MedPath

Axitinib Advanced Drug Monograph

Published:Jul 23, 2025

Generic Name

Axitinib

Brand Names

Inlyta, Axitinib Accord

Drug Type

Small Molecule

Chemical Formula

C22H18N4OS

CAS Number

319460-85-0

Associated Conditions

Advanced Renal Cell Carcinoma, Advanced Thyroid cancer

Axitinib (Inlyta®): A Comprehensive Oncological and Pharmacological Review

Executive Summary

Axitinib represents a significant advancement in the targeted therapy of advanced renal cell carcinoma (RCC). As a potent, second-generation, small-molecule inhibitor of vascular endothelial growth factor receptors (VEGFRs), it was developed to offer greater selectivity and potency over first-generation agents. Initially approved as a second-line monotherapy, its clinical utility was established in the pivotal Phase III AXIS trial, where it demonstrated a statistically significant and clinically meaningful improvement in progression-free survival (PFS) compared to sorafenib in patients who had failed one prior systemic therapy. This trial solidified Axitinib's role in the treatment algorithm for mRCC and validated PFS as a key regulatory endpoint in this setting.

The therapeutic landscape for RCC was subsequently revolutionized by the advent of immune checkpoint inhibitors (ICIs). Axitinib emerged as a premier combination partner, with landmark trials such as KEYNOTE-426 (with pembrolizumab) and JAVELIN Renal 101 (with avelumab) demonstrating superior overall survival (OS) and PFS for the combination regimens over sunitinib monotherapy in the first-line setting. These results established Axitinib-ICI combinations as a new standard of care. This enhanced efficacy, however, is associated with a more challenging toxicity profile, requiring careful patient selection and proactive management of adverse events.

Pharmacologically, Axitinib is characterized by rapid oral absorption, high plasma protein binding, and a short half-life, supporting a twice-daily dosing schedule. Its metabolism is predominantly mediated by the CYP3A4/5 enzyme system, making it susceptible to significant drug-drug interactions. A notable pharmacodynamic feature is the high incidence of hypertension, an on-target effect that may serve as a clinical biomarker of drug activity. Mechanisms of resistance involve the upregulation of alternative proangiogenic pathways and modulation of the tumor microenvironment. Future research is focused on optimizing combination strategies, identifying predictive biomarkers, and exploring Axitinib's potential in other malignancies and therapeutic settings, such as neoadjuvant therapy. This report provides a comprehensive analysis of Axitinib, from its fundamental chemical properties to its complex role in modern oncology.

1.0 Introduction: The Role of Axitinib in the Evolving Landscape of Renal Cell Carcinoma Therapy

1.1 Context of Renal Cell Carcinoma (RCC)

Renal cell carcinoma is the most common form of kidney cancer, with recent data showing approximately 65,000 new cases and over 13,000 deaths annually in the United States alone.[1] While the five-year survival rate for localized kidney cancer is relatively high at 70%, the prognosis for patients with advanced or metastatic disease (mRCC) has historically been poor.[1] For decades, surgery remained the only definitive treatment for localized disease, while traditional systemic chemotherapy offered little to no significant improvement in survival for patients with mRCC.[1] This therapeutic gap underscored the urgent need for novel treatment strategies based on the underlying biology of the disease.

1.2 The Angiogenesis-Targeted Therapy Revolution

A paradigm shift in the management of mRCC began with the elucidation of its molecular pathogenesis. A vast majority of clear-cell RCC, the most common histological subtype, is characterized by mutations or epigenetic silencing of the Von Hippel-Lindau (VHL) tumor suppressor gene.[2] The VHL protein is a critical component of an E3 ubiquitin ligase complex that targets hypoxia-inducible factors (HIF-1α and HIF-2α) for proteasomal degradation under normoxic conditions. In the absence of functional VHL protein, HIFs accumulate, translocate to the nucleus, and act as transcription factors, driving the expression of a suite of genes involved in tumorigenesis.[2]

Among the most critical HIF target genes are those encoding pro-angiogenic growth factors, particularly Vascular Endothelial Growth Factor (VEGF) and Platelet-Derived Growth Factor (PDGF).[2] These factors bind to their respective tyrosine kinase receptors on endothelial cells, initiating signaling cascades that promote endothelial cell proliferation, migration, and survival, ultimately leading to the formation of new blood vessels—a process known as angiogenesis.[2] This pathological angiogenesis is essential for providing tumors with the oxygen and nutrients required for growth, invasion, and metastasis.[6] The central role of the VHL/HIF/VEGF axis in RCC pathogenesis provided a clear and compelling rationale for the development of therapies targeting this pathway.

1.3 Emergence of Axitinib

Following this biological understanding, a new class of drugs, the small-molecule tyrosine kinase inhibitors (TKIs), was developed. First-generation agents like sunitinib and sorafenib demonstrated significant clinical activity by inhibiting VEGFR and other kinases, dramatically improving outcomes compared to the prior standard of care with cytokines.[1] However, these agents were associated with significant off-target toxicities and eventual development of resistance.

This context set the stage for the development of second-generation TKIs designed for greater potency and selectivity. Axitinib, developed by Pfizer and marketed under the brand name Inlyta®, emerged as a leading agent in this class.[6] It is an oral, potent, and highly selective inhibitor of VEGFRs 1, 2, and 3, with reported potency that is 50 to 450 times higher than that of first-generation inhibitors.[9] By more specifically and powerfully blocking the VEGF signaling pathway, Axitinib was engineered to maximize anti-angiogenic effects, thereby inhibiting tumor growth and progression, while potentially offering a more manageable safety profile.[4] Its development and subsequent clinical validation have marked another critical step forward in the targeted treatment of advanced RCC.

2.0 Chemical Profile and Formulation

2.1 Nomenclature and Identifiers

Axitinib is identified through a standardized set of chemical names, synonyms, brand names, and registration numbers that ensure its precise identification across scientific, clinical, and regulatory domains.

  • Chemical Name (IUPAC): The formal International Union of Pure and Applied Chemistry (IUPAC) name for the molecule is N-methyl-2-[[(E)-2-pyridin-2-ylethenyl]-1H-indazol-6-yl]sulfanyl]benzamide.[9]
  • Synonyms and Code Names: During its development by Pfizer, Axitinib was known by the code name AG013736 (also written as AG-013736 or AG 013736).[12]
  • Brand Names: The drug is most commonly marketed globally under the brand name Inlyta®.[8]
  • Registration Numbers: Key identifiers used in databases and regulatory filings include:
  • CAS Number: 319460-85-0.[8]
  • DrugBank ID: DB06626.[8]
  • PubChem CID: 6450551.[8]
  • UNII (Unique Ingredient Identifier): C9LVQ0YUXG.[8]
  • ChEMBL ID: CHEMBL1289926.[8]

2.2 Chemical Structure and Molecular Properties

Axitinib is a small molecule belonging to the indazole class of compounds, which forms its core heterocyclic structure.

  • Structural Description: Chemically, Axitinib is an indazole derivative. Its structure is characterized by an indazole ring substituted at two key positions: at position 3, there is a 2-(pyridin-2-yl)vinyl group, and at position 6, a 2-(N-methylaminocarboxy)phenylsulfanyl group is attached via a thioether linkage.[9] This specific arrangement of functional groups—including the indazole, pyridine, aryl sulfide, and benzamide moieties—is responsible for its high-affinity binding to the ATP pocket of VEGFRs.[2]
  • Molecular Formula: The empirical chemical formula for Axitinib is C22​H18​N4​OS.[8]
  • Molecular Weight: The calculated molecular weight of Axitinib is approximately 386.47 g·mol⁻¹ (often rounded to 386.5 g/mol).[8]

2.3 Physicochemical Properties and Formulation

The physical and chemical properties of Axitinib dictate its formulation, stability, and pharmacokinetic behavior.

  • Appearance: In its solid state, Axitinib is described as a white to off-white or tan crystalline powder.[12]
  • Solubility: Axitinib is poorly soluble in aqueous media, a characteristic common to many oral TKIs. It is practically insoluble in water and ethanol but is soluble in organic solvents like dimethyl sulfoxide (DMSO).[12] This low aqueous solubility is a key consideration for its oral drug formulation and absorption characteristics.
  • Melting Point: The melting point of Axitinib is reported to be in the range of 211–214°C.[21]
  • Formulation: For clinical use, Axitinib is formulated as an oral, red, film-coated immediate-release (FCIR) tablet.[9] Studies have been conducted using at least two different crystal polymorphs of the active pharmaceutical ingredient, designated Form IV and Form XLI, which can influence properties like solubility and dissolution rate.[22]

Table 1: Key Identifiers and Physicochemical Properties of Axitinib

PropertyValueSource(s)
IUPAC NameN-methyl-2-[[(E)-2-pyridin-2-ylethenyl]-1H-indazol-6-yl]sulfanyl]benzamide9
Common NameAxitinib8
Brand NameInlyta®8
CAS Number319460-85-08
DrugBank IDDB066268
Molecular FormulaC22​H18​N4​OS9
Molecular Weight386.47 g·mol⁻¹8
AppearanceWhite to off-white crystalline solid powder12
Solubility (DMSO)Soluble (e.g., 2.5 mg/mL, 42 mg/mL)12
Solubility (Water)Insoluble (<1 mg/mL)12
Solubility (Ethanol)Insoluble (<1 mg/mL)21
Melting Point211–214°C21

3.0 Clinical Pharmacology

3.1 Mechanism of Action: High-Potency Inhibition of VEGFR and Other Kinases

Axitinib's therapeutic effect is derived from its function as a highly potent and selective, second-generation tyrosine kinase inhibitor.[9] Its mechanism is centered on the blockade of key signaling pathways that drive tumor angiogenesis.

  • Primary Target and Potency: The primary molecular targets of Axitinib are the vascular endothelial growth factor receptors VEGFR-1, VEGFR-2, and VEGFR-3.[6] It inhibits these receptor tyrosine kinases at therapeutic plasma concentrations, demonstrating a potency that is reported to be 50 to 450 times greater than that of first-generation VEGFR inhibitors.[9] This exceptional potency is quantified by its very low half-maximal inhibitory concentrations ($IC_{50}$s): 0.1 nM for VEGFR-1, 0.2 nM for VEGFR-2, and 0.1–0.3 nM for VEGFR-3.[12] This high affinity is a defining characteristic of Axitinib and distinguishes it from earlier TKIs.[2]
  • Molecular Mechanism: Like other TKIs, Axitinib functions by competitively binding to the adenosine triphosphate (ATP)-binding site within the intracellular kinase domain of its target receptors.[6] Its small molecular structure allows it to fit snugly into a tunnel within the kinase domain, stabilizing the receptor in an inactive conformation.[2] This physical occupation of the ATP pocket prevents the binding of ATP and blocks the subsequent autophosphorylation of the receptor, which is the critical first step in signal transduction.[2] By inhibiting this phosphorylation event, Axitinib effectively halts the downstream signaling cascade, including the activation of key pathways such as protein kinase B (AKT) and mitogen-activated protein kinases (MAPK/ERK).[2]
  • Biological Effect: The direct consequence of inhibiting VEGFR signaling in endothelial cells is a profound anti-angiogenic effect.[6] Axitinib blocks VEGF-mediated endothelial cell proliferation, migration, and survival, thereby preventing the formation of new blood vessels that tumors require to grow and metastasize.[7] Preclinical studies in tumor xenograft mouse models have confirmed that Axitinib treatment leads to reduced microvessel density, a key marker of angiogenesis, and subsequent inhibition of tumor growth.[13]
  • Secondary Targets: In addition to its primary action on VEGFRs, Axitinib also exhibits inhibitory activity against other receptor tyrosine kinases at nanomolar concentrations, including the platelet-derived growth factor receptor beta (PDGFRβ) and c-KIT, with IC50​ values of 1.6 nM and 1.7 nM, respectively.[8] Inhibition of these pathways may contribute to its overall anti-tumor activity, as they are also implicated in cancer cell proliferation and survival.

3.2 Pharmacodynamics

Pharmacodynamics describes the effect of a drug on the body. For Axitinib, this includes a demonstrable relationship between drug exposure and clinical response, as well as on-target effects that can serve as clinical biomarkers.

  • Exposure-Response Relationship: Clinical data from trials in patients with mRCC have established a positive correlation between Axitinib exposure and clinical efficacy. An analysis revealed that patients who achieved a higher drug exposure, as measured by the area under the plasma concentration-time curve (AUC), on day 15 of the first treatment cycle had significantly longer median PFS and higher objective response rates (ORRs) compared to patients with sub-therapeutic exposure.[1] This finding provides a strong rationale for the dose-titration strategy employed with Axitinib, which aims to maximize exposure to an individually tolerable level.
  • Hypertension as a Biomarker: The high potency and selectivity of Axitinib for VEGFRs lead to a high incidence of on-target adverse events, most notably hypertension. Inhibition of VEGFR in normal vasculature is thought to decrease nitric oxide production and cause endothelial dysfunction, resulting in vasoconstriction and elevated blood pressure.[17] Crucially, a rise in diastolic blood pressure to ≥90 mmHg has been associated with improved clinical outcomes, suggesting that hypertension can serve as a pharmacodynamic biomarker of target engagement and drug activity.[1] This transforms hypertension from merely a side effect to be managed into a potential indicator that the drug is effectively inhibiting its target. This relationship underpins the clinical strategy of managing hypertension with anti-hypertensive medications to allow for continued dosing or even dose escalation of Axitinib, thereby optimizing the therapeutic benefit for the patient.
  • Cardiac Effects: A dedicated study was conducted to evaluate the effect of Axitinib on cardiac repolarization in 35 healthy subjects. The results showed no large, clinically significant changes in the mean QTc interval (i.e., >20 ms) from placebo. However, the study could not definitively rule out the possibility of small increases in the mean QTc interval (i.e., <10 ms).[23]

3.3 Pharmacokinetics: A Comprehensive Profile (ADME)

The pharmacokinetic profile of Axitinib describes its absorption, distribution, metabolism, and excretion (ADME), which collectively determine the drug's concentration-time profile in the body and inform its dosing regimen.

  • Absorption: Following oral administration, Axitinib is absorbed relatively quickly, with the time to reach maximum plasma concentration (Tmax​) ranging from 2.5 to 4.1 hours.[8] The mean absolute bioavailability of a 5 mg oral dose is 58%.[8] The rate and extent of absorption are not significantly affected by food; administration with a moderate-fat meal results in comparable exposure to administration under fasting conditions.[23] At steady state, Axitinib exhibits approximately linear pharmacokinetics over a dose range of 1 mg to 20 mg.[23]
  • Distribution: Axitinib is extensively bound to human plasma proteins, with a binding percentage greater than 99%.[8] It binds preferentially to albumin, with moderate binding to α1-acid glycoprotein.[2] The apparent volume of distribution ( Vd​) is 160 L, indicating distribution into tissues beyond the plasma volume.[8]
  • Metabolism: Axitinib undergoes extensive metabolism, primarily in the liver. The major metabolic pathway is oxidation via the cytochrome P450 (CYP) 3A4 and CYP3A5 enzyme systems.[7] Minor contributions to its metabolism (<10% each) are made by CYP1A2, CYP2C19, and uridine diphosphate glucuronosyltransferase (UGT) 1A1.[8] This heavy reliance on CYP3A4/5 is a critical clinical consideration, as it makes Axitinib highly susceptible to drug-drug interactions with inhibitors or inducers of these enzymes. The two major metabolites identified in human plasma are a sulfoxide product (M12) and an N-glucuronide product (M7); both are considered pharmacologically inactive, being at least 400-fold less potent against VEGFR-2 than the parent compound.[11]
  • Excretion: The elimination of Axitinib and its metabolites occurs primarily through hepatobiliary excretion. Following an oral dose, approximately 41% is recovered in the feces and 23% in the urine, with negligible urinary excretion of the unchanged drug.[8] Axitinib has a short effective plasma half-life ( t1/2​), which ranges from 2.5 to 6.1 hours.[8] This short half-life is consistent with the need for a twice-daily dosing regimen to maintain therapeutic plasma concentrations.
  • Population Pharmacokinetics: A two-compartment population pharmacokinetic model has been used to describe the drug's behavior.[11] Analysis of data from multiple trials has shown that while patient body weight can contribute to interindividual variability in pharmacokinetics, there are no clinically relevant effects of age, sex, race, renal function, or common genetic polymorphisms in UGT1A1 or CYP2C19 on the clearance of Axitinib.[11] However, moderate hepatic impairment results in a two-fold increase in drug exposure (AUC), necessitating a starting dose reduction in this population.[11]

Table 2: Summary of Key Pharmacokinetic Parameters of Axitinib

ParameterValueSource(s)
Absolute Bioavailability58%8
Time to Peak Concentration (Tmax​)2.5–4.1 hours8
Peak Concentration (Cmax​)27.8 ng/mL8
Area Under the Curve (AUC24​)265 ng·h/mL8
Volume of Distribution (Vd​)160 L8
Plasma Protein Binding>99%8
Terminal Half-life (t1/2​)2.5–6.1 hours8
Primary Metabolizing EnzymesCYP3A4, CYP3A58
Minor Metabolizing EnzymesCYP1A2, CYP2C19, UGT1A18
Primary Excretion RoutesFeces (41%), Urine (23%)8

4.0 Clinical Efficacy in Advanced Renal Cell Carcinoma

The clinical development of Axitinib has been marked by its successful validation in both second-line and first-line settings for advanced RCC, fundamentally altering treatment standards.

4.1 Second-Line Monotherapy: A Detailed Analysis of the Pivotal AXIS Trial (NCT00678392)

The AXIS trial was the landmark study that established the efficacy of Axitinib and led to its initial global regulatory approvals. It was the first Phase III trial to demonstrate the superiority of one targeted agent over another in the second-line mRCC setting.[26]

  • Study Design: AXIS was a randomized, open-label, multicenter, Phase III trial designed to directly compare the efficacy and safety of Axitinib versus sorafenib, an existing standard of care.[26]
  • Patient Population: The trial enrolled 723 patients with advanced or metastatic clear-cell RCC who had experienced disease progression after one prior systemic therapy.[27] Eligible prior therapies included sunitinib-based (54% of patients), cytokine-based (e.g., interferon-alfa or interleukin-2; 35%), bevacizumab-based (8%), or temsirolimus-based (3%) regimens.[26] Patients were required to have an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1 and were stratified by this status and their type of prior therapy.[26]
  • Treatment Arms: Patients were randomized in a 1:1 ratio to receive either Axitinib at a starting dose of 5 mg orally twice daily or sorafenib at a standard dose of 400 mg orally twice daily.[26] A key feature of the Axitinib arm was a provision for intra-patient dose titration; patients who tolerated the starting dose without significant hypertension or other adverse events could have their dose increased to 7 mg and then to 10 mg twice daily to optimize drug exposure and efficacy.[28]
  • Endpoints:
  • The primary endpoint was progression-free survival (PFS), as assessed by a blinded, independent radiology review committee.[26]
  • Secondary endpoints included overall survival (OS), objective response rate (ORR), duration of response, safety, and patient-reported outcomes.[26]
  • Key Efficacy Results:
  • Progression-Free Survival (PFS): The trial successfully met its primary endpoint. Axitinib demonstrated a statistically significant and clinically meaningful 43% improvement in median PFS compared to sorafenib. The median PFS was 6.7 months for the Axitinib arm versus 4.7 months for the sorafenib arm (Hazard Ratio 0.665; 95% Confidence Interval [CI] 0.544–0.812; p<0.0001).[27]
  • PFS by Prior Therapy Subgroup: The PFS benefit with Axitinib was consistent across the main patient subgroups. For patients previously treated with cytokines, the benefit was particularly pronounced, with a median PFS of 12.1 months for Axitinib versus 6.5 months for sorafenib (HR 0.46; p<0.0001).[29] For the larger subgroup of patients previously treated with sunitinib, Axitinib also showed a significant advantage, with a median PFS of 4.8 months versus 3.4 months for sorafenib (HR 0.74; p=0.0107).[29]
  • Objective Response Rate (ORR): Axitinib more than doubled the ORR compared to sorafenib, achieving a rate of 19.4% versus 9.4%, respectively (p=0.0001).[29]
  • Overall Survival (OS): Despite the clear benefit in PFS and ORR, the trial did not show a statistically significant difference in median OS. An updated analysis reported a median OS of 20.1 months with Axitinib compared to 19.2 months with sorafenib.[33] This lack of OS benefit is a common finding in second-line mRCC trials of this era and is widely attributed to the confounding effect of active post-study therapies. Patients progressing on the sorafenib arm had access to subsequent lines of treatment, which likely diluted the survival difference between the two initial arms. This observation reinforces the validity of PFS as a primary endpoint for regulatory approval in this disease setting, as it measures the direct effect of the therapy before being confounded by subsequent treatments.

4.2 First-Line Combination Therapy: The Paradigm Shift with Immune Checkpoint Inhibitors (ICIs)

Following its success as a monotherapy, Axitinib became a key agent in the next major evolution of RCC treatment: the combination of VEGFR TKIs with immune checkpoint inhibitors. The biological rationale for this approach is compelling; VEGFR inhibition can help normalize the tumor vasculature and modulate the immunosuppressive tumor microenvironment, potentially enhancing the infiltration and activity of T-cells unleashed by ICI therapy.[38]

  • KEYNOTE-426 Trial (Axitinib + Pembrolizumab):
  • Design: This pivotal, randomized, open-label Phase III trial evaluated Axitinib (5 mg twice daily) plus the anti-PD-1 antibody pembrolizumab (200 mg every 3 weeks) versus sunitinib monotherapy as a first-line treatment for patients with advanced clear-cell RCC.[41] The trial included 861 patients across all risk groups.[43]
  • Results: The combination of Axitinib and pembrolizumab demonstrated superiority over sunitinib across all key efficacy endpoints, leading to a new standard of care.
  • Overall Survival (OS): At the first interim analysis, the combination significantly improved OS, reducing the risk of death by 47% (HR 0.53; 95% CI 0.38–0.74; p<0.0001). The estimated 12-month survival rate was 89.9% for the combination arm versus 78.3% for the sunitinib arm.[42]
  • Progression-Free Survival (PFS): The combination also significantly prolonged PFS, with a median of 15.1 months versus 11.1 months for sunitinib (HR 0.69; 95% CI 0.57–0.84; p=0.0001).[41]
  • Objective Response Rate (ORR): The ORR was substantially higher with the combination at 59.3% (including a 6% complete response rate) compared to 36% for sunitinib.[41] The benefit was consistent across all risk groups and regardless of PD-L1 expression status.[43]
  • JAVELIN Renal 101 Trial (Axitinib + Avelumab):
  • Design: This Phase III trial similarly compared Axitinib plus the anti-PD-L1 antibody avelumab versus sunitinib monotherapy in 886 treatment-naïve patients with advanced RCC.[44]
  • Results: This combination also proved superior to sunitinib. At a minimum follow-up of 68 months, the combination showed a significantly longer median investigator-assessed PFS in the overall population (13.9 months vs. 8.5 months; HR 0.66; p ≤.0001).[45] The results were particularly strong in the sub-population of patients with PD-L1-positive tumors, which was one of the primary endpoints of the study.

Table 3: Summary of the AXIS Phase III Trial Design and Key Efficacy Outcomes

ParameterAxitinib Arm (n=361)Sorafenib Arm (n=362)Hazard Ratio (95% CI) / p-valueSource(s)
Study DesignRandomized, open-label, Phase III, second-line therapyN/AN/A26
Patient PopulationPatients with mRCC after failure of one prior systemic therapyN/AN/A26
Primary EndpointProgression-Free Survival (PFS)N/AN/A29
Median PFS (Overall)6.7 months4.7 monthsHR 0.665 (0.544–0.812); p<0.000128
Median PFS (Prior Cytokine)12.1 months6.5 monthsHR 0.464; p<0.000129
Median PFS (Prior Sunitinib)4.8 months3.4 monthsHR 0.741; p=0.010729
Objective Response Rate (ORR)19.4%9.4%p=0.000129
Median Overall Survival (OS)20.1 months19.2 monthsNot Statistically Significant34

Table 4: Summary of Key First-Line Combination Therapy Trials

ParameterAxitinib + Pembrolizumab (KEYNOTE-426)Axitinib + Avelumab (JAVELIN Renal 101)Sunitinib (Control)Key Statistics (vs. Sunitinib)Source(s)
Trial NameKEYNOTE-426JAVELIN Renal 101N/AN/A43
Patient PopulationTreatment-naïve advanced RCCTreatment-naïve advanced RCCN/AN/A41
Median PFS15.1 months13.9 months11.1 months (K-426) / 8.5 months (J-101)HR 0.69 (p=0.0001) / HR 0.66 (p≤.0001)42
Median OSNot Reached (at interim)Not Mature (at primary analysis)Not Reached (at interim)HR 0.53 (p<0.0001) for K-42642
ORR59.3%55.9% (PD-L1+)36% (K-426) / 25.5% (J-101)Statistically Significant41

5.0 Comparative Analysis in the RCC Armamentarium

Positioning Axitinib within the increasingly crowded therapeutic landscape for advanced RCC requires a nuanced comparison of its efficacy and safety relative to other key agents, both as a monotherapy and as a combination partner.

5.1 Efficacy and Safety vs. Other VEGFR TKIs (Sunitinib, Pazopanib, Cabozantinib)

Direct head-to-head trials provide the highest level of evidence, but indirect comparisons from network meta-analyses and real-world data also offer valuable context.

  • Direct Comparisons: The only major direct comparison for Axitinib monotherapy comes from the AXIS trial, which definitively established its superior PFS and ORR over sorafenib in the second-line setting.[28] There are no large-scale Phase III trials directly comparing Axitinib monotherapy to sunitinib, pazopanib, or cabozantinib in the same line of therapy. The COMPARZ trial established the non-inferiority of pazopanib to sunitinib in the first-line setting, while the CABOSUN trial showed superior PFS for cabozantinib over sunitinib in intermediate- and poor-risk first-line patients.[44]
  • Indirect Comparisons and Real-World Evidence: Network meta-analyses (NMAs) attempt to synthesize data from multiple trials. Several NMAs of first-line TKI monotherapies have concluded that there are no significant differences in PFS among cabozantinib, sunitinib, and pazopanib.[49] In the second-line setting, the picture is more complex. One comprehensive NMA found that cabozantinib and nivolumab both demonstrated longer OS than everolimus. A sensitivity analysis within that same review suggested that both cabozantinib and nivolumab were also superior to Axitinib in terms of OS.[50] This highlights a key challenge of NMAs: results can be influenced by the specific trials included and the statistical models used. Real-world evidence provides a practical perspective. One retrospective study compared cabozantinib and Axitinib as second-line treatments following progression on first-line immunotherapy. The study found that the overall effectiveness was similar, with a median PFS of 11.0 months for cabozantinib and 9.5 months for Axitinib. However, a notable difference emerged in the poor-risk subgroup, where cabozantinib appeared to be more effective. Cabozantinib was associated with slightly more serious side effects.[51]

5.2 Interpreting Safety Profiles: Monotherapy vs. Combination Regimens

The choice of TKI is often driven as much by its safety and tolerability profile as by its efficacy. This decision has become more complex with the advent of combination therapies.

  • Distinct Monotherapy Profiles: Each TKI has a characteristic toxicity profile. Sunitinib is frequently associated with fatigue, hematologic toxicities, and gastrointestinal issues.[44] Pazopanib is noted for a higher incidence of liver function test elevations.[44] Axitinib's profile is dominated by on-target VEGFR-related toxicities, such as hypertension, dysphonia (hoarseness), and diarrhea.[29] Cabozantinib also causes significant diarrhea, hypertension, and hand-foot syndrome.[44]
  • The Increased Burden of Combination Therapy: A consistent finding across multiple analyses is that the combination of a TKI with an ICI results in a significantly poorer safety profile compared to TKI monotherapy.[53] This is not merely an additive effect; the combination can exacerbate known toxicities and introduce new, immune-related adverse events. For instance, a meta-analysis showed that the rate of treatment discontinuation due to adverse events for Axitinib was 8.9% when used as a single agent but nearly tripled to 26.6% when used in combination regimens.[54] This increased toxicity creates a critical efficacy-toxicity trade-off. While combination therapies like Axitinib plus pembrolizumab offer unprecedented survival benefits and are the standard of care for most eligible patients, the significant increase in the risk of severe adverse events and treatment discontinuation cannot be ignored. This dynamic preserves a clinical niche for TKI monotherapy, particularly for patients who may not be suitable candidates for the more aggressive combination approach, such as the elderly or those with significant comorbidities.[49] Therefore, treatment selection in the modern era of mRCC requires a highly individualized, patient-centered discussion that carefully weighs the profound efficacy gains of combination therapy against its substantial toxicity burden.

Table 5: Comparative Safety Profile of Axitinib vs. Other TKIs in Advanced RCC (Monotherapy)

Adverse EventAxitinibSunitinibPazopanibCabozantinib
HypertensionVery Common (40% all grades; 16% Grade ≥3) 24CommonCommonCommon 44
DiarrheaVery Common (55% all grades; 11% Grade ≥3) 38Common, often a dose-limiting toxicity 44Less prominent than sunitinibVery Common 44
Hand-Foot SyndromeLess common than sorafenib/sunitinib (27%) 29Very Common and often severe 44Less common than sunitinibCommon 44
Fatigue/AstheniaVery Common (39% all grades; 11% Grade ≥3) 38Very CommonCommonCommon 44
Hepatotoxicity (LFTs)Can occur; increased with pembrolizumab 38Less common than pazopanibMore prominent than sunitinib 44Can occur
Nausea/VomitingCommon (32% Nausea, 24% Vomiting) 17CommonCommonCommon
Dysphonia (Hoarseness)Common (31%) 29Less commonLess commonLess common

6.0 Safety, Tolerability, and Risk Management

A thorough understanding of Axitinib's safety profile and a proactive approach to risk management are essential for optimizing patient outcomes and maintaining treatment adherence.

6.1 Comprehensive Adverse Event Profile

The adverse events associated with Axitinib are largely predictable and related to its potent inhibition of the VEGFR pathway. The profile can be more complex when used in combination with an ICI.

  • Serious Adverse Events: Axitinib is associated with several serious and potentially life-threatening adverse events that require vigilant monitoring and immediate intervention:
  • High Blood Pressure (Hypertension): Hypertension is very common and can be severe, including instances of hypertensive crisis.[17]
  • Thromboembolic Events: Both arterial (e.g., heart attack, stroke) and venous (e.g., deep vein thrombosis, pulmonary embolism) blood clots have been observed and can be fatal.[17]
  • Hemorrhage: Bleeding events, some of which have been fatal, have been reported. Patients with untreated brain metastases or recent gastrointestinal bleeding were excluded from trials and should not receive Axitinib.[17]
  • Heart Failure: Cases of cardiac failure, which can be fatal, have been observed.[17]
  • Gastrointestinal Perforation and Fistula: These are rare but serious complications, including fatal cases, that can occur. Caution is advised in patients at risk.[17]
  • Thyroid Dysfunction: Hypothyroidism is a common class effect of VEGFR TKIs and may require thyroid hormone replacement therapy.[17]
  • Impaired Wound Healing: Due to its anti-angiogenic mechanism, Axitinib can interfere with wound healing.[17]
  • Reversible Posterior Leukoencephalopathy Syndrome (RPLS): This is a rare neurological disorder characterized by headache, seizures, confusion, and vision loss, which has been observed in patients treated with Axitinib. Treatment should be permanently discontinued if RPLS occurs.[16]
  • Proteinuria: The development of protein in the urine should be monitored before and during treatment. Dose reduction or interruption may be necessary.[17]
  • Hepatotoxicity: Liver enzyme elevations can occur. The frequency of Grade 3 and 4 elevations is higher when Axitinib is used in combination with pembrolizumab.[17]
  • Common Adverse Events: The most frequently reported adverse events include:
  • When used as monotherapy: Diarrhea, hypertension, fatigue, decreased appetite, nausea, dysphonia (hoarseness), and palmar-plantar erythrodysesthesia (hand-foot syndrome) are the most common.[1]
  • When used with pembrolizumab: The profile includes diarrhea, fatigue/asthenia, hypertension, hepatotoxicity, hypothyroidism, decreased appetite, rash, and stomatitis (mouth sores).[17]

6.2 Management of Clinically Important Toxicities

Proactive management strategies are crucial for mitigating the impact of Axitinib's side effects.

  • Hypertension: This is the most common dose-limiting toxicity. It is imperative that patients' blood pressure is well-controlled before starting Axitinib. Blood pressure should be monitored regularly throughout treatment, as increases can occur as early as four days after initiation. Standard anti-hypertensive medications should be used as needed. If hypertension persists despite medical management, the Axitinib dose should be reduced. In cases of severe and persistent hypertension or hypertensive crisis, Axitinib should be discontinued.[24]
  • Wound Healing: To minimize the risk of wound healing complications, it is recommended to stop Axitinib at least 24 to 48 hours prior to any scheduled surgery. The healthcare provider will determine when it is safe to resume treatment post-surgery.[17]

6.3 Drug-Drug Interactions, Contraindications, and Special Populations

Axitinib's pharmacokinetic profile necessitates careful consideration of concomitant medications and specific patient populations.

  • CYP3A4/5 Interactions:
  • Inhibitors: Co-administration with strong inhibitors of CYP3A4/5 (e.g., ketoconazole, itraconazole, clarithromycin) should be avoided. These agents can increase Axitinib plasma concentrations by up to two-fold, thereby increasing the risk of toxicity. If co-administration is unavoidable, a dose reduction of Axitinib is recommended.[6]
  • Inducers: Co-administration with strong inducers of CYP3A4/5 (e.g., rifampin, carbamazepine, phenytoin, St. John's Wort) should be avoided. These agents can dramatically decrease Axitinib plasma concentrations by over 70%, potentially rendering the treatment ineffective.[6]
  • Dietary Interactions: Patients should be advised to avoid eating grapefruit or drinking grapefruit juice, as these are well-known inhibitors of CYP3A4 and can increase Axitinib plasma levels.[17]
  • Contraindications and Cautions: Axitinib has not been studied in patients with untreated brain metastases or recent active gastrointestinal bleeding and should not be used in these populations.[24] Caution is warranted in patients at risk for gastrointestinal perforation.[24]
  • Special Populations:
  • Pregnancy and Fertility: Axitinib can cause fetal harm and should not be used during pregnancy. Women of childbearing potential must have a negative pregnancy test before starting treatment and use effective contraception during treatment and for one week after the final dose. Male patients with female partners who can become pregnant must also use effective contraception during the same period. Axitinib may also impair fertility in both men and women.[17]
  • Hepatic Impairment: Patients with moderate hepatic impairment have approximately double the systemic exposure to Axitinib. Therefore, a reduction in the starting dose is required for these patients.[11]

Table 6: Common and Serious Adverse Events Associated with Axitinib

CategoryAdverse EventDescription and ManagementSource(s)
Common (≥20%)DiarrheaFrequent loose stools. Manage with anti-diarrheal agents, hydration, and dietary modification. Dose modification may be needed.17
HypertensionElevated blood pressure. Must be well-controlled pre-initiation. Monitor frequently and treat with anti-hypertensives. Dose reduce or discontinue if severe.17
Fatigue/AstheniaFeeling of tiredness and weakness. Manage with activity pacing and supportive care.17
Decreased AppetiteLoss of interest in food. Encourage small, frequent, nutrient-dense meals.17
Nausea/VomitingManage with anti-emetics and hydration.17
DysphoniaHoarseness or change in voice quality.29
SeriousThromboembolic EventsArterial or venous blood clots. Can be fatal. Requires immediate medical attention.17
HemorrhageSerious bleeding. Can be fatal. Requires immediate medical attention.17
GI PerforationTear in the stomach or intestinal wall. A medical emergency.17
Heart FailureCan be fatal. Monitor for signs such as shortness of breath and edema.17
RPLSRare neurological syndrome. Permanently discontinue Axitinib if it occurs.16
HepatotoxicityLiver enzyme elevation. Monitor LFTs before and during treatment. More frequent with ICI combination.17

Table 7: Clinically Significant Drug-Drug Interactions with Axitinib

Interacting Agent ClassExamplesEffect on Axitinib Plasma LevelsClinical RecommendationSource(s)
Strong CYP3A4/5 InhibitorsKetoconazole, Itraconazole, Clarithromycin, Grapefruit JuiceIncrease (up to 2-fold)Avoid co-administration. If necessary, reduce Axitinib starting dose by approximately half.6
Strong CYP3A4/5 InducersRifampin, Carbamazepine, Phenytoin, St. John's WortDecrease (by >70%)Avoid co-administration. If necessary, a dose increase of Axitinib may be considered.6

7.0 Global Regulatory History

Axitinib's path to becoming a global standard of care for advanced RCC is reflected in its approval history with major regulatory agencies, which followed a logical progression from second-line monotherapy to first-line combination therapy.

7.1 U.S. Food and Drug Administration (FDA)

The FDA's approval timeline for Axitinib (Inlyta) highlights its expanding role in RCC treatment.

  • Initial Approval (Second-Line Monotherapy): On December 7, 2011, the FDA's Oncologic Drugs Advisory Committee (ODAC) voted unanimously to recommend the approval of Axitinib for the second-line treatment of advanced RCC.[8] Following this recommendation, the FDA granted full approval on January 27, 2012, for the treatment of advanced RCC after the failure of one prior systemic therapy.[6] This approval was based on the robust data from the AXIS trial, which demonstrated a significant PFS advantage for Axitinib over sorafenib.[32]
  • First-Line Combination Approvals: The subsequent success of Axitinib in combination with ICIs led to two critical label expansions, establishing it as a cornerstone of first-line therapy.
  • With Pembrolizumab: On April 22, 2019, the FDA approved the combination of Axitinib and pembrolizumab for the first-line treatment of patients with advanced RCC, based on the superior OS, PFS, and ORR demonstrated in the KEYNOTE-426 trial.[42]
  • With Avelumab: Shortly thereafter, on May 14, 2019, the FDA approved the combination of Axitinib and avelumab for the same indication, based on the positive PFS results from the JAVELIN Renal 101 trial.[56]

7.2 European Medicines Agency (EMA)

The EMA's review and approval process mirrored that of the FDA, establishing Axitinib's role in Europe.

  • Initial Approval: Following a positive opinion from the Committee for Medicinal Products for Human Use (CHMP), the European Commission granted a marketing authorisation for Inlyta on September 3, 2012.[8] The approved indication was for the treatment of adult patients with advanced RCC after failure of prior treatment with sunitinib or a cytokine.[27] The CHMP's recommendation was based on the results of the AXIS trial, which showed that Axitinib extended the median time patients lived without their disease worsening by two months compared to sorafenib (6.7 months vs. 4.7 months).[27] The committee concluded that the drug's benefits outweighed its risks, noting that its side effects were manageable and consistent with its class.[27]
  • Recent Developments: The patent protection for Axitinib has led to the development of generic versions. In July 2024, the CHMP adopted a positive opinion recommending the granting of a marketing authorization for a generic medicinal product, Axitinib Accord, for the treatment of adults with RCC.[8]

8.0 Mechanisms of Therapeutic Resistance

While VEGFR TKIs like Axitinib have transformed the treatment of mRCC, the development of therapeutic resistance, either intrinsic (primary) or acquired (secondary), remains a major clinical challenge that ultimately limits their long-term efficacy.[59] Understanding the molecular mechanisms underlying this resistance is critical for developing strategies to overcome or delay it.

8.1 Upregulation of Alternative Proangiogenic Pathways (Angiogenic Escape)

One of the primary mechanisms of acquired resistance to anti-VEGF therapy is the activation of alternative, redundant proangiogenic signaling pathways. Effective blockade of the VEGFR pathway by Axitinib can induce tumor hypoxia, which paradoxically acts as a powerful stimulus for the tumor to upregulate other growth factors and receptors to restore its blood supply, a process often termed "angiogenic escape" or "angiogenic switch".[62]

  • Key Alternative Pathways:
  • HGF/c-Met Pathway: The hepatocyte growth factor (HGF) and its receptor, c-Met, represent a critical escape pathway. Overexpression of HGF and/or c-Met has been identified as a key driver of resistance to anti-VEGF therapies in RCC and other tumors.[48] Preclinical studies have shown that combining a VEGFR inhibitor with a c-Met inhibitor like crizotinib can enhance anti-tumor activity in sunitinib-resistant RCC models, providing a strong rationale for targeting this pathway.[59]
  • Fibroblast Growth Factor (FGF) Pathway: Upregulation of fibroblast growth factors (e.g., FGF1, FGF2) and their receptors (FGFRs) can also mediate resistance by activating pro-survival and pro-angiogenic signaling cascades like the MAPK/ERK and PI3K/Akt pathways.[63]
  • Other Proangiogenic Factors: A host of other factors have been implicated in angiogenic escape, including interleukins (IL-6, IL-8), angiopoietins (Ang1/2), and ephrins (EFNA1/2). Elevated levels of these cytokines have been correlated with shorter PFS and OS in patients treated with TKIs.[63]

8.2 The Role of the Tumor Microenvironment (TME) and Intracellular Mechanisms

Resistance is not solely driven by tumor cells but is also heavily influenced by complex interactions within the tumor microenvironment and by cell-intrinsic adaptations.

  • Immunomodulation and the TME: The TME is a dynamic ecosystem of cancer cells, immune cells, stromal cells, and vasculature. VEGFR inhibitors, including Axitinib, can modulate this environment. There is emerging evidence that Axitinib's interaction with the immune system may be dose-dependent. A compelling case report described a patient with RCC resistant to both anti-PD-1 therapy and standard-dose Axitinib. In this patient, standard-dose Axitinib was associated with an increase in peripheral immunosuppressive regulatory T-cells (Tregs). However, upon reducing the Axitinib dose, the Treg population decreased, and the patient achieved a partial response to the combination therapy.[64] This suggests that while high-dose Axitinib is potently anti-angiogenic, a lower dose might create a more favorable immune microenvironment, thereby reversing resistance to immunotherapy. This highlights Axitinib's potential dual role as both an anti-angiogenic agent and an immunomodulator, a concept that is central to its success in combination regimens.
  • Intracellular Signaling and Drug Efflux: Tumor cells can develop resistance through intrinsic mechanisms. For example, studies have shown that downregulation of the tumor suppressor protein Keap1 can lead to the stabilization and upregulation of the transcription factor Nrf2. Elevated Nrf2 activity can protect cancer cells from oxidative stress and has been linked to Axitinib resistance in RCC cell lines.[66] Additionally, general mechanisms of drug resistance, such as the sequestration of TKI molecules within cellular lysosomes or their active removal from the cell by efflux pumps like the multidrug resistance pump, can reduce the effective intracellular concentration of the drug, leading to treatment failure.[3]

8.3 Strategies to Overcome or Delay Resistance

The understanding of these resistance mechanisms informs the development of next-generation therapeutic strategies.

  • Rational Combination Therapy: This is the most successful strategy to date. By combining Axitinib with an agent that has a different mechanism of action, such as an ICI, it is possible to attack the tumor on multiple fronts simultaneously. The ICI can overcome immune evasion while the TKI suppresses angiogenesis and potentially modulates the TME to be more permissive to an anti-tumor immune response.[39]
  • Sequential Therapy: Using different TKIs in sequence can be an effective strategy. Because different TKIs have varying affinities for VEGFRs and other off-target kinases, a tumor that has become resistant to one agent (e.g., sunitinib) may still be sensitive to another (e.g., Axitinib).[5] The AXIS trial provided robust evidence for this concept.[29]
  • Targeting Specific Resistance Pathways: A more targeted approach involves combining Axitinib with an inhibitor of a known escape pathway. As mentioned, preclinical data support the combination of Axitinib with a c-Met inhibitor to overcome resistance mediated by that pathway.[59]

9.0 Future Perspectives and Ongoing Research

Axitinib's established efficacy and manageable safety profile have positioned it as a versatile agent for ongoing research, with investigations aimed at optimizing its use in RCC and expanding its application to other cancers.

9.1 Expanding Roles in RCC Treatment

Research continues to explore the utility of Axitinib across the full spectrum of RCC disease states, beyond its approved indications in the metastatic setting.

  • Neoadjuvant and Adjuvant Therapy: A significant area of investigation is the use of Axitinib in the perioperative setting. Neoadjuvant therapy (given before surgery) with Axitinib, often in combination with an ICI like toripalimab or pembrolizumab, is being studied in patients with locally advanced RCC.[46] The goals are to shrink the primary tumor and any associated tumor thrombus to facilitate a safer and more complete surgical resection, and to treat micrometastatic disease early. Early-phase trials have shown that this approach is feasible, with encouraging objective response rates and acceptable toxicity.[46] Adjuvant therapy (given after surgery) is another potential application, aiming to eradicate residual disease and prevent recurrence in high-risk patients.[69]
  • Third-line and Beyond: While Axitinib is approved for second-line monotherapy, real-world evidence and clinical practice demonstrate its continued activity in later lines of treatment. A retrospective study found Axitinib to be a safe and effective option for patients receiving it as a third-line or subsequent therapy, highlighting its durable role in the sequential treatment of mRCC.[71]

9.2 Novel Combination Strategies

The success of combining Axitinib with pembrolizumab and avelumab has spurred extensive research into other novel combination strategies, primarily with next-generation immunotherapies.

  • Next-Generation ICI Combinations: The Phase III RENOTORCH trial, conducted in China, evaluated Axitinib in combination with toripalimab, an anti-PD-1 antibody. The study met its primary endpoint, showing that the combination significantly improved PFS and ORR compared to sunitinib monotherapy in patients with intermediate- to high-risk advanced RCC.[46] This led to the approval of the combination in China and provides further evidence for the efficacy of the Axitinib-ICI backbone.[73] Numerous other trials are actively investigating Axitinib in combination with other ICIs, including nivolumab, and even triplet combinations with agents like tiragolumab (an anti-TIGIT antibody).[68]

9.3 Investigational Roles in Other Malignancies

Given that angiogenesis is a hallmark of many solid tumors, Axitinib's potent anti-VEGFR activity has prompted its investigation in a variety of other cancers.

  • Broad Anti-Tumor Activity: Early clinical trials and preclinical models have demonstrated Axitinib's anti-tumor activity in malignancies beyond RCC, including breast cancer, metastatic melanoma, thyroid cancer, and advanced non-small cell lung cancer (NSCLC).[4] Active clinical trials are currently exploring Axitinib, typically in combination with ICIs, for the treatment of advanced mucosal melanoma and head and neck cancers.[68]
  • Targeting Specific Mutations: An intriguing area of research stems from a 2015 study which found that Axitinib can effectively inhibit the BCR-ABL1 fusion protein, specifically the T315I mutant isoform.[8] This mutation confers resistance to standard TKIs like imatinib in patients with chronic myeloid leukemia (CML) and adult acute lymphoblastic leukemia (ALL). This finding suggests a potential niche application for Axitinib in hematologic malignancies characterized by this specific resistance mutation, an area of application completely distinct from its primary role in solid tumors.

10.0 Expert Synthesis and Strategic Recommendations

10.1 Synthesis of Axitinib's Profile

Axitinib has firmly established itself as a critical component in the therapeutic armamentarium for advanced renal cell carcinoma. Its clinical profile is defined by its high potency and selectivity as a second-generation VEGFR tyrosine kinase inhibitor, which translates into a robust anti-angiogenic effect. Its clinical journey reflects the broader evolution of mRCC treatment over the past decade. It was initially validated as a superior second-line monotherapy to sorafenib based on a clear progression-free survival benefit in the AXIS trial. Subsequently, it was repositioned as a cornerstone of first-line therapy through its synergistic and survival-prolonging partnership with immune checkpoint inhibitors in the KEYNOTE-426 and JAVELIN Renal 101 trials.

The drug's pharmacologic characteristics—a short half-life supporting twice-daily dosing, a predictable on-target toxicity profile dominated by hypertension, and a clear exposure-response relationship—allow for an individualized dose-titration strategy. The management of its on-target toxicities, particularly hypertension, is not merely a supportive care measure but an integral part of optimizing therapy, as blood pressure elevation serves as a pharmacodynamic biomarker of target engagement.

10.2 Clinical Positioning and Recommendations

Based on the comprehensive evidence, the strategic positioning of Axitinib can be summarized as follows:

  • First-Line Metastatic RCC: For treatment-naïve patients with advanced clear-cell RCC who are eligible for combination therapy, an Axitinib-based regimen (in combination with pembrolizumab or avelumab) represents a standard of care. This approach offers a proven overall survival benefit compared to TKI monotherapy. The clinical decision-making process must involve a careful assessment of the patient's performance status, comorbidities, and potential to tolerate the compounded toxicities of the TKI-ICI combination. Proactive management of both TKI-related and immune-related adverse events is paramount.
  • Second-Line and Subsequent Therapy: Axitinib monotherapy remains a highly effective and important option in the second-line setting and beyond. It is a standard of care for patients who have progressed after a first-line therapy, particularly in contexts where prior treatment did not include Axitinib (e.g., following an ipilimumab/nivolumab combination or in regions where TKI monotherapy is still used first-line). Its efficacy after progression on sunitinib is well-established by the AXIS trial. Real-world data also support its use in later lines of therapy, providing a valuable option for heavily pre-treated patients.

10.3 Future Research Imperatives

To further refine the role of Axitinib and improve outcomes for patients, future research should prioritize several key areas:

  • Optimizing Combination Therapy: The primary challenge with current Axitinib-ICI combinations is toxicity management. Future research should focus on alternative dosing strategies, such as adaptive dosing or lower continuous dosing schedules, to determine if the synergistic efficacy can be maintained while mitigating the high rates of adverse events and treatment discontinuation. Understanding the dose-dependent immunomodulatory effects of Axitinib is crucial to this effort.
  • Biomarker Development: The field urgently needs robust predictive biomarkers to guide the selection of first-line therapy. Research should move beyond PD-L1 status to investigate more sophisticated composite biomarkers (e.g., genomic signatures, TME characteristics, circulating tumor DNA) that can identify patients most likely to derive maximal benefit from an Axitinib-ICI combination versus an alternative regimen (e.g., dual ICI or another TKI-ICI pair). Further prospective validation of hypertension as a pharmacodynamic biomarker could help guide real-time dose optimization.
  • Overcoming Resistance: As resistance to Axitinib-ICI combinations inevitably occurs, a deeper understanding of the underlying mechanisms is essential. Future clinical trials should focus on rationally designed triplet or sequential therapies that target known resistance pathways, such as the c-Met or FGF pathways, to extend the duration of clinical benefit and provide effective options for patients upon progression.

11.0 References

[1]

Works cited

  1. Axitinib: Newly Approved for Renal Cell Carcinoma - PMC, accessed July 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4093354/
  2. Axitinib: A Review of its Safety and Efficacy in the Treatment of Adults with Advanced Renal Cell Carcinoma, accessed July 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3825605/
  3. Renal Cell Cancer – Insights in Drug Resistance Mechanisms - Anticancer Research, accessed July 23, 2025, https://ar.iiarjournals.org/content/anticanres/43/11/4781.full-text.pdf
  4. Axitinib: A Review of its Safety and Efficacy in the Treatment of Adults with Advanced Renal Cell Carcinoma - ResearchGate, accessed July 23, 2025, https://www.researchgate.net/publication/258703636_Axitinib_A_Review_of_its_Safety_and_Efficacy_in_the_Treatment_of_Adults_with_Advanced_Renal_Cell_Carcinoma
  5. Renal Cell Cancer – Insights in Drug Resistance Mechanisms - Anticancer Research, accessed July 23, 2025, https://ar.iiarjournals.org/content/43/11/4781
  6. What is Axitinib used for? - Patsnap Synapse, accessed July 23, 2025, https://synapse.patsnap.com/article/what-is-axitinib-used-for
  7. What is the mechanism of Axitinib? - Patsnap Synapse, accessed July 23, 2025, https://synapse.patsnap.com/article/what-is-the-mechanism-of-axitinib
  8. Axitinib - Wikipedia, accessed July 23, 2025, https://en.wikipedia.org/wiki/Axitinib
  9. Axitinib | C22H18N4OS | CID 6450551 - PubChem, accessed July 23, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Axitinib
  10. go.drugbank.com, accessed July 23, 2025, https://go.drugbank.com/drugs/DB06626#:~:text=Axitinib%20is%20a%20second%20generation,2%2C%20VEGFR%2D3).&text=Through%20this%20mechanism%20of%20action,angiogenesis%2C%20tumour%20growth%20and%20metastases..%26text%3DThrough%2520this%2520mechanism%2520of%2520action,angiogenesis%252C%2520tumour%2520growth%2520and%2520metastases.&sa=D&source=editors&ust=1753250264349027&usg=AOvVaw2uVfNag4D7wZnkShR4stl-)
  11. Clinical pharmacology of axitinib - PubMed, accessed July 23, 2025, https://pubmed.ncbi.nlm.nih.gov/23677771/
  12. Axitinib | AG013736 | CAS#319460-85-0 - MedKoo Biosciences, accessed July 23, 2025, https://www.medkoo.com/products/4585
  13. Axitinib (AG-013736, CAS Number: 319460-85-0) | Cayman Chemical, accessed July 23, 2025, https://www.caymanchem.com/product/13813/axitinib
  14. Axitinib | CAS 319460-85-0 | VEGFR kinase inhibitor - StressMarq Biosciences Inc., accessed July 23, 2025, https://www.stressmarq.com/products/small-molecules/inhibitor/axitinib-sih-494/
  15. Definition of axitinib - NCI Drug Dictionary, accessed July 23, 2025, https://www.cancer.gov/publications/dictionaries/cancer-drug/def/axitinib
  16. Axitinib (Inlyta) | Cancer information, accessed July 23, 2025, https://www.cancerresearchuk.org/about-cancer/treatment/drugs/axitinib
  17. INLYTA® (axitinib) Home Page | Safety Info, accessed July 23, 2025, https://www.inlyta.com/
  18. pubchem.ncbi.nlm.nih.gov, accessed July 23, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Axitinib#:~:text=Axitinib%20is%20an%20indazole%20substituted,a%20first%20line%20systemic%20treatment.
  19. www.tocris.com, accessed July 23, 2025, https://www.tocris.com/products/axitinib_4350#:~:text=Preparing%20Stock%20Solutions%20for%20Axitinib,the%20product%20molecular%20weight%20386.47.
  20. Compound: AXITINIB (CHEMBL1289926) - ChEMBL - EMBL-EBI, accessed July 23, 2025, https://www.ebi.ac.uk/chembl/explore/compound/CHEMBL1289926
  21. 319460-85-0 Axitinib AKSci A819, accessed July 23, 2025, https://aksci.com/item_detail.php?cat=A819
  22. Population pharmacokinetic analysis of axitinib in healthy volunteers - PMC, accessed July 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3952722/
  23. INLYTA® (axitinib) Clinical Pharmacology | Pfizer Medical Information - US, accessed July 23, 2025, https://www.pfizermedicalinformation.com/inlyta/clinical-pharmacology
  24. Inlyta (axitinib) tablets Label - accessdata.fda.gov, accessed July 23, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/202324lbl.pdf
  25. Axitinib for the Management of Metastatic Renal Cell Carcinoma - PMC, accessed July 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3585900/
  26. Axitinib versus sorafenib in advanced renal cell carcinoma ..., accessed July 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4056058/
  27. Inlyta | European Medicines Agency (EMA), accessed July 23, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/inlyta
  28. Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial | Request PDF - ResearchGate, accessed July 23, 2025, https://www.researchgate.net/publication/51772730_Comparative_effectiveness_of_axitinib_versus_sorafenib_in_advanced_renal_cell_carcinoma_AXIS_a_randomised_phase_3_trial
  29. Axitinib versus sorafenib as second-line therapy for metastatic renal ..., accessed July 23, 2025, https://ascopubs.org/doi/10.1200/jco.2011.29.15_suppl.4503
  30. Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): A randomised phase 3 trial - Northwestern Scholars, accessed July 23, 2025, https://www.scholars.northwestern.edu/en/publications/comparative-effectiveness-of-axitinib-versus-sorafenib-in-advance
  31. Study Results | Axitinib (AG 013736) As Second Line Therapy For Metastatic Renal Cell Cancer | ClinicalTrials.gov, accessed July 23, 2025, https://clinicaltrials.gov/study/NCT00678392?tab=results
  32. U.S. Food And Drug Administration Approves Pfizer's INLYTA® (axitinib) For Patients With Previously Treated Advanced Renal Cell Carcinoma (RCC), accessed July 23, 2025, https://www.pfizer.com/news/press-release/press-release-detail/u_s_food_and_drug_administration_approves_pfizer_s_inlyta_axitinib_for_patients_with_previously_treated_advanced_renal_cell_carcinoma_rcc
  33. INLYTA® (axitinib) Clinical Trial Results | Safety Info, accessed July 23, 2025, https://www.inlyta.com/axitinib/clinical-trial-results
  34. The European Medicines Agency approval of axitinib (Inlyta) for the treatment of advanced renal cell carcinoma after failure of prior treatment with sunitinib or a cytokine - PubMed, accessed July 23, 2025, https://pubmed.ncbi.nlm.nih.gov/25616431/
  35. Axitinib after Treatment Failure with Sunitinib or Cytokines in Advanced Renal Cell Carcinoma—Systematic Literature Review of Clinical and Real-World Evidence - MDPI, accessed July 23, 2025, https://www.mdpi.com/2072-6694/16/15/2706
  36. The European Medicines Agency Approval of Axitinib (Inlyta) for the Treatment of Advanced Renal Cell Carcinoma After Failure of Prior Treatment With Sunitinib or a Cytokine: Summary of the Scientific Assessment of the Committee for Medicinal Products for Human Use - ResearchGate, accessed July 23, 2025, https://www.researchgate.net/publication/271329753_The_European_Medicines_Agency_Approval_of_Axitinib_Inlyta_for_the_Treatment_of_Advanced_Renal_Cell_Carcinoma_After_Failure_of_Prior_Treatment_With_Sunitinib_or_a_Cytokine_Summary_of_the_Scientific_Ass
  37. Real-world experience of second-line axitinib in metastatic renal cell carcinoma: analysis of the Swedish population, accessed July 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11376413/
  38. INLYTA® (axitinib) Mechanism Of Action | 2nd-Line | Safety Info, accessed July 23, 2025, https://inlyta.pfizerpro.com/monotherapy/vegfr-pathway
  39. Full article: Second-Line Treatment with Axitinib Plus Toripalimab in Metastatic Renal Cell Carcinoma: A Retrospective Multicenter Study - Taylor & Francis Online, accessed July 23, 2025, https://www.tandfonline.com/doi/full/10.2217/fon-2021-1267
  40. Axitinib: From preclinical development to future clinical perspectives in renal cell carcinoma | Request PDF - ResearchGate, accessed July 23, 2025, https://www.researchgate.net/publication/277782417_Axitinib_From_preclinical_development_to_future_clinical_perspectives_in_renal_cell_carcinoma
  41. Clinical Trial Results in Patients With Advanced Kidney Cancer (Renal Cell Carcinoma) - Keytruda, accessed July 23, 2025, https://www.keytruda.com/kidney-cancer/combination-therapy-clinical-trial-results/
  42. Combining Immunotherapy and Targeted Therapy for Kidney Cancer | The AACR, accessed July 23, 2025, https://www.aacr.org/patients-caregivers/progress-against-cancer/combining-immunotherapy-and-targeted-therapy-for-kidney-cancer/
  43. KEYTRUDA® (pembrolizumab) in Combination with Inlyta® (axitinib) Reduced Risk of Death by Nearly Half Compared to Sunitinib as First-Line Treatment for Advanced Renal Cell Carcinoma (RCC) - Merck.com, accessed July 23, 2025, https://www.merck.com/news/keytruda-pembrolizumab-in-combination-with-inlyta-axitinib-reduced-risk-of-death-by-nearly-half-compared-to-sunitinib-as-first-line-treatment-for-advanced-renal-cell-carcinoma-rcc/
  44. Recap: Safety and Efficacy of Available Treatment Options and Considerations for Management of RCC - CancerNetwork, accessed July 23, 2025, https://www.cancernetwork.com/view/recap-safety-and-efficacy-of-available-treatment-options-and-considerations-for-management-of-rcc
  45. Real-World Study Supports Avelumab Plus Axitinib in RCC - Targeted Oncology, accessed July 23, 2025, https://www.targetedonc.com/view/real-world-study-supports-avelumab-plus-axitinib-in-rcc
  46. Neoadjuvant toripalimab combined with axitinib in patients with locally advanced clear cell renal cell carcinoma: a single-arm, phase II trial | Journal for ImmunoTherapy of Cancer, accessed July 23, 2025, https://jitc.bmj.com/content/12/6/e008475
  47. Cabozantinib Versus Sunitinib As Initial Targeted Therapy for Patients With Metastatic Renal Cell Carcinoma of Poor or Intermedi - Cancer Center of Kansas, accessed July 23, 2025, https://cancercenterofkansas.com/wp-content/uploads/2016/11/JCO_Publishment_CABOzantinib-vs-SUNitinib-11.15.16.pdf
  48. CABOSUN Trial: Cabozantinib vs Sunitinib for Renal Cell Carcinoma - Targeted Oncology, accessed July 23, 2025, https://www.targetedonc.com/view/cabosun-trial-cabozantinib-vs-sunitinib-for-renal-cell-carcinoma
  49. Efficacy and Safety of Approved First-Line Tyrosine Kinase Inhibitor ..., accessed July 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7004428/
  50. Axitinib, cabozantinib, everolimus, nivolumab, sunitinib and best ..., accessed July 23, 2025, https://njl-admin.nihr.ac.uk/document/download/2012065
  51. Comparison of cabozantinib with axitinib as second-line treatment for metastatic kidney cancer, accessed July 23, 2025, https://actionkidneycancer.org/real-world-comparison-of-cabozantinib-with-axitinib-as-second-line-treatment-after-immunotherapy-for-metastatic-kidney-cancer/
  52. Sunitinib versus Pazopanib Dilemma in Renal Cell Carcinoma: New Insights into the In Vitro Metabolic Impact, Efficacy, and Safety - MDPI, accessed July 23, 2025, https://www.mdpi.com/1422-0067/23/17/9898
  53. Comparative safety of tyrosine kinase inhibitors in the treatment of metastatic renal cell carcinoma: a systematic review and network meta-analysis - PMC, accessed July 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10512702/
  54. Tolerability of axitinib in advanced renal cell carcinoma: A meta-analysis., accessed July 23, 2025, https://ascopubs.org/doi/10.1200/JCO.2021.39.15_suppl.e16536
  55. Axitinib - NCI, accessed July 23, 2025, https://www.cancer.gov/about-cancer/treatment/drugs/axitinib
  56. Inlyta (axitinib) FDA Approval History - Drugs.com, accessed July 23, 2025, https://www.drugs.com/history/inlyta.html
  57. Label: INLYTA- axitinib tablet, film coated - DailyMed, accessed July 23, 2025, https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=8a903e31-936e-4ed7-8a59-59f32374f338
  58. The European Medicines Agency Approval of Axitinib (Inlyta) for the Treatment of Advanced Renal Cell Carcinoma After Failure of Prior Treatment With Sunitinib or a Cytokine - PubMed Central, accessed July 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4319625/
  59. Molecular effects of axitinib and crizotinib in the sunitinib-resistant... - ResearchGate, accessed July 23, 2025, https://www.researchgate.net/figure/Molecular-effects-of-axitinib-and-crizotinib-in-the-sunitinib-resistant-RP-R-01-ccRCC_fig6_268039113
  60. Molecular Mechanisms of Drug Resistance in Clear Cell Renal Cell Carcinoma - MDPI, accessed July 23, 2025, https://www.mdpi.com/2072-6694/17/10/1613
  61. Determinants of resistance to VEGF-TKI and immune checkpoint inhibitors in metastatic renal cell carcinoma - PubMed, accessed July 23, 2025, https://pubmed.ncbi.nlm.nih.gov/34099013/
  62. Hypothesized mechanisms of response and resistance to targeted therapy... - ResearchGate, accessed July 23, 2025, https://www.researchgate.net/figure/Hypothesized-mechanisms-of-response-and-resistance-to-targeted-therapy-in-metastatic-RCCC_fig1_41548313
  63. Mechanisms of tyrosine kinase inhibitor resistance in renal cell carcinoma - PMC, accessed July 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10792482/
  64. Axitinib Reverses Resistance to Anti-Programmed Cell Death-1 Therapy in a Patient With Renal Cell Carcinoma - PubMed, accessed July 23, 2025, https://pubmed.ncbi.nlm.nih.gov/34764951/
  65. Axitinib Reverses Resistance to Anti-Programmed Cell ... - Frontiers, accessed July 23, 2025, https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2021.728750/full
  66. Downregulation of Keap1 contributes to poor prognosis and Axitinib ..., accessed July 23, 2025, https://www.spandidos-publications.com/10.3892/ijmm.2019.4134
  67. Neoadjuvant toripalimab combined with axitinib in patients with locally advanced clear cell renal cell carcinoma: a single-arm, phase II trial - PubMed, accessed July 23, 2025, https://pubmed.ncbi.nlm.nih.gov/38862251/
  68. Clinical Trials Using Axitinib - NCI, accessed July 23, 2025, https://www.cancer.gov/research/participate/clinical-trials/intervention/axitinib?pn=1
  69. Axitinib in metastatic renal cell carcinoma: beyond the second-line ..., accessed July 23, 2025, https://pubmed.ncbi.nlm.nih.gov/28707479/
  70. Axitinib: from preclinical development to future clinical perspectives in renal cell carcinoma, accessed July 23, 2025, https://pubmed.ncbi.nlm.nih.gov/26039031/
  71. Axitinib as a third or further line of treatment in renal cancer: a single institution experience, accessed July 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7265645/
  72. RENOTORCH Study of Toripalimab and Axitinib Meets Primary End Point in RCC, accessed July 23, 2025, https://www.targetedonc.com/view/renotorch-study-of-toripalimab-and-axitinib-meets-primary-end-point-in-rcc
  73. Toripalimab plus axitinib approved in China for renal cell carcinoma - Urology Times, accessed July 23, 2025, https://www.urologytimes.com/view/toripalimab-plus-axitinib-approved-in-china-for-renal-cell-carcinoma
  74. Find Kidney Cancer Clinical Trials - gumdrop, accessed July 23, 2025, https://www.gumdroptrials.org/kidney-cancer-trials/
  75. Clinical study | Renal Cell Carcinoma RO7247669 | ForPati, accessed July 23, 2025, https://genentech-clinicaltrials.com/en/trials/cancer/rcc/a-study-of-immune-checkpoint-inhibitor-combinations-wit-47221.html
  76. Axitinib in combination with pembrolizumab in patients with advanced renal cell cancer: a non-randomised, open-label, dose-finding, and dose-expansion phase 1b trial - PMC, accessed July 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6860026/
  77. Tumour Unknown Status Phase 2 Trials for Axitinib (DB06626) | DrugBank Online, accessed July 23, 2025, https://go.drugbank.com/indications/DBCOND0059731/clinical_trials/DB06626?phase=2&status=unknown_status
  78. Carcinoma Completed Phase Trials for Axitinib (DB06626) | DrugBank Online, accessed July 23, 2025, https://go.drugbank.com/indications/DBCOND0022280/clinical_trials/DB06626?status=completed
  79. Axitinib: MedlinePlus Drug Information, accessed July 23, 2025, https://medlineplus.gov/druginfo/meds/a612017.html
  80. medlineplus.gov, accessed July 23, 2025, https://medlineplus.gov/druginfo/meds/a612017.html#:~:text=Axitinib%20is%20used%20in%20combination,of%20medications%20called%20kinase%20inhibitors.
  81. 202324Orig1s000 - accessdata.fda.gov, accessed July 23, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2012/202324Orig1s000MedR.pdf
  82. Axitinib | C22H18N4OS - ChemSpider, accessed July 23, 2025, https://www.chemspider.com/Chemical-Structure.4953153.html
  83. Axitinib | CAS 319460-85-0 | Cayman Chemical | Biomol.com, accessed July 23, 2025, https://www.biomol.com/products/chemicals/biochemicals/axitinib-cay13813-50
  84. Axitinib | C22H18N4OS | CID 6450551 - PubChem, accessed July 23, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/6450551
  85. Axitinib: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed July 23, 2025, https://go.drugbank.com/drugs/DB06626
  86. Study Results | A Study of Avelumab in Combination With Axitinib In Non-Small Cell Lung Cancer (NSCLC) or Urothelial Cancer (Javelin Medley VEGF) | ClinicalTrials.gov, accessed July 23, 2025, https://clinicaltrials.gov/study/NCT03472560?term=AREA%5BBasicSearch%5D(axitinib%20and%20lung%20cancer)&rank=4&tab=results
  87. Safety of Ceasing Combination Therapy After 2 Years in Patients With Renal Cell Carcinoma - The ASCO Post, accessed July 23, 2025, https://ascopost.com/news/february-2025/safety-of-ceasing-combination-therapy-after-2-years-in-patients-with-renal-cell-carcinoma/
  88. Advanced Renal Cell Carcinoma Clinical Trials - NCI - National Cancer Institute, accessed July 23, 2025, https://www.cancer.gov/research/participate/clinical-trials/disease/advanced-renal-cell-carcinoma?pn=1
  89. Neoadjuvant Axitinib in Locally Advanced Renal Cell Carcinoma (RCC) | ClinicalTrials.gov, accessed July 23, 2025, https://clinicaltrials.gov/study/NCT01263769
  90. Chardan Capital downgrades Clearside Biomedical stock rating to Neutral - Investing.com, accessed July 23, 2025, https://in.investing.com/news/analyst-ratings/chardan-capital-downgrades-clearside-biomedical-stock-rating-to-neutral-93CH-4917205
  91. Axitinib Reverses Resistance to Anti-Programmed Cell Death-1 Therapy in a Patient With Renal Cell Carcinoma - Frontiers, accessed July 23, 2025, https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2021.728750/pdf
  92. Neoadjuvant combination of pazopanib or axitinib and programmed cell death protein-1-activated dendritic cell-cytokine-induced killer cells immunotherapy may facilitate surgery in patients with renal cell carcinoma - Translational Andrology and Urology, accessed July 23, 2025, https://tau.amegroups.org/article/view/70505/html
  93. Comparative safety of tyrosine kinase inhibitors in the treatment of metastatic renal cell carcinoma: a systematic review and network meta-analysis - PubMed, accessed July 23, 2025, https://pubmed.ncbi.nlm.nih.gov/37745049/
  94. A Dose Finding Study To Evaluate Safety, Drug Interaction, Tumor Markers Of Axitinib In Combination With MK-3475 In Adult Patients With Previously Untreated Advanced Renal Cell Cancer | ClinicalTrials.gov, accessed July 23, 2025, https://clinicaltrials.gov/study/NCT02133742
  95. Full article: Real-world experience of second-line axitinib in metastatic renal cell carcinoma: analysis of the Swedish population - Taylor and Francis, accessed July 23, 2025, https://www.tandfonline.com/doi/full/10.1080/14796694.2024.2351352
  96. Re: Comparative Effectiveness of Axitinib versus Sorafenib in Advanced Renal Cell Carcinoma (AXIS): A Randomised Phase 3 Trial | Journal of Urology, accessed July 23, 2025, https://www.auajournals.org/doi/10.1016/j.juro.2012.04.069
  97. Axitinib versus sorafenib in advanced renal cell carcinoma: subanalyses by prior therapy from a randomised phase III trial - PubMed, accessed July 23, 2025, https://pubmed.ncbi.nlm.nih.gov/24823696/
  98. Cabozantinib Versus Standard-of-Care Comparators in the Treatment of Advanced/Metastatic Renal Cell Carcinoma in Treatment-naïve Patients: a Systematic Review and Network Meta-Analysis - PubMed Central, accessed July 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5887000/
  99. Comparative effectiveness of pazopanib and sunitinib as first-line therapy for patients with advanced/metastatic renal cell carcinoma in a U.S. community oncology setting. - ASCO, accessed July 23, 2025, https://www.asco.org/abstracts-presentations/ABSTRACT158228
  100. Evolving Treatment Paradigm in Metastatic Renal Cell Carcinoma - ASCO Publications, accessed July 23, 2025, https://ascopubs.org/doi/10.1200/EDBK_174469
  101. Current Technologies and Future Perspectives in Immunotherapy towards a Clinical Oncology Approach - MDPI, accessed July 23, 2025, https://www.mdpi.com/2227-9059/12/1/217

Published at: July 23, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.