MedPath

panitumumab Advanced Drug Monograph

Published:Jul 17, 2025

Generic Name

panitumumab

Brand Names

Vectibix

Drug Type

Biotech

CAS Number

339177-26-3

Associated Conditions

Metastatic Colorectal Cancer (CRC)

Panitumumab (Vectibix®): A Comprehensive Monograph on a Targeted Therapy for Metastatic Colorectal Cancer

Executive Summary and Key Findings

Panitumumab, marketed under the brand name Vectibix®, is a cornerstone of targeted therapy for metastatic colorectal cancer (mCRC). It is a recombinant, fully human IgG2 kappa monoclonal antibody that functions as a high-affinity antagonist of the human epidermal growth factor receptor (EGFR).[1] By competitively inhibiting the binding of endogenous ligands like EGF and TGF-α, panitumumab blocks the downstream signaling cascades—primarily the RAS-RAF-MEK-ERK and PI3K-AKT-mTOR pathways—that drive tumor cell proliferation, survival, and angiogenesis.[3] Its efficacy is critically dependent on the molecular profile of the tumor. The paramount finding from over a decade of clinical research is that panitumumab provides benefit exclusively to patients with wild-type

RAS (both KRAS and NRAS) tumors, and its use in RAS-mutant disease is associated with a lack of benefit and potential harm.[5]

Pivotal clinical trials have precisely defined its role in the mCRC treatment algorithm. The PRIME study established its efficacy in the first-line setting in combination with FOLFOX chemotherapy for RAS wild-type patients, demonstrating significant improvements in both progression-free survival (PFS) and overall survival (OS).[7] The ASPECCT trial confirmed its non-inferiority to the chimeric anti-EGFR antibody cetuximab in the chemorefractory setting, establishing it as a valid therapeutic alternative.[9] More recently, the PARADIGM trial provided practice-changing evidence that for patients with left-sided,

RAS wild-type tumors, first-line treatment with panitumumab plus FOLFOX is superior to bevacizumab plus FOLFOX in extending overall survival, solidifying primary tumor location as a key predictive biomarker.[11] In a strategic reversal of its contraindication in

RAS-mutant disease, panitumumab was recently approved in combination with the KRAS G12C inhibitor sotorasib for KRAS G12C-mutated mCRC, where it functions to block pathway reactivation and overcome therapeutic resistance.[13]

The safety profile of panitumumab is well-characterized and directly linked to its on-target EGFR inhibition in normal tissues. It carries a boxed warning for severe dermatologic toxicity, which occurs in the vast majority of patients and can lead to serious infectious complications.[14] Other clinically significant adverse events include electrolyte disturbances, most notably hypomagnesemia, and a low but serious risk of infusion-related reactions.[5] This report provides an exhaustive analysis of panitumumab's molecular biology, pharmacology, the pivotal clinical data supporting its use, its comprehensive safety profile, and its evolving position within the precision oncology landscape for metastatic colorectal cancer.

Molecular Profile and Pharmacological Action

Identity and Physicochemical Properties

Panitumumab is a protein-based biologic therapeutic classified as a recombinant, fully human IgG2 kappa monoclonal antibody.[1] It is produced using recombinant DNA technology within a mammalian cell line, specifically Chinese Hamster Ovary (CHO) cells.[19] Its fully human nature is a key structural feature, distinguishing it from chimeric antibodies and minimizing its potential for immunogenicity in patients.[17]

The drug is marketed globally by Amgen Inc. under the trade name Vectibix®.[1] During its development, it was also known by the identifier ABX-EGF.[1] Its chemical name is Disulfide with human monoclonal ABX-EGF light chain anti-(human epidermal growth factor receptor) (human monoclonal ABX-EGF heavy chain) immunoglobulin dimer, and it has the molecular formula

C6306​H9732​N1672​O1994​S46​.[22]

As a pharmaceutical product, panitumumab is supplied as a sterile, colorless concentrate for solution for infusion at a concentration of 20 mg/mL.[19] The solution has a pH ranging from 5.6 to 6.0 and may contain visible, translucent-to-white, amorphous proteinaceous particles, which are a normal characteristic of the formulation and are removed by an in-line filter during administration.[19]

Table 1: Panitumumab: Key Drug Identifiers and Properties

PropertyDetailSource(s)
Brand NameVectibix®1
Generic NamePanitumumab1
Drug ClassAntineoplastic Agent; Epidermal Growth Factor Receptor (EGFR) Antagonist1
ManufacturerAmgen Inc.23
DrugBank IDDB012691
CAS Number339177-26-322
Molecular FormulaC6306​H9732​N1672​O1994​S46​22
ATC CodeL01FE0226
Antibody TypeRecombinant Monoclonal Antibody (mAb), IgG2 kappa1
Antibody SourceFully Human24
TargetEpidermal Growth Factor Receptor (EGFR / HER1 / c-ErbB-1)1

The Epidermal Growth Factor Receptor (EGFR) Pathway: The Therapeutic Target

The therapeutic activity of panitumumab is predicated on its interaction with the Epidermal Growth Factor Receptor (EGFR). EGFR, also known as HER1 or by its gene name c-ErbB-1, is a transmembrane glycoprotein belonging to the type I receptor tyrosine kinase subfamily. This family also includes other critical receptors such as HER2 (human epidermal growth factor receptor 2), HER3, and HER4.[1]

Under normal physiological conditions, EGFR is expressed in various epithelial tissues, including the skin and hair follicles, where it plays a fundamental role in regulating cell growth and differentiation.[20] In the context of oncology, EGFR is frequently overexpressed in a wide range of human cancers, most notably in cancers of the colon and rectum.[1] This overexpression can contribute to uncontrolled tumor growth and progression.

The activation of the EGFR pathway is initiated by the binding of its specific ligands, such as Epidermal Growth Factor (EGF) and Transforming Growth Factor-alpha (TGF-α), to the receptor's extracellular domain.[3] This ligand binding induces a conformational change that facilitates receptor dimerization (either homodimerization with another EGFR molecule or heterodimerization with other HER family members). Dimerization, in turn, activates the intracellular tyrosine kinase domain of the receptor, leading to the autophosphorylation of specific tyrosine residues.[1]

These phosphorylated tyrosine residues serve as docking sites for a host of intracellular signaling proteins, triggering the activation of complex downstream signaling cascades. The two most critical pathways for cancer biology that are activated by EGFR are:

  1. The RAS-RAF-MEK-ERK Pathway: This pathway, also known as the MAPK pathway, is a primary regulator of cellular proliferation.[3]
  2. The Phosphatidylinositol 3-Kinase (PI3K)-AKT-mTOR Pathway: This pathway is a central mediator of cell survival, promoting anti-apoptotic signals.[2]

Together, these pathways regulate the transcription of genes essential for cellular growth, survival, motility, and angiogenesis (the formation of new blood vessels to supply the tumor), making the EGFR pathway a prime target for anticancer therapy.[1]

Mechanism of Action: High-Affinity EGFR Blockade

Panitumumab exerts its antineoplastic effects through a direct and potent blockade of the EGFR signaling pathway. It is engineered to bind with very high affinity and specificity to the extracellular ligand-binding domain III of the human EGFR, a site that partially overlaps with the binding site for natural ligands like EGF.[1] The dissociation constant (

KD​), a measure of binding affinity, for panitumumab is approximately 0.05 nM, indicating a significantly tighter bond than that of its natural ligands and even other therapeutic antibodies.[31]

By occupying this critical domain on the receptor, panitumumab acts as a pure antagonist, competitively inhibiting the binding of EGF, TGF-α, and other ligands.[1] This blockade prevents the necessary first steps of pathway activation: ligand-induced receptor dimerization and subsequent autophosphorylation of the intracellular kinase domains.[1]

The ultimate pharmacodynamic consequences of this upstream blockade are the comprehensive inhibition of EGFR-dependent downstream signaling. This results in several key anti-tumor effects:

  • Inhibition of Cell Growth and Proliferation: By shutting down the RAS-RAF-MEK-ERK pathway, panitumumab halts the signals that drive uncontrolled cell division.[3]
  • Induction of Apoptosis: By inhibiting the pro-survival PI3K-AKT-mTOR pathway, the balance shifts towards programmed cell death.[2]
  • Decreased Angiogenesis and Cytokine Production: The blockade of EGFR signaling reduces the tumor's production of key growth factors, such as Vascular Endothelial Growth Factor (VEGF), and pro-inflammatory cytokines, impairing the tumor's ability to build a blood supply and manipulate its microenvironment.[2]
  • Receptor Internalization: Binding of panitumumab to EGFR also leads to the internalization and subsequent degradation of the receptor, effectively removing it from the cell surface and further diminishing the cell's signaling capacity.[2]

A crucial distinction between panitumumab and the other major anti-EGFR antibody, cetuximab, lies in their immunoglobulin isotype. Panitumumab is a fully human IgG2 antibody, while cetuximab is a chimeric (mouse-human) IgG1 antibody.[21] This structural difference has direct clinical consequences. The Fc region of IgG1 antibodies can engage with immune effector cells (like Natural Killer cells) to mediate antibody-dependent cellular cytotoxicity (ADCC) and can activate the complement pathway.[21] In contrast, the IgG2 isotype of panitumumab is considered largely immunologically inert and does not significantly elicit these immune-mediated functions.[21] This difference in molecular design not only impacts the potential mechanisms of tumor cell killing but also contributes to a more favorable safety profile regarding infusion reactions. Because panitumumab is fully human and lacks the murine component found in cetuximab, it is far less likely to be recognized as foreign by the patient's immune system. This lower immunogenicity translates directly to a significantly lower incidence of hypersensitivity and infusion-related reactions, a key clinical advantage that allows panitumumab to be administered without the need for routine premedication with antihistamines.[25]

Pharmacokinetics: A Target-Mediated Profile

The pharmacokinetics (PK) of panitumumab, which describe its absorption, distribution, metabolism, and excretion, are characteristic of a monoclonal antibody with high target affinity. It exhibits what is known as target-mediated drug disposition, meaning its clearance from the body is partly dependent on its binding to the EGFR target.[2]

At doses lower than 2 mg/kg, this target-mediated clearance pathway becomes saturated, leading to non-linear pharmacokinetics where clearance decreases as the dose increases.[17] However, at the clinically recommended dose of 6 mg/kg administered every 14 days, the EGFR binding sites are saturated, and the drug's pharmacokinetics become approximately linear and dose-proportional.[17]

  • Distribution: Following intravenous administration, panitumumab has a relatively small volume of distribution, primarily confined to the vascular and interstitial spaces. The volume of the central compartment is approximately 0.042 L/kg (or 42 mL/kg).[18] With the bi-weekly dosing schedule, panitumumab concentrations reach a steady state after the third infusion.[17]
  • Elimination and Half-Life: The elimination of panitumumab is governed by two main pathways: a non-specific, linear clearance pathway through the reticuloendothelial system, and the saturable, target-mediated pathway involving binding to EGFR, followed by internalization and intracellular degradation.[18] The terminal elimination half-life is approximately 7.5 days, with clinical studies reporting a range of 4 to 11 days.[1]
  • Clearance: Systemic clearance of panitumumab is low, with a typical value of approximately 4.9 mL/kg/day.[1]
  • Special Populations: Clinical studies have shown that the pharmacokinetics of panitumumab are not meaningfully affected by patient age, gender, race, or the level of EGFR membrane expression on tumor cells.[17] Formal pharmacokinetic studies in patients with renal or hepatic impairment have not been conducted.[27] However, a case report of a patient with Child-Pugh class B liver dysfunction treated with panitumumab found that the PK parameters (AUC, half-life, and clearance) were within the range of historical controls with normal liver function, suggesting that dose adjustments may not be required in this population.[35]

The Central Role of RAS Status and Predictive Biomarkers

The clinical utility of panitumumab is inextricably linked to the molecular genetics of the patient's tumor. The evolution of its indication from a broad label to a highly specific, biomarker-defined population serves as a paradigm for the development of precision oncology. This progression reflects the field's growing understanding that targeting a pathway is only effective if the pathway is not constitutively activated downstream of the point of inhibition.

RAS Wild-Type as a Prerequisite for Efficacy

The single most important predictive biomarker for panitumumab therapy is the mutation status of the RAS family of genes (KRAS and NRAS). It is now unequivocally established that the efficacy of panitumumab is strictly limited to patients whose tumors are RAS wild-type (WT), meaning they do not harbor activating mutations in these genes.[5]

The rationale for this is rooted in the biology of the EGFR signaling cascade. In RAS-WT tumors, the pathway operates in a linear fashion, where downstream signaling is dependent on upstream activation of EGFR by its ligands. Therefore, blocking EGFR with panitumumab effectively shuts down the entire cascade.[1]

Reflecting this critical dependency, regulatory agencies worldwide, including the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA), have made RAS mutation testing mandatory before initiating treatment with Vectibix®.[6] This testing must be performed by an experienced laboratory using a validated, FDA-approved companion diagnostic test. The comprehensive analysis must confirm the absence of activating somatic mutations in all clinically relevant exons of both genes:

  • KRAS: Exon 2 (codons 12, 13), Exon 3 (codons 59, 61), and Exon 4 (codons 117, 146).[5]
  • NRAS: Exon 2 (codons 12, 13), Exon 3 (codons 59, 61), and Exon 4 (codons 117, 146).[2]

Detrimental Outcomes in RAS-Mutant Colorectal Cancer

Panitumumab is explicitly not indicated for the treatment of patients with RAS-mutant (MT) mCRC or for patients whose RAS mutation status is unknown.[36] This is a formal Limitation of Use in the U.S. and a contraindication in combination with oxaliplatin-based chemotherapy in Europe.[19]

The presence of an activating mutation in a RAS gene creates a protein that is "stuck" in the "on" position, continuously signaling through the RAF-MEK-ERK pathway regardless of the status of the upstream EGFR.[1] In this scenario, blocking EGFR with panitumumab is futile, as the downstream pathway has been hijacked and rendered independent of EGFR regulation. This molecular bypass mechanism explains the complete lack of response to panitumumab observed in patients with

RAS-MT tumors.[5]

Crucially, the data indicate more than just a lack of benefit; they point to the potential for significant harm. Retrospective and exploratory analyses of randomized clinical trials, such as the PRIME study, have shown that patients with RAS-MT mCRC who received panitumumab in combination with FOLFOX chemotherapy experienced worse outcomes, including a shorter progression-free survival and a trend toward decreased overall survival, compared to patients who received FOLFOX alone.[5] This finding underscores the absolute necessity of accurate biomarker testing to avoid exposing patients to the toxicities of an ineffective and potentially detrimental therapy.

Emerging and Secondary Biomarkers

Beyond the primary RAS WT/MT dichotomy, the therapeutic landscape for panitumumab is further refined by a number of secondary and emerging biomarkers that influence treatment decisions and strategy.

Primary Tumor Location

The anatomical location of the primary tumor has emerged as a powerful predictive biomarker. Data from several trials, most prospectively and definitively from the Japanese PARADIGM trial, have shown that the benefit of first-line anti-EGFR therapy is largely confined to patients whose primary tumors arise in the left side of the colon (descending colon, sigmoid colon, and rectum).[11] In this population, panitumumab plus FOLFOX was shown to be superior to bevacizumab plus FOLFOX for improving overall survival.[11] Conversely, patients with right-sided tumors (cecum, ascending colon, transverse colon) appear to derive less benefit from anti-EGFR therapy and may be better served by an anti-VEGF agent like bevacizumab in the first-line setting. This finding has been incorporated into major clinical practice guidelines.[5]

BRAF V600E Mutation

The BRAF gene encodes a protein kinase that acts downstream of RAS. A specific activating mutation, V600E, is found in a subset of mCRC patients (typically those who are RAS-WT) and is associated with a particularly aggressive disease course and poor prognosis.[14] While these tumors are

RAS-WT, their downstream pathway is activated by the mutant BRAF protein. In this specific molecular subtype, anti-EGFR therapy with panitumumab is not used as a single agent but is recommended as part of a combination regimen with a targeted BRAF inhibitor, such as encorafenib, to achieve dual pathway blockade.[36]

KRAS G12C Mutation: A New Paradigm

In a significant evolution of its use, panitumumab gained a new indication in a RAS-mutant population. The FDA approved panitumumab in combination with sotorasib, a specific inhibitor of the KRAS G12C mutant protein, for patients with KRAS G12C-mutated mCRC who have progressed on prior chemotherapy.[13] This indication represents a sophisticated understanding of tumor biology. While sotorasib directly inhibits the mutant KRAS G12C protein, tumor cells can develop resistance by reactivating the upstream EGFR pathway in a feedback loop. The addition of panitumumab serves to block this escape mechanism. This "vertical inhibition" strategy, targeting the pathway at two different levels, was validated in the CodeBreaK 300 trial, which showed superior PFS for the combination versus standard of care.[13] This is the only approved indication for panitumumab in a

RAS-mutant setting and highlights its potential role in overcoming acquired resistance to other targeted agents.

EGFR Ectodomain Mutations

For patients on anti-EGFR therapy, a common mechanism of acquired resistance is the development of new mutations in the EGFR gene itself, specifically in the extracellular domain where the antibodies bind. One such mutation, S492R, has been shown to confer clinical resistance to cetuximab. However, structural and biophysical studies have revealed that panitumumab can still bind effectively to EGFR harboring the S492R mutation due to subtle differences in its binding interface, which features a central cavity that can accommodate the mutated residue.[30] This provides a molecular rationale for why a patient who develops resistance to cetuximab via an S492R mutation might remain sensitive to panitumumab, highlighting that the two antibodies are not perfectly interchangeable.

Clinical Evidence in Metastatic Colorectal Cancer (mCRC)

The clinical development program for panitumumab has been extensive, involving numerous large-scale, randomized Phase 3 trials that have meticulously defined its role across different lines of therapy and in comparison to other standard-of-care agents. This body of evidence provides a robust foundation for its use in specific, biomarker-defined patient populations.

First-Line Treatment in Combination with Chemotherapy

The cornerstone trial establishing panitumumab in the first-line setting is the PRIME (Panitumumab Randomized trial In combination with chemotherapy for Metastatic colorectal cancer to determine Efficacy) study (NCT00364013).[45] This was a pivotal, global, open-label Phase 3 trial that randomized 1,183 patients with previously untreated mCRC to receive either panitumumab plus the FOLFOX4 chemotherapy regimen (oxaliplatin, fluorouracil, and leucovorin) or FOLFOX4 alone.[7] The primary endpoint was progression-free survival (PFS) in the population of patients with wild-type (

WT) KRAS exon 2 tumors.[48]

The results from the PRIME trial were practice-defining:

  • Efficacy in WT KRAS Population: In the prospectively defined WT KRAS patient group, the addition of panitumumab to FOLFOX4 led to a statistically significant and clinically meaningful improvement in PFS. The median PFS was 10.0 months in the panitumumab-FOLFOX4 arm compared to 8.6 months in the FOLFOX4-alone arm (Hazard Ratio = 0.80; 95% Confidence Interval [CI], 0.67–0.95; p=0.01).[8] Furthermore, an updated, exploratory analysis with more mature data demonstrated a significant improvement in overall survival (OS), with a median OS of 23.8 months for the combination versus 19.4 months for chemotherapy alone (HR = 0.83; 95% CI, 0.70–0.98; p=0.03).[7] The objective response rate (ORR) was also higher in the panitumumab arm (55% vs. 48%).[7]
  • Harm in MT KRAS Population: In stark contrast, the trial confirmed the lack of benefit and potential for harm in patients with mutant (MT) KRAS tumors. In this subgroup, patients treated with panitumumab plus FOLFOX4 had a shorter PFS compared to those receiving FOLFOX4 alone (HR = 1.27).[45]

The PRIME study was instrumental in securing the full FDA approval for panitumumab in the first-line setting for WT KRAS mCRC and unequivocally established the necessity of biomarker testing to guide treatment selection.[7]

Second-Line and Monotherapy in Chemorefractory Disease

Panitumumab's utility has also been proven in patients whose disease has progressed on initial therapies. A large Phase 3 trial (NCT00339183) evaluated panitumumab in the second-line setting. This study randomized 1,186 patients who had progressed after first-line fluoropyrimidine-based chemotherapy to receive either panitumumab in combination with the FOLFIRI regimen (irinotecan, fluorouracil, leucovorin) or FOLFIRI alone.[37] In the

WT KRAS subgroup, the panitumumab-FOLFIRI combination significantly improved PFS, with a median of 5.9 months versus 3.9 months for FOLFIRI alone (p=0.004). A positive but not statistically significant trend toward improved OS was also observed (14.5 vs. 12.5 months).[37]

In the chemorefractory setting (after progression on all standard chemotherapies), panitumumab's efficacy as a monotherapy was established in an early Phase 3 trial. This study compared panitumumab plus Best Supportive Care (BSC) to BSC alone in 463 heavily pretreated patients.[2] The results in the retrospectively analyzed

WT KRAS population were striking:

  • Panitumumab monotherapy led to a significant improvement in PFS, with a median of 12.3 weeks versus 7.3 weeks for BSC alone (HR = 0.45; p<0.0001).[2]
  • The ORR was 17% in the WT KRAS group treated with panitumumab, compared to 0% in the MT KRAS group, further highlighting the biomarker's predictive power.[43]

These monotherapy data formed the basis for panitumumab's initial accelerated FDA approval in 2006 and its indication for patients with chemorefractory mCRC.[7]

Head-to-Head Comparator Analysis: Panitumumab versus Cetuximab

Given that both panitumumab and cetuximab are anti-EGFR antibodies used in mCRC, a direct comparison was essential. The ASPECCT (A Study of Panitumumab Efficacy and Safety Compared to Cetuximab) trial (NCT01001377) was a global, randomized, open-label, Phase 3 non-inferiority study designed for this purpose.[9] It enrolled nearly 1,000 patients with chemorefractory

WT KRAS exon 2 mCRC and randomized them to receive either panitumumab monotherapy or cetuximab monotherapy.[10]

The trial successfully met its primary endpoint, demonstrating that panitumumab was non-inferior to cetuximab for overall survival. The median OS was 10.2 months for panitumumab versus 9.9 months for cetuximab (HR = 0.94).[10] Secondary endpoints, including PFS (4.2 vs. 4.4 months) and ORR (22% vs. 19.8%), were also similar between the two arms, suggesting they are broadly equivalent options in this clinical setting.[9]

However, an important finding emerged from post-hoc subgroup analyses. In the subset of patients who had received prior treatment with the anti-VEGF antibody bevacizumab, panitumumab appeared to provide a greater benefit. A combined analysis of ASPECCT and a similar Japanese trial (WJOG6510G) showed that in this prior-bevacizumab population, panitumumab was associated with a significantly longer OS (median 12.8 vs. 10.1 months; HR = 0.72) and PFS compared to cetuximab.[32] This suggests that for patients progressing on a bevacizumab-containing regimen, panitumumab may be the preferred anti-EGFR agent.

Head-to-Head Comparator Analysis: Panitumumab versus Bevacizumab

The optimal first-line biologic partner for chemotherapy in RAS-WT mCRC was a major clinical question, leading to the PARADIGM (Panitumumab and RAS, Diagnostically-useful Gene Mutation for mCRC) trial (NCT02394795).[11] This large, randomized, Phase 3 trial conducted in Japan compared first-line mFOLFOX6 plus panitumumab (anti-EGFR) versus mFOLFOX6 plus bevacizumab (anti-VEGF) in 823 patients with

RAS-WT mCRC.[11] The study was prospectively designed to analyze the primary endpoint of OS based on the location of the primary tumor.[11]

The results of PARADIGM were practice-changing:

  • In the pre-specified analysis of patients with left-sided primary tumors, the panitumumab-containing regimen demonstrated a statistically significant and clinically superior overall survival compared to the bevacizumab-containing regimen. The median OS was 37.9 months for the panitumumab arm versus 34.3 months for the bevacizumab arm (HR = 0.82; p=0.031).[11]
  • This was the first prospective trial to definitively show the superiority of an anti-EGFR antibody over an anti-VEGF antibody in the first-line treatment of left-sided, RAS-WT mCRC, firmly establishing primary tumor sidedness as a critical factor in treatment selection.[11]

Novel Combination Therapy for KRAS G12C-Mutated mCRC

The CodeBreaK 300 trial (NCT05198934) established a new role for panitumumab. This randomized, controlled study evaluated the KRAS G12C inhibitor sotorasib, either alone or in combination with panitumumab, against standard of care (trifluridine/tipiracil or regorafenib) in patients with chemorefractory KRAS G12C-mutated mCRC.[13] The combination of sotorasib (960 mg) plus panitumumab resulted in a significantly improved PFS compared to standard of care, with a median of 5.6 months versus 2.2 months (HR = 0.49; p=0.006). The ORR was 26% for the combination versus 0% for standard of care.[13] These compelling results led to the FDA's approval of this combination in January 2024, creating a new standard of care and the first-ever indication for panitumumab in a

RAS-mutant population.[13]

Negative Trial Data: Defining Boundaries

An important part of defining a drug's role is understanding where it should not be used. A randomized clinical trial designed to evaluate panitumumab in combination with both chemotherapy and bevacizumab in the first-line setting was terminated early.[50] The interim analysis showed that the addition of panitumumab to a bevacizumab-and-chemotherapy backbone resulted in inferior PFS, increased toxicity, and a higher incidence of death compared to bevacizumab and chemotherapy alone.[40] This trial clearly established that the dual blockade of both the EGFR and VEGF pathways is not beneficial and is potentially harmful, and this combination is not recommended.[34]

Table 2: Summary of Pivotal Clinical Trials of Panitumumab in mCRC

Trial / NCT IDPhaseClinical SettingPatient Population (RAS Status)Treatment ArmsPrimary EndpointKey Efficacy Results (Median OS/PFS, HR)
PRIME (NCT00364013)31st-LineUntreated, KRAS-WTPanitumumab + FOLFOX4 vs. FOLFOX4PFSPFS: 10.0 vs 8.6 mos (HR=0.80, p=0.01) OS: 23.8 vs 19.4 mos (HR=0.83, p=0.03)
NCT0033918332nd-LinePost-1st-Line Chemo, KRAS-WTPanitumumab + FOLFIRI vs. FOLFIRIPFSPFS: 5.9 vs 3.9 mos (p=0.004) OS: 14.5 vs 12.5 mos (NS)
Monotherapy3ChemorefractoryPost-Chemo, KRAS-WTPanitumumab + BSC vs. BSCPFSPFS: 12.3 vs 7.3 wks (HR=0.45, p<0.0001)
ASPECCT (NCT01001377)3ChemorefractoryPost-Chemo, KRAS-WTPanitumumab vs. CetuximabOS (Non-inferiority)OS: 10.2 vs 9.9 mos (HR=0.94, Non-inferior)
PARADIGM (NCT02394795)31st-LineUntreated, RAS-WT, Left-SidedPanitumumab + mFOLFOX6 vs. Bevacizumab + mFOLFOX6OSOS: 37.9 vs 34.3 mos (HR=0.82, p=0.031)
CodeBreaK 300 (NCT05198934)3ChemorefractoryPost-Chemo, KRAS G12C-MutantSotorasib + Panitumumab vs. SoCPFSPFS: 5.6 vs 2.2 mos (HR=0.49, p=0.006)

BSC=Best Supportive Care; HR=Hazard Ratio; mos=months; NS=Not Significant; OS=Overall Survival; PFS=Progression-Free Survival; SoC=Standard of Care; wks=weeks

The collective evidence from these trials illustrates a clear, data-driven path for the use of panitumumab. It is not a drug for all colorectal cancer patients. Its application requires a stratified approach, where treatment decisions are guided by a sequence of biomarker assessments: first RAS status, then primary tumor location, and finally, consideration of prior therapies and other specific mutations like BRAF or KRAS G12C. This evidence base provides a precise and complex roadmap for clinicians to optimize outcomes by selecting the right treatment for the right patient at the right time.

Comprehensive Safety and Tolerability Profile

The safety profile of panitumumab is well-defined and is dominated by adverse events that are a direct and predictable consequence of its on-target inhibition of EGFR in normal tissues. The predictability of this toxicity profile allows for proactive monitoring and management strategies to mitigate severity and ensure patient safety.

Boxed Warning: Severe Dermatologic and Soft Tissue Toxicity

The most prominent and frequent adverse event associated with panitumumab is dermatologic toxicity, for which it carries a Boxed Warning in its FDA-approved labeling.[14]

  • Incidence: Skin and soft tissue toxicities are nearly universal, occurring in 90% to 96% of patients treated with panitumumab, whether as a monotherapy or in combination with chemotherapy.[1] While most reactions are Grade 1 or 2, severe (Grade 3 or 4) reactions are common, affecting 12-15% of patients on monotherapy and as many as 32% of patients receiving panitumumab with FOLFOX chemotherapy.[5] The median time to onset is approximately 10 to 12 days after the first dose.[15]
  • Manifestations: The clinical presentation is characteristic of EGFR inhibitor-induced rash and includes acneiform dermatitis (pustular rash resembling acne), pruritus (itching), erythema (redness), generalized rash, skin exfoliation (peeling), skin fissures (cracks), and paronychia (inflammation and infection of the nail folds).[5]
  • Complications: Severe dermatologic reactions are not merely cosmetic; they can lead to serious and life-threatening infectious sequelae. Reported complications include cellulitis, abscesses requiring drainage, necrotizing fasciitis, and sepsis, which have in rare cases been fatal.[1] Additionally, rare cases of severe bullous mucocutaneous diseases, such as Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN), have been observed in post-marketing reports.[5]
  • Correlation with Efficacy: An intriguing and consistent observation across multiple clinical trials is that patients who develop more severe skin toxicities tend to have better clinical outcomes, including longer progression-free and overall survival.[10] This suggests that the rash serves as a pharmacodynamic marker, indicating that the drug is effectively engaging and inhibiting its EGFR target in both tumor and normal tissues.

Infusion-Related Reactions (IRRs)

As a parenterally administered monoclonal antibody, panitumumab carries a risk of infusion-related reactions (IRRs). However, due to its fully human structure, the incidence is relatively low compared to chimeric antibodies.

  • Incidence and Symptoms: IRRs occur in approximately 1-4% of patients, with severe (Grade 3-4) reactions reported in about 1%.[17] Symptoms typically occur within 24 hours of an infusion and may include fever, chills, dyspnea (shortness of breath), bronchospasm, and hypotension.[17]
  • Severe and Late-Onset Reactions: Although rare, fatal infusion reactions have been reported in post-marketing surveillance.[22] Patients must also be counseled about the possibility of late-onset hypersensitivity reactions, which can occur more than 24 hours after an infusion. A fatal case of angioedema has been reported in this context.[18]

Electrolyte Depletion: Hypomagnesemia and Hypocalcemia

A frequent and clinically significant metabolic complication of panitumumab therapy is electrolyte depletion, particularly hypomagnesemia.

  • Incidence and Severity: Panitumumab often causes a progressive decrease in serum magnesium levels, which can become severe (Grade 3-4).[4] Hypomagnesemia of any grade is reported in 38-51% of patients, with severe cases occurring in 1-6%.[5] When combined with sotorasib, the incidence rises to 69%, with 16% being severe.[5] Severe hypomagnesemia can lead to secondary hypocalcemia and hypokalemia.[17]
  • Mechanism and Monitoring: The mechanism is believed to involve on-target EGFR inhibition in the kidneys and gastrointestinal tract, impairing magnesium reabsorption and absorption.[4] Due to its frequency and potential severity, it is mandatory to monitor serum electrolytes—especially magnesium, calcium, and potassium—before initiating therapy, periodically during treatment, and for up to 8 weeks after the final dose. Electrolyte repletion, often requiring intravenous supplementation, should be administered as appropriate.[16]

Other Significant Toxicities

  • Ocular Toxicity: Serious ocular adverse events have been reported, including keratitis (inflammation of the cornea), ulcerative keratitis, and, in rare cases, corneal perforation. Patients should be monitored for signs of ocular toxicity like eye pain, redness, or vision changes, and treatment should be interrupted or discontinued for acute or worsening keratitis.[1]
  • Pulmonary Toxicity: Although rare (<1%), panitumumab has been associated with cases of interstitial lung disease (ILD) and pulmonary fibrosis, which can be fatal. Panitumumab is contraindicated in patients with a history of these conditions. Any patient developing acute or worsening pulmonary symptoms (e.g., dyspnea, cough) on treatment should be evaluated for ILD, and the drug should be permanently discontinued if it is diagnosed.[18]
  • Gastrointestinal Toxicity: Severe diarrhea is a known side effect that can lead to dehydration and subsequent acute renal failure and other complications. The risk of severe diarrhea is particularly elevated when panitumumab is combined with irinotecan-based chemotherapy regimens.[26]
  • Photosensitivity: As EGFR is crucial for skin health, its inhibition can lead to increased sensitivity to sunlight. Sunlight exposure can exacerbate the dermatologic toxicities associated with panitumumab. Patients must be advised to limit sun exposure and consistently use broad-spectrum sunscreen (SPF >15) and protective clothing (e.g., hats) during treatment.[18]

Management of Adverse Events and Dose Modifications

The predictable nature of panitumumab's toxicities allows for standardized management protocols, including proactive measures and clear guidelines for dose modification.

Table 3: Incidence of Key Adverse Reactions with Panitumumab (%)

Adverse ReactionMonotherapy (n=229)With FOLFOX (n=585)With Sotorasib (n=126)
Dermatologic Toxicity (All Grades)90%96%94%
Severe (G3-4) Dermatologic Toxicity15%32% (G3), 1% (G4)16% (G3)
Hypomagnesemia (All Grades)38%51%69%
Severe (G3-4) Hypomagnesemia3.9% (G3/4)11% (G3/4)16.4% (G3/4)
Severe (G3-4) Infusion Reaction~1%~1%Not specified
Diarrhea (All Grades)24%66% (with FOLFOX)51% (with sotorasib)

Data compiled from [5]

Proactive management of dermatologic toxicity is recommended, including the use of moisturizers, sunscreens, topical steroid creams (e.g., 1% hydrocortisone), and, in some cases, prophylactic oral antibiotics like doxycycline.[19] For toxicities that do arise, clear dose modification rules are in place.

Table 4: Guidelines for Dose Modification Due to Dermatologic Toxicity

EventRequired Action
1st Occurrence of Grade 3 ReactionWithhold 1-2 doses. If reaction improves to ≤ Grade 2, reinitiate at 100% of the original dose.
2nd Occurrence of Grade 3 ReactionWithhold 1-2 doses. If reaction improves to ≤ Grade 2, reinitiate at 80% of the original dose.
3rd Occurrence of Grade 3 ReactionWithhold 1-2 doses. If reaction improves to ≤ Grade 2, reinitiate at 60% of the original dose.
4th Occurrence of Grade 3 ReactionPermanently discontinue Vectibix®.
Any Grade 4 ReactionPermanently discontinue Vectibix®.
Grade 3 Reaction that does not recoverPermanently discontinue Vectibix® after withholding 1-2 doses.

Guidelines based on [55]

For infusion reactions, a mild or moderate (Grade 1-2) reaction requires a 50% reduction in the infusion rate for the duration of that infusion. A severe (Grade 3-4) reaction necessitates immediate and permanent termination of the therapy.[22]

Dosage, Administration, and Practical Considerations

The administration of panitumumab requires adherence to specific protocols for dosing, preparation, and infusion to ensure patient safety and therapeutic efficacy. Treatment should be supervised by a physician experienced in the use of anticancer therapy.[19]

Recommended Dosing Regimens

The standard, FDA-approved dose of panitumumab for all its indications in mCRC is 6 mg/kg of body weight, administered as an intravenous (IV) infusion once every 14 days.[19] This bi-weekly schedule is maintained until disease progression or the development of unacceptable toxicity.[40]

This dosing regimen applies to its use as:

  • Monotherapy in the chemorefractory setting.[40]
  • First-line therapy in combination with FOLFOX or FOLFIRI.[19]
  • Combination therapy with sotorasib for KRAS G12C-mutated mCRC.[40]

Unlike chimeric antibodies, no loading dose is required for panitumumab.[25] Furthermore, routine premedication with antihistamines (e.g., diphenhydramine) is not required, although appropriate medical resources for the treatment of severe infusion reactions must be readily available during administration.[25]

Preparation and Intravenous Administration Protocol

Aseptic technique must be used throughout the preparation process to maintain sterility.[25]

Preparation

  1. Inspect Vial: Before use, parenteral drug products should be visually inspected. The panitumumab solution should be colorless. It may contain a small quantity of visible, translucent-to-white, amorphous proteinaceous particles, which is acceptable. The vial should not be shaken. Do not use if the solution is discolored.[25]
  2. Withdraw Drug: The necessary amount of panitumumab to achieve a dose of 6 mg/kg should be withdrawn from the single-use vial(s). It is critical to use a 21-gauge or larger (i.e., smaller bore) hypodermic needle for this step. Needle-free devices, such as vial adapters, must not be used to withdraw the vial contents.[25]
  3. Dilute Solution: The withdrawn panitumumab must be diluted in 0.9% Sodium Chloride Injection, USP. The final volume of the infusion bag depends on the total dose:
  • For doses ≤ 1,000 mg, dilute to a total volume of 100 mL.
  • For doses > 1,000 mg, dilute to a total volume of 150 mL. The final concentration of panitumumab in the infusion solution must not exceed 10 mg/mL.19
  1. Mix Solution: The diluted solution should be mixed by gentle inversion. Do not shake the bag, as this can cause protein aggregation and denaturation.[25] Any unused portion of the drug remaining in the vial must be discarded.[25]

Administration

  1. Infusion Setup: Panitumumab must be administered via an infusion pump. It must not be given as an intravenous push or bolus. The infusion line must include a low-protein-binding 0.2 µm or 0.22 µm in-line filter to remove any proteinaceous particles.[19]
  2. IV Line Management: To prevent mixing with other drugs or solutions, the IV line should be flushed with 0.9% sodium chloride before and after the panitumumab infusion. Panitumumab should not be mixed with or administered in the same line as other medicinal products.[19]
  3. Infusion Times: The duration of the infusion is based on the total dose and the patient's tolerance:
  • Initial Infusion (all doses ≤ 1,000 mg): The first dose should be infused over 60 minutes.[25]
  • Subsequent Infusions (doses ≤ 1,000 mg): If the first infusion is well-tolerated, subsequent infusions may be administered over a shorter period of 30 to 60 minutes.[19]
  • All Doses > 1,000 mg: Any dose exceeding 1,000 mg should be infused over 90 minutes.[25]

Stability and Storage

Unopened vials of Vectibix® must be stored under refrigeration at 2°C to 8°C (36°F to 46°F) in the original carton to protect from light. Vials must not be frozen.[55] The diluted infusion solution is stable for up to 6 hours if stored at room temperature, or for up to 24 hours if stored under refrigeration (2°C to 8°C). The diluted solution should not be frozen.[25]

The Evolving Therapeutic Landscape

Panitumumab does not exist in a therapeutic vacuum. Its clinical value and strategic positioning are best understood in comparison to other targeted agents, particularly the other anti-EGFR antibody cetuximab, and in the context of a rapidly advancing field of research that continues to refine its use and explore new applications.

Comparative Assessment: Panitumumab vs. Cetuximab

While both panitumumab and cetuximab target EGFR and are often considered in the same clinical contexts, they are distinct molecular entities with differences that are clinically relevant. They should be considered similar but not interchangeable, with specific scenarios potentially favoring one agent over the other.

This nuanced view is supported by a direct comparison of their properties. The ASPECCT trial established their broad equivalence in the overall chemorefractory population based on the primary endpoint of non-inferiority for OS.[9] However, key differences in structure, safety, and activity in specific subgroups allow for more personalized treatment decisions. For example, panitumumab's fully human structure confers a clear safety advantage regarding infusion reactions, a practical benefit that simplifies administration and improves the patient experience.[32] Furthermore, the accumulating evidence suggesting superior efficacy for panitumumab in patients with prior bevacizumab exposure provides a data-driven rationale for preferring it in this common clinical scenario.[32] Finally, the differential sensitivity to acquired EGFR resistance mutations like S492R underscores that they are not biologically identical, and resistance to one may not automatically imply resistance to the other.[30]

Table 5: Comparative Profile of Panitumumab and Cetuximab

FeaturePanitumumab (Vectibix®)Cetuximab (Erbitux®)Source(s)
Antibody Class/IsotypeFully Human IgG2Chimeric (Mouse-Human) IgG121
Binding Affinity (KD​)High (~0.05 nM)Moderate (~0.39 nM)31
Immune Effector Function (ADCC)Minimal to noneYes21
Dosing Schedule6 mg/kg every 2 weeks400 mg/m² load, then 250 mg/m² weekly (or 500 mg/m² q2w)33
Premedication RequiredNo (routine)Yes (H1 antagonist)33
Head-to-Head OS (ASPECCT)Non-inferior (Median 10.2 mos)Non-inferior (Median 9.9 mos)10
OS after Prior BevacizumabMay be superior (Median OS 12.8 mos)(Median OS 10.1 mos)32
Rate of Severe (G3/4) Infusion Reactions~1%~3% (US label); higher in some reports (~9%)32
Resistance to S492R MutationNo (remains effective)Yes30

Current and Future Clinical Investigations

The clinical development of panitumumab continues, with research focused on optimizing its use, overcoming resistance, and expanding its application into new combinations and contexts. A review of the ClinicalTrials.gov database reveals a dynamic landscape of ongoing investigation.[58]

  • Combination with Immunotherapy: A key area of interest is combining anti-EGFR therapy with immune checkpoint inhibitors to treat microsatellite stable (MSS) mCRC, which is typically unresponsive to immunotherapy alone. The rationale is that EGFR inhibition may promote T-cell infiltration and alter the tumor microenvironment, sensitizing these "cold" tumors. A Phase II trial combining panitumumab with ipilimumab (anti-CTLA-4) and nivolumab (anti-PD-1) in refractory RAS-WT, MSS mCRC showed a promising ORR of 32.1% and durable responses in some patients, suggesting this is a viable strategy worthy of further study.[61]
  • Overcoming Acquired Resistance: Research is actively exploring ways to treat patients who develop resistance to panitumumab. Since a common resistance mechanism is the acquisition of RAS mutations, trials are evaluating the combination of panitumumab with inhibitors of the downstream pathway. For example, the NCT03087071 trial is studying panitumumab with or without the MEK inhibitor trametinib in patients who have progressed on prior anti-EGFR therapy, with treatment allocated based on the patient's specific molecular mechanism of resistance identified in circulating tumor DNA.[62]
  • Exploration in Other Cancers: While panitumumab's success has been primarily in mCRC, its activity has been explored in other EGFR-expressing tumors. Early-phase trials have investigated its use in head and neck squamous cell carcinoma, urothelial carcinoma, and cholangiocarcinoma, although with more limited efficacy demonstrated to date compared to its role in colorectal cancer.[26]
  • Molecular Imaging Applications: A novel and promising application for panitumumab is its use as a targeting agent for molecular imaging. By conjugating panitumumab to a radioisotope (e.g., Zirconium-89 for PET scans, Indium-111 for SPECT) or a near-infrared fluorescent dye (e.g., IRDye800), researchers are developing agents that can specifically identify EGFR-expressing tumor cells in vivo. Ongoing trials are evaluating these conjugates for improved diagnosis, nodal staging, and surgical guidance in cancers of the head and neck and pancreas.[57]

Concluding Remarks and Strategic Outlook

Panitumumab (Vectibix®) has firmly established itself as a critical agent in the management of metastatic colorectal cancer. Its journey from a broadly targeted antibody to a highly specific, biomarker-driven therapy exemplifies the trajectory of modern precision oncology. The wealth of clinical evidence underscores a fundamental principle: the profound efficacy of panitumumab is unlocked only through meticulous patient selection. Its use is predicated on the mandatory confirmation of wild-type RAS status, a requirement born from definitive data showing not only a lack of benefit but also potential harm in RAS-mutant tumors.

The therapeutic algorithm for panitumumab is now highly refined. The PARADIGM trial has provided Level 1 evidence for its superiority over bevacizumab-based therapy in the first-line treatment of RAS-wild-type, left-sided mCRC, making it a standard of care in this large patient population. In the chemorefractory setting, the ASPECCT trial has demonstrated its non-inferiority to cetuximab, providing a valuable therapeutic choice with a distinct safety and administration profile, while subgroup analyses suggest it may be the preferred anti-EGFR agent following progression on bevacizumab.

The strategic outlook for panitumumab is one of continued refinement and novel application. The recent approval in combination with sotorasib for KRAS G12C-mutated cancer signals a paradigm shift, repositioning panitumumab from a therapy targeting a primary oncogenic pathway to a crucial partner agent used to block adaptive resistance to other targeted drugs. Future growth will likely be driven by its integration into even more sophisticated, biomarker-guided combination strategies, particularly with immune checkpoint inhibitors in MSS tumors and with other targeted agents to overcome acquired resistance. As our understanding of tumor biology deepens, panitumumab is poised to remain a vital and evolving tool in the oncologist's armamentarium, delivering significant clinical benefit to carefully selected patients with metastatic colorectal cancer.

Works cited

  1. Panitumumab: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed July 17, 2025, https://go.drugbank.com/drugs/DB01269
  2. Treatment of metastatic colorectal cancer: focus on panitumumab - Dove Medical Press, accessed July 17, 2025, https://www.dovepress.com/treatment-of-metastatic-colorectal-cancer-focus-on-panitumumab-peer-reviewed-fulltext-article-CMAR
  3. What is the mechanism of Panitumumab? - Patsnap Synapse, accessed July 17, 2025, https://synapse.patsnap.com/article/what-is-the-mechanism-of-panitumumab
  4. Panitumumab - PMC, accessed July 17, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2780272/
  5. Targeted Treatment for WT RAS mCRC | Vectibix® (panitumumab), accessed July 17, 2025, https://www.vectibixhcp.com/targeted-1l-treatment
  6. Annotation of FDA Label for panitumumab and KRAS, NRAS - PharmGKB, accessed July 17, 2025, https://www.pharmgkb.org/labelAnnotation/PA166104809
  7. Panitumumab Gains FDA Approval for KRAS Wild-Type mCRC - Targeted Oncology, accessed July 17, 2025, https://www.targetedonc.com/view/panitumumab-gains-fda-approval-for-kras-wild-type-mcrc
  8. Final results from PRIME: randomized phase III study of panitumumab with FOLFOX4 for first-line treatment of metastatic colorectal cancer - PubMed, accessed July 17, 2025, https://pubmed.ncbi.nlm.nih.gov/24718886/
  9. Phase 3 Trial Comparing Vectibix® (Panitumumab) to Erbitux® (Cetuximab) Meets Primary Endpoint Of Non-Inferiority Of Overall Survival - Amgen, accessed July 17, 2025, https://www.amgen.com/newsroom/press-releases/2013/09/phase-3-trial-comparing-vectibix-panitumumab-to-erbitux-cetuximab-meets-primary-endpoint-of-noninferiority-of-overall-survival
  10. Final results and outcomes by prior bevacizumab exposure, skin toxicity, and hypomagnesaemia from ASPECCT: randomized phase 3 non-inferiority study of panitumumab versus cetuximab in chemorefractory wild-type KRAS exon 2 metastatic colorectal cancer - PubMed, accessed July 17, 2025, https://pubmed.ncbi.nlm.nih.gov/27716478/
  11. Data from the PARADIGM Trial in Chemotherapy-naive Japanese Patients with Unresectable Advanced Recurrent Colorectal Cancer and Wild-type RAS Gene Presented at the ASCO 2022 Plenary Session - Takeda Oncology, accessed July 17, 2025, https://www.takedaoncology.com/news/news-releases/Data%20from%20the%20PARADIGM%20Trial%20in%20Chemotherapy-naive%20Japanese%20Patients%20with%20Unresectable%20Advanced%20Recurrent%20Colorectal%20Cancer%20and%20Wild-type%20RAS%20Gene%20Presented%20at%20the%20ASCO%202022%20Plenary%20Session/
  12. Panitumumab and RAS, Diagnostically-useful Gene Mutation for mCRC - Cancer Trial Results, accessed July 17, 2025, https://clin.larvol.com/trial-detail/NCT02394795
  13. FDA approves sotorasib with panitumumab for KRAS G12C-mutated colorectal cancer, accessed July 17, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-sotorasib-panitumumab-kras-g12c-mutated-colorectal-cancer
  14. Panitumumab (Vectibix) Criteria for Use - Ascension Personalized Care, accessed July 17, 2025, https://www.ascensionpersonalizedcare.com/-/media/project/aca/aca/pharmacy/pa-required/panitumumab-vectibix-criteria-for-use.pdf
  15. Considerations for Dermatologic Toxicities | Vectibix® (panitumumab), accessed July 17, 2025, https://www.vectibixhcp.com/dermatologic-toxicities
  16. Safety & Adverse Events| LUMAKRAS® (sotorasib) + Vectibix® (panitumumab), accessed July 17, 2025, https://www.lumakrashcp.com/mcrc/safety
  17. PANitumumab | Cancer Care Ontario, accessed July 17, 2025, https://www.cancercareontario.ca/en/system/files_force/panitumumab.pdf?download=1
  18. DRUG NAME: Panitumumab - BC Cancer, accessed July 17, 2025, http://www.bccancer.bc.ca/drug-database-site/Drug%20Index/Panitumumab%20monograph.pdf
  19. Vectibix, INN-panitumumab - European Medicines Agency, accessed July 17, 2025, https://www.ema.europa.eu/en/documents/product-information/vectibix-epar-product-information_en.pdf
  20. Vectibix 20 mg/mL concentrate for solution for infusion - Summary of Product Characteristics (SmPC) - (emc) | 6178, accessed July 17, 2025, https://www.medicines.org.uk/emc/product/6178/smpc
  21. Distinguishing Features of Cetuximab and Panitumumab in Colorectal Cancer and Other Solid Tumors - Frontiers, accessed July 17, 2025, https://www.frontiersin.org/journals/oncology/articles/10.3389/fonc.2019.00849/full
  22. Panitumumab Monograph for Professionals - Drugs.com, accessed July 17, 2025, https://www.drugs.com/monograph/panitumumab.html
  23. Amgen | A Worldwide Pioneer in Biotechnology, accessed July 17, 2025, https://www.amgen.com/
  24. Panitumumab | CAS NO.:339177-26-3 - GlpBio, accessed July 17, 2025, https://www.glpbio.com/research-area/panitumumab.html
  25. Dosing & Administration | Vectibix® (panitumumab), accessed July 17, 2025, https://www.vectibixhcp.com/dosing-administration
  26. Panitumumab - Wikipedia, accessed July 17, 2025, https://en.wikipedia.org/wiki/Panitumumab
  27. Panitumumab (Vectibix) - PMC, accessed July 17, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8013163/
  28. Panitumumab (ABX-EGF) | Anti-EGFR Monoclonal Antibody | MedChemExpress, accessed July 17, 2025, https://www.medchemexpress.com/panitumumab.html
  29. Panitumumab (Vectibix) - Medical Clinical Policy Bulletins - Aetna, accessed July 17, 2025, https://www.aetna.com/cpb/medical/data/700_799/0748.html
  30. The Panitumumab EGFR Complex Reveals a Binding Mechanism That Overcomes Cetuximab Induced Resistance - Our journal portfolio - PLOS, accessed July 17, 2025, https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0163366
  31. Distinguishing Features of Cetuximab and Panitumumab in Colorectal, accessed July 17, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6763619/
  32. Panitumumab versus cetuximab in patients with wild-type KRAS exon 2 metastatic colorectal cancer who received prior bevacizumab therapy: A combined analysis of individual patient data from ASPECCT and WJOG6510G. - ASCO, accessed July 17, 2025, https://www.asco.org/abstracts-presentations/ABSTRACT202515
  33. ASPECCT Study Design & Clinical Data | Vectibix® (panitumumab), accessed July 17, 2025, https://www.vectibixhcp.com/efficacy/aspecct-clinical-data
  34. name of the medicinal product - Vectibix, INN-panitumumab, accessed July 17, 2025, https://ec.europa.eu/health/documents/community-register/2010/2010042978468/anx_78468_en.pdf
  35. Pharmacokinetics (PK) of panitumumab in a single patient with metastatic colorectal cancer (mCRC) and liver dysfunction. - ASCO, accessed July 17, 2025, https://www.asco.org/abstracts-presentations/ABSTRACT105080
  36. Panitumumab (Vectibix®), accessed July 17, 2025, https://www.bcbst.com/mpmanual/!SSL!/WebHelp/Panitumumab.htm
  37. Metastatic colorectal cancer: treatment with panitumumab - SciELO, accessed July 17, 2025, https://www.scielo.br/j/ramb/a/bG9xBn4QKDnLx4TP4JHzXtg/
  38. FDA Approves Vectibix® (Panitumumab) For Use In Wild-Type RAS Metastatic Colorectal Cancer - Amgen, accessed July 17, 2025, https://www.amgen.com/newsroom/press-releases/2017/06/fda-approves-vectibix-panitumumab-for-use-in-wildtype-ras-metastatic-colorectal-cancer
  39. Vectibix | European Medicines Agency (EMA), accessed July 17, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/vectibix
  40. Vectibix (panitumumab) dosing, indications, interactions, adverse effects, and more, accessed July 17, 2025, https://reference.medscape.com/drug/vectibix-panitumumab-342261
  41. VECTIBIX (panitumumab) Label - accessdata.fda.gov, accessed July 17, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/125147s210lbl.pdf
  42. Study Details | Panitumumab and RAS, Diagnostically-useful Gene Mutation for mCRC | ClinicalTrials.gov, accessed July 17, 2025, https://clinicaltrials.gov/study/NCT02394795
  43. Wild-Type KRAS Is Required for Panitumumab Efficacy in Patients With Metastatic Colorectal Cancer | Journal of Clinical Oncology - ASCO Publications, accessed July 17, 2025, https://ascopubs.org/doi/10.1200/JCO.2007.14.7116
  44. Vectibix® (panitumumab) Administration | mCRC Treatment, accessed July 17, 2025, https://www.vectibix.com/dosing-and-administration
  45. (PDF) Final results from PRIME: Randomized phase III study of panitumumab with FOLFOX4 for first-line treatment of metastatic colorectal cancer - ResearchGate, accessed July 17, 2025, https://www.researchgate.net/publication/261520708_Final_results_from_PRIME_Randomized_phase_III_study_of_panitumumab_with_FOLFOX4_for_first-line_treatment_of_metastatic_colorectal_cancer
  46. mCRC Clinical Trial Results | Vectibix® (panitumumab), accessed July 17, 2025, https://www.vectibix.com/learn-about-vectibix-clinical-trial-results
  47. Study Details | PRIME: Panitumumab Randomized Trial In Combination With Chemotherapy for Metastatic Colorectal Cancer to Determine Efficacy | ClinicalTrials.gov, accessed July 17, 2025, https://clinicaltrials.gov/study/NCT00364013
  48. Overall survival (OS) analysis from PRIME: Randomized phase III study of panitumumab (pmab) with FOLFOX4 for first-line metastatic colorectal cancer (mCRC). - ASCO Publications, accessed July 17, 2025, https://ascopubs.org/doi/10.1200/jco.2013.31.15_suppl.3620
  49. Study Details | Comparison of Treatment Effect of Chemotherapy With Panitumumab to ... - ClinicalTrials.gov, accessed July 17, 2025, https://www.clinicaltrials.gov/study/NCT00339183
  50. FDA review of a panitumumab (Vectibix) clinical trial for first-line treatment of metastatic colorectal cancer - PubMed, accessed July 17, 2025, https://pubmed.ncbi.nlm.nih.gov/19282350/
  51. ASPECCT: A Study of Panitumumab Efficacy and Safety Compared to Cetuximab in Patients With KRAS Wild-Type Metastatic Colorectal Cancer | ClinicalTrials.gov, accessed July 17, 2025, https://clinicaltrials.gov/study/NCT01001377
  52. Efficacy of panitumumab vs cetuximab in patients with wild-type KRAS exon 2 metastatic colorectal cancer treated with prior bevacizumab: Results from ASPECCT. - ASCO, accessed July 17, 2025, https://www.asco.org/abstracts-presentations/ABSTRACT165575
  53. A multicenter, randomized, phase III study of 5-fluorouracil, leucovorin, and oxaliplatin (mFOLFOX6) plus panitumumab or bevacizumab as first-line treatment in patients with RAS (KRAS/NRAS) wild-type metastatic colorectal cancer. - ASCO, accessed July 17, 2025, https://www.asco.org/abstracts-presentations/ABSTRACT156460
  54. Amgen® SupportPlus | Vectibix® (panitumumab), accessed July 17, 2025, https://www.vectibix.com/resources-amgen-supportplus
  55. Panitumumab Dosage Guide + Max Dose, Adjustments - Drugs.com, accessed July 17, 2025, https://www.drugs.com/dosage/panitumumab.html
  56. Vectibix Dosage Guide - Drugs.com, accessed July 17, 2025, https://www.drugs.com/dosage/vectibix.html
  57. Vectibix (panitumumab) - accessdata.fda.gov, accessed July 17, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2009/125147s080lbl.pdf
  58. Search for: "Panitumumab" | Card Results - ClinicalTrials.gov, accessed July 17, 2025, https://www.clinicaltrials.gov/search?intr=%22Panitumumab%22&viewType=Table
  59. Search for: Other terms: PANITUMUMAB | Card Results | ClinicalTrials.gov, accessed July 17, 2025, https://www.clinicaltrials.gov/ct2/results?submit=Search&term=PANITUMUMAB
  60. Clinical Trials Using Panitumumab - NCI, accessed July 17, 2025, https://www.cancer.gov/research/participate/clinical-trials/intervention/panitumumab?pn=1
  61. Phase II Study of Ipilimumab, Nivolumab, and Panitumumab in Patients With KRAS/NRAS/BRAF Wild-Type Microsatellite Stable Metastatic Colorectal Cancer | JCO Oncology Advances - ASCO Publications, accessed July 17, 2025, https://ascopubs.org/doi/10.1200/OA-24-00086
  62. Panitumumab With or Without Trametinib in Treating Patients With Stage IV Colorectal Cancer | ClinicalTrials.gov, accessed July 17, 2025, https://www.clinicaltrials.gov/study/NCT03087071
  63. A Phase II Enrichment Study of Panitumumab as a Single Agent or in Combination with Trametinib in Anti-EGFR-Refractory Stage IV Colorectal Cancer Patients, accessed July 17, 2025, https://www.mdanderson.org/patients-family/diagnosis-treatment/clinical-trials/clinical-trials-index/clinical-trials-detail.ID2016-0338.html

Published at: July 17, 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.