improved safety profiles and longer-lasting effects. This contrasts with high-dose IL-2, which requires intensive inpatient monitoring. However, careful patient selection and monitoring remain important.

Q: What biomarkers predict response to cytokine therapy?

A: Current biomarkers include baseline immune cell counts, tumor-infiltrating lymphocytes, PD-L1 expression, and tumor mutational burden. However, reliable predictive biomarkers remain an active area of research, and no single test definitively predicts response.

Q: How are dosing schedules determined for engineered cytokines?

A: Dosing depends on the specific modifications made to each cytokine. Half-life extension allows less frequent dosing – some engineered variants can be given weekly or monthly instead of multiple times daily. Phase 1 trials establish optimal dosing through carefully designed dose-escalation studies.

Q: What are the most serious side effects to monitor?

A: The most serious effects include severe hypotension, pulmonary edema from vascular leak syndrome, cardiac arrhythmias, and kidney dysfunction. Most engineered variants have reduced risk of these effects compared to natural cytokines, but monitoring remains important.

Q: Can patients develop resistance to engineered cytokines?

A: Yes, resistance can develop through several mechanisms: production of neutralizing antibodies against the engineered protein, downregulation of cytokine receptors on target cells, or changes in the tumor microenvironment that prevent immune cell function.

Q: How do the costs of engineered cytokines compare to other cancer treatments?

A: Engineered cytokines are expensive due to complex manufacturing and development costs. However, they may be cost-effective given reduced hospitalization needs, the potential for long-lasting responses, and the ability to be combined with other therapies. Cost varies significantly between different products.

Q: What combination therapies show the most promise?

A: Combinations with checkpoint inhibitors (anti-PD-1/PD-L1) show strong clinical results. Engineered cytokines also enhance adoptive cell therapies and may overcome resistance to targeted therapies. Optimal combinations depend on cancer type and individual patient factors.

 


References:   Top Of Page

Bentebibel, S. E., Hurwitz, M. E., Bernatchez, C., Haymaker, C., Hudgens, C. W., Kluger, H. M., … & Diab, A. (2019). A first-in-human study and biomarker analysis of NKTR-214, a novel IL2Rβγ-biased cytokine, in patients with advanced or metastatic solid tumors. Cancer Discovery, 9(6), 711-721.

Berraondo, P., Sanmamed, M. F., Ochoa, M. C., Etxeberria, I., Aznar, M. A., Pérez-Gracia, J. L., … & Melero, I. (2019). Cytokines in clinical cancer immunotherapy. British Journal of Cancer, 120(1), 6-15.

Charych, D., Khalili, S., Dixit, V., Kirk, P., Chang, T., Langowski, J., … & Zalevsky, J. (2017). Modeling the receptor pharmacology, pharmacokinetics, and pharmacodynamics of NKTR-214, a kinetically-controlled interleukin-2 (IL2) receptor agonist for cancer immunotherapy. PLoS One, 12(7), e0179431.

Conlon, K. C., Lugli, E., Welles, H. C., Rosenberg, S. A., Fojo, A. T., Morris, J. C., … & Waldmann, T. A. (2015). Redistribution, hyperproliferation, activation of natural killer cells and CD8 T cells, and cytokine production during the first-in-human clinical trial of recombinant human interleukin-15 in patients with cancer. Journal of Clinical Oncology, 33(1), 74-82.

Deckers, J., Anbergen, T., Hokke, A. M., de Dreu, A., Schrijver, D. P., de Bruin, K., … & Sparidans, R. W. (2022). Engineering cytokine therapeutics. Nature Reviews Bioengineering, 1(4), 286-303.

Dougan, M., Dranoff, G., & Dougan, S. K. (2019). GM-CSF, IL-3, and IL-5 family of cytokines: regulators of inflammation. Immunity, 50(4), 796-811.

Garralda, E., Navarro, A., Hanna, G. J., Sukari, A., Fayette, J., Gillison, M. L., … & Melero, I. (2021). ARTACUS-101: A first-in-human phase 1 study of ARTBV-Fc-IL2, a novel anti-PD-L1/IL-2 fusion protein, in patients with advanced solid tumors. Journal of Clinical Oncology, 39(15_suppl), 2551-2551.

Hernandez, R., Põder, J., LaPorte, K. M., & Malek, T. R. (2022). Engineering IL-2 for immunotherapy of autoimmunity and cancer. Nature Reviews Immunology, 22(10), 614-628.

Khoryati, L., Pham, M. N., Sherve, M., Kumari, S., Bogdani, M., Campbell, D. J., & Gavin, M. A. (2020). Regulatory T cell expansion by a highly CD25-selective IL-2 mutein arrests ongoing autoimmunity. bioRxiv.

Klein, C., Waldhauer, I., Nicolini, V. G., Freimoser-Grundschober, A., Nayak, T., Vugts, D. J., … & Umaña, P. (2017). Cergutuzumab amunaleukin (CEA-IL2v), a CEA-targeted IL-2 variant-based immunocytokine for combination cancer immunotherapy: overcoming limitations of aldesleukin and conventional IL-2-based immunocytokines. Oncoimmunology, 6(3), e1277306.

Lopes, J. E., Fisher, J. L., Flick, H. L., Wang, C., Sun, L., Ernstoff, M. S., … & Farid, S. S. (2020). ALKS 4230: a novel engineered IL-2 fusion protein with an improved cellular selectivity profile for cancer immunotherapy. Journal for Immunotherapy of Cancer, 8(1), e000673.

Margolin, K., Morishima, C., Velcheti, V., Miller, J. S., Lee, S. M., Silk, A. W., … & Thompson, J. A. (2022). Phase I trial of ALT-803, a novel recombinant IL-15 complex, in patients with advanced solid tumors. Clinical Cancer Research, 28(22), 4882-4893.

Milling, L., Zhang, Y., & Irvine, D. J. (2017). Delivering safer immunotherapies for cancer. Advanced Drug Delivery Reviews, 114, 79-101.

Mortara, L., Balza, E., Bruno, A., Poggi, A., Orecchia, P., & Carnemolla, B. (2018). Anti-cancer therapies employing IL-2 cytokine tumor targeting: contribution of innate, adaptive, and immunosuppressive cells in the anti-tumor efficacy. Frontiers in Immunology, 9, 2905.

Overwijk, W. W., Tagliaferri, M. A., & Zalevsky, J. (2021). Engineering IL-2 to give new life to T cell immunotherapy. Annual Review of Medicine, 72, 281-311.

Ptacin, J. L., Caffaro, C. E., Ma, L., San Jose Gall, K. M., Aerni, H. R., Acuff, N. V., … & Mijalis, A. J. (2021). An engineered IL-2 reprogrammed for anti-tumor therapy using a semi-synthetic organism. Nature Communications, 12(1), 1-14.

Puskas, J., Skrombolas, D., Sedlacek, A., Lord, E., Sullivan, M., & Frelinger, J. (2021). Development of an attenuated interleukin-2 fusion protein that can be activated by tumour-expressed proteases. Immunology, 164(3), 531-540.

Ren, Z., Zhang, A., Sun, Z., Liang, Y., Ye, J., Qiao, J., … & Li, B. (2022). Selective delivery of low-affinity IL-2 to PD-1+ T cells rejuvenates anti-tumor immunity with reduced toxicity. Journal of Clinical Investigation, 132(3), e153604.

Rosenberg, S. A. (2014). IL-2: the first effective immunotherapy for human cancer. Journal of Immunology, 192(12), 5451-5458.

Sahin, D., Arenas-Ramirez, N., Rath, M., Karakus, U., Hümbelin, M., van Gogh, M., … & Boyman, O. (2020). An IL-2-grafted antibody immunotherapy with potent efficacy against metastatic cancer. Nature Communications, 11(1), 1-12.

Sharma, M., Khong, H., Fa’ak, F., Bentebibel, S. E., Janssen, L. M., Chesson, B. C., … & Diab, A. (2020). Bempegaldesleukin selectively depletes intratumoral Tregs and potentiates T cell-mediated cancer therapy. Nature Communications, 11(1), 1-11.

Silva, D. A., Yu, S., Ulge, U. Y., Spangler, J. B., Jude, K. M., Labão-Almeida, C., … & Baker, D. (2019). De novo design of potent and selective mimics of IL-2 and IL-15. Nature, 565(7738), 186-191.

Sun, Z., Ren, Z., Yang, K., Liu, Z., Cao, S., Deng, S., … & Li, B. (2019). A next-generation tumor-targeting IL-2 preferentially promotes tumor-infiltrating CD8+ T-cell response and effective tumor control. Nature Communications, 10(1), 1-12.

Waldmann, T. A. (2018). Cytokines in cancer immunotherapy. Cold Spring Harbor Perspectives in Biology, 10(12), a028472.

Ward, N. C., Yu, A., Moro, A., Ban, Y., Chen, X., Hsiung, S., … & West, J. (2020). IL-2/CD25: a long-acting fusion protein that promotes immune tolerance by selectively targeting the IL-2 receptor on regulatory T cells. Journal of Immunology, 205(6), 1701-1712.

Wrangle, J. M., Velcheti, V., Patel, M. R., Garrett-Mayer, E., Hill, E. G., Ravenel, J. G., … & Rubinstein, M. P. (2018). ALT-803, an IL-15 superagonist, in combination with nivolumab in patients with metastatic non-small cell lung cancer: a non-randomised, open-label, phase 1b trial. The Lancet Oncology, 19(5), 694-704.

Zhang, Y., & Zhang, Z. (2020). The history and advances in cancer immunotherapy: understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cellular & Molecular Immunology, 17(8), 807-821.


[Internal Medicine -Home]

 

Video Section


 

Recent Articles

Cardiology

   


 

 

About Author

Similar Articles

Leave a Reply


thpxl