C25H26F6N2O2
552292-08-7
Delayed chemotherapy-induced nausea and vomiting
Interleukin-2 (IL-2) is a pleiotropic cytokine that functions as a central signaling molecule within the immune system, orchestrating the complex interplay between immune activation and tolerance.[1] Naturally produced by activated CD4+ and CD8+ T lymphocytes, IL-2 was originally identified as T-cell growth factor due to its critical role in promoting the proliferation, survival, and differentiation of various lymphocyte populations.[3] Its physiological functions are multifaceted. On one hand, IL-2 is a potent pro-inflammatory mediator, driving the expansion and enhancing the cytotoxic capabilities of effector T-cells and Natural Killer (NK) cells, which are essential for clearing pathogens and eliminating malignant cells.[2] On the other hand, it is indispensable for the maintenance of peripheral immune tolerance. IL-2 is fundamentally required for the development, survival, and function of regulatory T-cells (Tregs), a specialized subset of T-cells that suppress excessive immune responses and prevent autoimmunity.[1] This inherent duality—simultaneously promoting aggressive immune attacks and enforcing immune restraint—is determined by the concentration of IL-2 and the specific cellular context. This biological paradox is the cornerstone that explains its divergent therapeutic applications, from high-dose cancer immunotherapy to low-dose treatment for autoimmune disorders.
The therapeutic potential of IL-2 led to the development of aldesleukin, a recombinant analog of human IL-2, marketed under the brand name Proleukin®.[3] Aldesleukin is produced using recombinant DNA technology in a genetically engineered strain of Escherichia coli that contains a modified version of the human IL-2 gene.[6] The resulting product is a highly purified protein with a molecular weight of approximately 15,300 daltons.[6]
Critically, aldesleukin is not biochemically identical to native human IL-2. Its chemical name is des-alanyl-1, serine-125 human interleukin-2.[6] This nomenclature reflects two key modifications from the native protein: the N-terminal alanine was removed (des-alanyl-1), and the cysteine residue at position 125 was replaced with a serine.[6] These alterations were engineered to facilitate stable and efficient production in the bacterial expression system. While aldesleukin possesses the biological activities of native IL-2, its non-native structure creates the potential for immunogenicity.[6] The host immune system can recognize the recombinant protein as foreign, leading to the formation of anti-drug antibodies. Indeed, the development of non-neutralizing anti-interleukin-2 antibodies has been observed in a small fraction of patients (less than 1%), although the clinical significance of this phenomenon remains unknown.[9] This represents a direct link between the biopharmaceutical engineering required for the drug's manufacture and a tangible immunological consequence in patients.
For clinical use, Proleukin is supplied as a sterile, white to off-white, preservative-free lyophilized cake in single-use vials.[6] It is intended for intravenous administration following reconstitution with sterile water for injection and subsequent dilution.[6] The biological potency of aldesleukin is not measured by mass but by a functional lymphocyte proliferation bioassay, with activity expressed in International Units (IU) as established by the World Health Organization.[6] The standard conversion is 18 million IU of Proleukin being equivalent to 1.1 mg of protein.[6]
Aldesleukin is classified as a form of immunotherapy, a therapeutic modality that harnesses the patient's own immune system to fight disease.[3] More specifically, it is categorized as a biologic response modulator and a cytokine.[10] This classification fundamentally distinguishes it from conventional chemotherapy. Whereas cytotoxic chemotherapy agents work by directly killing rapidly dividing cells, including both cancer cells and healthy cells, aldesleukin has no direct cytotoxic effect.[10] Instead, its anti-tumor activity is entirely indirect; it functions by stimulating and activating the host's immune cells, which in turn recognize and destroy malignant cells.[10] This distinction is crucial for understanding its unique mechanism of action, its pattern of clinical efficacy, and its distinct and often severe toxicity profile.
| Table 1: Summary of Aldesleukin (Proleukin®) Characteristics | |
|---|---|
| Generic Name | Aldesleukin |
| Brand Name | Proleukin® |
| Synonyms | Recombinant Human Interleukin-2 (rIL-2), r-serHuIL-2 |
| Chemical Name | des-alanyl-1, serine-125 human interleukin-2 |
| Pharmacological Class | Immunotherapy, Cytokine, Biologic Response Modifier |
| Molecular Weight | ~ daltons |
| Production Method | Recombinant DNA technology (E. coli strain) |
| Formulation | Lyophilized powder for injection/infusion |
| Potency | mg protein = million International Units (IU) |
Data compiled from sources.[6]
The biological effects of aldesleukin are mediated through its binding to the IL-2 receptor (IL-2R), a multi-subunit complex expressed on the surface of various immune cells.[8] The IL-2R exists in three forms with distinct affinities for IL-2, and the differential expression of these receptor forms on various cell types underpins the dose-dependent effects of the drug.
Aldesleukin binding to the IL-2R initiates a cascade of intracellular signaling events. The binding event leads to the heterodimerization of the cytoplasmic domains of the β and γc chains.[8] This conformational change activates the Janus kinase 3 (Jak3), a tyrosine kinase associated with the receptor complex. Activated Jak3 then phosphorylates key tyrosine residues on the IL-2Rβ chain, creating docking sites for various cytoplasmic signaling molecules, most notably the Signal Transducer and Activator of Transcription 5 (STAT5).[8] The recruitment and subsequent phosphorylation of STAT5 leads to its dimerization, translocation to the nucleus, and binding to specific DNA sequences, ultimately driving the expression of genes critical for lymphocyte proliferation, survival, and effector function.
The primary anti-tumor mechanism of high-dose aldesleukin stems from its potent stimulation of cytotoxic effector lymphocytes.[8] By activating the IL-2R signaling pathway, aldesleukin enhances lymphocyte mitogenesis (cell division) and stimulates the long-term growth of IL-2-dependent cell lines, fulfilling its role as a T-cell growth factor.[4]
Beyond proliferation, aldesleukin dramatically enhances the cytotoxic (cell-killing) capacity of these immune cells.[8] It induces the activity of two key populations of anti-tumor effector cells:
This broad activation of cellular immunity is a hallmark of aldesleukin therapy and is clinically observed as profound lymphocytosis (increased lymphocytes), eosinophilia (increased eosinophils), and thrombocytopenia (decreased platelets).[9] The ultimate therapeutic goal of this process is to induce a robust, T-cell-mediated regression of tumors.[8] The importance of this mechanism is underscored by its application in clinical trials, where aldesleukin is used either to activate NK cells in vivo or to stimulate donor NK cells ex vivo before their adoptive transfer into patients.[15]
The immunomodulatory effects of aldesleukin are not confined to the direct stimulation of lymphocytes. Activation of the IL-2 pathway triggers a secondary cascade, leading to the production and release of a host of other pro-inflammatory cytokines by activated immune cells.[9] These include:
This cytokine cascade creates a highly inflamed tumor microenvironment and contributes to a systemic state of immune activation. This broad, non-specific amplification of the immune response is a critical component of aldesleukin's anti-tumor efficacy. However, it is also a double-edged sword. The same systemic release of TNF and IL-1 that contributes to tumor destruction is also directly responsible for many of the drug's most severe toxicities. The intense flu-like syndrome (fever, chills, rigors) experienced by nearly all patients is a direct consequence of this IL-1/TNF surge.[9] The most life-threatening toxicity, Capillary Leak Syndrome, is the extreme manifestation of the systemic vascular inflammation driven by this cytokine storm. Therefore, the adverse effects of aldesleukin are not aberrant or unexpected reactions; they are the predictable and systemic consequences of its powerful, intended immunomodulatory mechanism. The toxicity is inextricably linked to the efficacy.
Aldesleukin exhibits a distinct pharmacokinetic profile characterized by rapid distribution and clearance. Following intravenous infusion, the drug distributes rapidly, primarily to well-perfused organs such as the lungs, liver, kidneys, and spleen.[9] It exists in solution as biologically active, non-covalently bound microaggregates, a physical property that may be influenced by the formulation's solubilizing agent, sodium dodecyl sulfate.[6]
The drug has a short terminal half-life, reported to be between 80 and 120 minutes, which necessitates the frequent, every-8-hour dosing schedule used in high-dose regimens to maintain therapeutic concentrations.[9] Aldesleukin is not cleared as an intact molecule. Instead, it is primarily metabolized to its constituent amino acids within the cells of the proximal convoluted tubules of the kidney.[9] Consequently, only trace amounts of the active drug are found in the urine.[14] This renal metabolism means that drug clearance is not dependent on glomerular filtration rate, but severe renal dysfunction can still arise as a secondary consequence of drug-induced hypotension and reduced organ perfusion.[9]
High-dose aldesleukin therapy represents a potent but highly toxic treatment reserved for specific, advanced malignancies. Its use is defined by a narrow therapeutic index and the potential for rare but exceptionally durable responses.
Regulatory Approvals
Clinical Efficacy in Metastatic Melanoma
The approval of aldesleukin for metastatic melanoma was a landmark event in oncology, as it was the first immunotherapy to demonstrate the ability to induce long-term, complete remissions in this notoriously difficult-to-treat disease.20 An integrated analysis of 270 patients from eight clinical studies established the benchmark efficacy rates:
The most significant aspect of these responses was their profound durability. Among patients who achieved a CR, the median duration of response exceeded 59 months, with many patients remaining disease-free for over a decade.[20] This established the principle that immunotherapy could be curative for a subset of patients with advanced cancer, a concept that has driven the field of immuno-oncology ever since.[20]
Clinical Efficacy in Metastatic Renal Cell Carcinoma
Similar to melanoma, the efficacy of aldesleukin in metastatic RCC is characterized by a low overall response rate but the potential for durable remissions. Objective responses are observed in approximately 16% of patients.9 It was this ability to induce durable responses in a small fraction of patients that led to its initial FDA approval in 1992.19
However, the role of aldesleukin in RCC has undergone a significant re-evaluation in the modern era. The advent of newer, better-tolerated, and more broadly effective agents—first targeted therapies like vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors (TKIs) and later immune checkpoint inhibitors (ICIs)—has dramatically changed the treatment landscape.[19] This shift is formally reflected in the most current clinical practice guidelines. As of version 3.2025, the National Comprehensive Cancer Network (NCCN) Kidney Cancer guidelines no longer recommend Proleukin for the treatment of renal cell carcinoma.[22] This change marks a pivotal moment in the drug's history, illustrating a broader trend in medicine where a pioneering but difficult therapy is superseded by its more refined successors. The legacy of aldesleukin in RCC is now less about its direct clinical application and more about the paradigm of immunotherapy it helped to create.
The profound toxicity of high-dose aldesleukin necessitates a highly controlled and specialized approach to its administration, centered on stringent patient selection and intensive inpatient monitoring.
Patient Selection
Careful patient selection is mandatory and is the first and most critical step in mitigating risk.6 Treatment is strictly limited to a small subset of patients who meet the following criteria:
Administration Setting
Due to the high incidence of severe, life-threatening adverse events, particularly Capillary Leak Syndrome, aldesleukin must be administered in a hospital setting under the direct supervision of a qualified physician experienced in its use.10 The clinical course frequently requires management in an intensive care unit (ICU) to provide the necessary level of monitoring and supportive care (e.g., vasopressors, mechanical ventilation).10
High-Dose Intravenous Regimen
The standard high-dose regimen is identical for both metastatic melanoma and RCC and is structured as an intensive, cyclical course of therapy.22
| Table 2: High-Dose Aldesleukin Dosing Regimen for Metastatic RCC and Melanoma | |||
|---|---|---|---|
| Phase | Days | Action | Dose Limit |
| Cycle 1 | 1-5 | Administer IU/kg IV over 15 minutes every 8 hours. | Maximum of 14 doses |
| Rest Period | 6-14 | No treatment. | 9 days of rest |
| Cycle 2 | 15-19 | Administer IU/kg IV over 15 minutes every 8 hours. | Maximum of 14 doses |
| Notes: A full course comprises both cycles (maximum of 28 doses). Retreatment may be considered after ≥7 weeks of rest from hospital discharge if tumor shrinkage is observed. Doses are frequently withheld due to toxicity; patients in clinical trials received a median of 18-20 doses per course. |
Data compiled from sources.[7]
Alternative Administration Routes
While high-dose IV infusion is the standard for systemic therapy, other routes are used in specific clinical or investigational contexts. These include continuous intravenous infusion, low-dose subcutaneous injection (primarily for non-oncologic indications), and intralesional injection directly into cutaneous tumor nodules to achieve high local drug concentrations with reduced systemic toxicity.7
Beyond its primary FDA-approved indications, aldesleukin serves important roles in other specialized oncologic settings.
The clinical utility of high-dose aldesleukin is profoundly limited by its severe and predictable toxicity profile. The risk of life-threatening adverse events necessitates stringent patient selection, inpatient administration in specialized centers, and intensive management by an experienced multidisciplinary team.
Proleukin's prescribing information includes multiple black box warnings, the most serious class of warning issued by the FDA. These warnings highlight potentially fatal risks and restrict the drug's use.[23] Key warnings include:
The most significant and life-threatening toxicity associated with aldesleukin is Capillary Leak Syndrome (CLS).[14] This syndrome is the unifying pathophysiology behind many of the drug's most severe organ-specific toxicities.
Beyond CLS, high-dose aldesleukin is associated with a wide array of severe and common adverse events affecting nearly every organ system.
The severe toxicity profile and the complex management it requires have a direct impact on healthcare delivery. The need for advanced cardiac and pulmonary screening, coupled with the requirement for an on-site ICU and a highly experienced medical team, effectively concentrates the administration of high-dose aldesleukin at a small number of large, specialized academic medical centers.[10] This creates significant logistical and geographical barriers to access for many patients. Consequently, a patient's eligibility for this potentially curative therapy is often determined not only by their disease and physiological fitness but also by their proximity to a center of excellence capable of safely managing the treatment. This practical limitation has been a major factor in its limited adoption and has contributed to its current niche status in oncology.
| Table 3: Systemic Adverse Events Associated with High-Dose Aldesleukin (Incidence >10%) | |
|---|---|
| System | Adverse Event (Incidence) |
| Cardiovascular | Hypotension (71%), Tachycardia (23%), Arrhythmia (12%), Vasodilation (13%) |
| Constitutional | Chills (52%), Fever (29%), Malaise (27%), Asthenia (23%), Weight Gain (16%) |
| Gastrointestinal | Diarrhea (67%), Vomiting (50%), Nausea (35%), Stomatitis (22%), Anorexia (20%), Abdominal Pain (11%) |
| Renal/Genitourinary | Oliguria (63%), Increased Creatinine (33%) |
| Neurologic | Confusion (34%), Somnolence (22%), Dizziness (11%), Anxiety (12%) |
| Respiratory | Dyspnea (43%), Lung Disorder/Congestion (24%), Cough (11%), Rhinitis (10%) |
| Dermatologic | Rash (42%), Pruritus (24%), Exfoliative Dermatitis (18%) |
| Hematologic | Thrombocytopenia (37%), Anemia (29%), Leukopenia (16%), Eosinophilia (Profound) |
| Metabolic/Laboratory | Bilirubinemia (40%), Increased SGOT (23%), Acidosis (12%), Hypomagnesemia (12%), Hypocalcemia (11%) |
| Infection | Infection (13%) |
Data compiled from sources.[9]
Absolute Contraindications
Aldesleukin is strictly contraindicated in patients with:
Clinically Significant Drug Interactions
While high-dose aldesleukin is a potent immunostimulant used to fight cancer, a completely different therapeutic paradigm has emerged based on the administration of much lower doses to treat autoimmune and inflammatory diseases. This dichotomy is rooted in the fundamental immunobiology of the IL-2 receptor system.
The scientific basis for low-dose IL-2 therapy rests on the differential expression and affinity of IL-2 receptors on different lymphocyte populations.[5]
In many autoimmune diseases, such as systemic lupus erythematosus (SLE), there is a relative deficiency of IL-2 and/or a functional impairment of Tregs.[1] The therapeutic hypothesis of low-dose IL-2 is that administering small, subcutaneous doses can selectively stimulate and expand the highly sensitive Treg population without significantly activating the pro-inflammatory effector cells.[34] The goal is not to stimulate the immune system, but rather to restore its balance and re-establish self-tolerance, effectively turning a pro-inflammatory "sledgehammer" into an anti-inflammatory "scalpel".[1] This successful application of a deep biological insight—the differential receptor affinity model—to repurpose an existing drug for a completely opposite therapeutic goal represents a powerful example of reverse translational medicine. It demonstrates that the biological effect of a cytokine is not fixed but is highly dependent on concentration and context, opening new avenues for the nuanced application of other immunomodulators.
This immunological rationale has been validated in a growing number of clinical trials across a spectrum of autoimmune and inflammatory conditions.
| Table 4: Comparison of High-Dose vs. Low-Dose Interleukin-2 Therapy | ||
|---|---|---|
| Parameter | High-Dose IL-2 (Oncology) | Low-Dose IL-2 (Autoimmunity) |
| Therapeutic Goal | Immune Stimulation (Anti-Tumor) | Immune Tolerance (Anti-Autoimmune) |
| Primary Cellular Target | Effector T-Cells, NK Cells | Regulatory T-Cells (Tregs) |
| Key Mechanism | Enhance Cytotoxicity, Induce Cytokine Cascade | Selective Treg Expansion, Restore Homeostasis |
| Typical Indication | Metastatic Melanoma, RCC | Systemic Lupus Erythematosus (Investigational) |
| Typical Dose | IU/kg | million IU/day (total) |
| Route of Administration | Intravenous (IV) Infusion | Subcutaneous (SC) Injection |
| Site of Care | Hospital / ICU | Outpatient / Home |
| Key Toxicity | Capillary Leak Syndrome | Generally well-tolerated, injection site reactions |
Data compiled from sources.[1]
The significant limitations of aldesleukin—namely its severe toxicity and narrow therapeutic window—have driven extensive research into two main areas: combining it with other immunotherapies to enhance efficacy, and engineering novel IL-2 variants with improved properties.
A compelling rationale exists for combining aldesleukin with immune checkpoint inhibitors (ICIs) such as anti-PD-1 (e.g., pembrolizumab, nivolumab) or anti-CTLA-4 (e.g., ipilimumab) antibodies. These two classes of drugs have complementary mechanisms of action.[20] Aldesleukin provides a powerful, non-specific "accelerator" or "go" signal to the immune system, driving the proliferation and activation of T-cells. In contrast, ICIs work by "releasing the brakes," blocking the inhibitory pathways that tumors exploit to evade immune destruction.[39] The hypothesis is that combining these two approaches—simultaneously stimulating T-cells and removing the signals that suppress them—could lead to a more profound and durable anti-tumor response.[39]
This hypothesis is being actively tested in numerous clinical trials.
The quest to uncouple the potent efficacy of IL-2 from its debilitating toxicity has led to a major effort in protein engineering to create next-generation IL-2 analogs with superior therapeutic properties. The primary goals of this research are to:
Bempegaldesleukin (NKTR-214): A Case Study in Rational Design and Clinical Disappointment
Bempegaldesleukin (BEMPEG, NKTR-214) was one of the most advanced next-generation IL-2 candidates, designed with a clear and logical scientific rationale.43 It is a prodrug consisting of recombinant IL-2 with multiple polyethylene glycol (PEG) chains covalently attached.44
The failure of bempegaldesleukin serves as a profound cautionary tale in drug development. It reveals a critical gap in our understanding of IL-2 biology within the complex human tumor microenvironment. The logical assumption that simply biasing receptor affinity away from Tregs and towards effector cells would result in superior anti-tumor efficacy proved to be incorrect. This outcome forces a re-evaluation of fundamental assumptions, suggesting that the role of Tregs may be more complex than previously thought, or that the specific pharmacokinetics of the prodrug failed to achieve the intended biological effect where it mattered most. It is a powerful lesson that a successful next-generation immunotherapy requires more than just an elegant molecular engineering solution; it must contend with the still-unpredictable complexity of the human immune response to cancer.
Other Novel Approaches
Research continues on other strategies to improve IL-2 therapy. These include immunocytokines (antibody-cytokine fusion proteins) and immunotoxins, which link IL-2 to a tumor-targeting antibody or a toxin, respectively.51 The goal of these approaches is to deliver the IL-2 signal or a cytotoxic payload directly to the tumor site, thereby maximizing local anti-tumor activity while minimizing systemic exposure and toxicity.51
Aldesleukin, as the first therapeutic recombinant cytokine, occupies a unique and paradoxical position in the history of medicine. Its legacy is twofold. First, it is a foundational pillar of modern immuno-oncology. By demonstrating that systemic administration of a single immune-stimulating agent could induce durable, complete remissions in patients with advanced, incurable cancers like melanoma and renal cell carcinoma, aldesleukin provided the first definitive proof-of-concept for immunotherapy as a curative modality. It fundamentally altered the perception of what was possible in oncology and paved the way for the development of all subsequent immunotherapies, including the now-ubiquitous immune checkpoint inhibitors.
Simultaneously, aldesleukin carries a second, darker legacy as a cautionary tale of extreme toxicity. Its association with life-threatening Capillary Leak Syndrome and a host of other severe adverse events established a benchmark for the potential dangers of broad, non-specific immune stimulation. The challenges of managing its toxicity have profoundly shaped the development of all subsequent immunotherapies, driving a relentless focus on improving the therapeutic index and uncoupling efficacy from severe, systemic side effects.
In the current therapeutic landscape, dominated by better-tolerated and more broadly effective immune checkpoint inhibitors and targeted therapies, high-dose aldesleukin has been relegated to a small but important clinical niche. Its use as a frontline therapy has diminished significantly, as evidenced by its removal from NCCN guidelines for renal cell carcinoma. However, it remains a viable, albeit challenging, option for a very select group of young, highly fit patients with metastatic melanoma or RCC. In this specific population, the small but real potential for a durable, long-term cure may still be deemed to outweigh the significant risks and toxicities of treatment, particularly after other options have been exhausted. Furthermore, aldesleukin retains a critical role in specialized applications, including the management of steroid-refractory graft-versus-host disease and as an essential supportive agent for promoting the efficacy of adoptive cell therapies like TILs.
The story of human interleukin-2 therapy is a powerful narrative of scientific evolution. It began with the identification of a native cytokine, progressed to the development of a potent but challenging therapeutic agent in aldesleukin, and has now entered an era of sophisticated bioengineering aimed at creating safer, more effective next-generation molecules. The clinical failure of rationally designed agents like bempegaldesleukin underscores the immense complexity of the immune system and highlights the gaps that remain in our understanding.
The future of IL-2-based therapy lies in achieving greater specificity. The ultimate goal is to engineer variants that can precisely target and activate anti-tumor effector cells preferentially within the tumor microenvironment, deliver a sustained and optimal signal, and avoid the systemic toxicities and off-target effects that have limited the utility of aldesleukin. The journey of IL-2—from a fundamental biological discovery to a pioneering drug and a template for future innovation—vividly demonstrates both the immense power of the immune system to cure cancer and the profound challenge of harnessing that power safely and effectively.
Published at: October 8, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.