APP下载

Mechanistic considerations for the use of monoclonal antibodies for cancer therapy

2014-09-26PatrickGlassmanJosephBalthasar

Cancer Biology & Medicine 2014年1期

Patrick M.Glassman, Joseph P.Balthasar

Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA

Introduction

The use of antibodies in oncology dates back to the work of Héricourt and Richet, who in the late 1800s, described serotherapy as a potential approach to treating malignancies1.Several decades later, Paul Ehrlich proposed the ‘magic bullet’hypothesis, which suggested that drugs could be developed that were highly selective for pathogenic cells, thereby granting the drug high potency with minimal off-site toxicity2.Despite this early work, it was not until Köhler and Milstein described hybridoma technology in 19753that development of monoclonal antibodies (mAbs) became a feasible approach in drug development.In the decades immediately after this breakthrough, coinciding with the advent of improved molecular biology techniques, it became possible for mAbs to be generated with increasing proportions of ‘human’ content.As a result,investigators are now able to produce chimeric, humanized,and fully human mAbs.Antibody platforms which incorporate human regions provide many benefits over the initially developed rodent mAbs, including the potential for reduced immunogenicity, improved effector function, and improved pharmacokinetic properties due to higher affinity interactions with the neonatal Fc receptor (FcRn) in humans.

Currently, there are 14 therapeutic mAbs approved by the FDA for use in oncology (Table 1).Of these, ten are administered as‘naked’ mAbs, two are radioimmunoconjugates (ibritumomab tiuxetan and131I tositumomab), and two are antibody-drug conjugates (ADC) (brentuximab vedotin and trastuzumab emtansine).Additionally, as of July 2013, there are 10 mAbs in late-stage (Phase II/III and Phase III) clinical trials15(Table 2).These products are utilized as major components in the therapeutic regimens for a wide variety of solid and liquid cancers.

Table 1 FDA-approved mAbs for use in oncology

Table 2 mAbs currently in late stage clinical trials

Incorporation of mAbs into chemotherapeutic regimens has led to significant improvements in patient outcomes for a variety of cancers, most notably being the addition of rituximab to the standard CHOP (cyclophosphamide, doxorubicin, vincristine,and prednisone) regimen for the treatment of large B-cell lymphoma17,18.

In this review, we describe key considerations specific to the clinical application of mAb-based therapeutics in oncology,including pharmacologic mechanisms of action, clinical targets,and pharmacokinetic intricacies.Additionally, we summarize the clinical applications of marketed mAbs and those in late stage clinical trials.

Pharmacokinetic (PK) considerations

All of the approved mAbs are members of the immune gamma globulin (IgG) family.When studied in healthy, human subjects,endogenous (or pooled) IgG antibodies are often found to demonstrate predictable, linear pharmacokinetics, with small volumes of distribution (~3-9 L), low rates of clearance (8-12 mL/h),and long biological half-lives (~20-25 d)19.However, therapeutic IgG mAbs often exhibit complex, non-linear pharmacokinetics,with substantial between- and within-patient variability.Main determinants of mAb disposition are discussed below; a more detailed description of general mAb PK/PD expectations may be found in the 2008 review by Wang et al.20.

Target mediated drug disposition (TMDD)

For most drug molecules, the interaction between the drug and its pharmacological receptor does not contribute substantially to the kinetics of drug distribution or elimination.However,as proposed by Gerhard Levy in 199421, and as described by the mathematical modeling of Mager and Jusko in 200122,in cases where target-drug binding affinity is very high, the interaction between target and drug may play a significant role in drug pharmacokinetics.This phenomenon, known as targetmediated drug disposition (TMDD), leads to non-linear,saturable distribution and elimination kinetics.High affinity mAb-target binding contributes to the apparent volume of mAb distribution, as a high degree of binding leads to a high ratio of the quantity of mAb bound to cellular target proteins, relative to the concentration of mAb in blood.Additionally, in many cases,mAb-target binding precipitates the endocytosis of the mAbtarget complex, with subsequent intracellular catabolism and elimination of the antibody.As such, target binding may lead to efficient mAb elimination.With increasing doses of mAb, the target becomes increasingly saturated with antibody, and this saturation leads to decreases in the apparent volume of mAb distribution and to decreases in the rate of antibody clearance(i.e., non-linear, dose-dependent pharmacokinetics).

For mAb exhibiting TMDD, intra- and inter-patient variability in target expression often is a prime determinant of pharmacokinetic variability.For example, patients with large tumor loads, and large amounts of tumor-associated target, may show much more rapid and extensive mAb distribution and elimination than observed in healthy individuals or in patients with low tumor volume.In many cases, administration of mAb leads to the destruction of cells that express the target and, consequently,mAb pharmacokinetics may be influenced by the therapeutic effects of the mAb.For example, in the clinical investigation of the pharmacokinetics of an anti-CD3 mAb, Meijer et al.observed that mAb elimination was more rapid for the first dose relative to the rate of mAb elimination observed for later doses (i.e., following the second, fourth, or tenth dose in a multiple-dose regimen).This finding was explained by the effect of the mAb on CD3-positive cells (i.e., depletion), which led to a reduction in target-mediated mAb clearance with increased treatment23.As such, for mAbs that exhibit TMDD, the ‘baseline’ target expression level, as well as the influence of mAb dosing on target expression, should be considered when evaluating mAb pharmacokinetics.Knowledge of changes in target expression due to disease progression or response to treatment may be crucial for the accurate prediction of the PK/PD of subsequent doses of mAbs.

Tumor distribution

Due to the large molecular weight and high polarity of antibodies,mAb demonstrate very slow rates of diffusion across cell membranes and, thus, comparatively slow rates of extravasation and tissue distribution (i.e., relative to small-molecule drugs).In comparison to the distribution of mAb in healthy tissues,distribution of mAb within tumors may be further impeded due to irregularities in the tumor vasculature, and due to high interstitial pressure in tumors, as described by Jain24.Moreover,the high affinity binding of mAb to target proteins within solid tumors may act as a barrier to distribution, as explained by the‘binding site barrier’ hypothesis.The impact of mAb binding on tumor distribution has been well illustrated by Fujimori et al., who utilized a modeling analysis demonstrating that high affinity (KA>1.0×109M–1) mAbs exhibit heterogeneous tumor distribution, with the majority of the molecules being ‘stuck’ at sites proximal to the point of extravasation within the tumor.The results of their simulations suggested that moderate affinity mAb(KA=5×107–1×108M–1) would allow optimal distribution25.Their predictions have been supported by experimental work performed by several investigators, including Juweid et al., who demonstrated that, following low doses, mAb intra-tumoral distribution was limited to areas adjacent to blood vessels, and the extent of tumor distribution was enhanced following high doses of mAb, consistent with the saturation of the binding site barrier26.

Pharmacologic mechanisms of action

As shown in Table 1 and Table 2, mAbs have been developed to engage a wide variety of cell surface and soluble target proteins.While several factors play a role in the pharmacologic mechanism of action for mAbs, the nature of the target and its role in tumor growth are crucial players in determining how mAb will exert therapeutic effects.Therapeutic responses to mAbs may be mediated through either the Fab or Fc region of the antibody.Key pharmacodynamic mechanisms for mAbs in oncology include:inhibition of cell signaling, induction of apoptosis, antibodydependent cellular cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC), and targeting a toxic payload to tumor cells(Figure 1).Additionally, there has been some interest in the development of mAbs known as ‘superagonists’ that stimulate immune function to accelerate immune clearance of tumor cells.It is important to note that a single mAb may act through a combination of mechanisms to achieve anti-tumor effects.

Inhibition of cell signaling

Monoclonal antibodies may antagonize cell signaling pathways by several mechanisms, including neutralization of soluble signaling factors (e.g., vascular endothelial growth factor,hepatocyte growth factor), binding to and blocking cell surface receptors (i.e., preventing receptor engagement with signaling factors), and by decreasing the expression of cell surface receptors.Of these mechanisms, perhaps the most interesting is the action of mAb to decrease receptor expression, which may be accomplished by ‘stripping’ the receptor from the cell surface or by accelerating the internalization and catabolism of the receptor.Dose requirements for the inhibition of signaling may be, in some cases, estimated based on the rate of production of the antibody target, whether it is a receptor or a soluble ligand.In most cases, blockade of cell signaling will not require engagement of the Fc domain of the mAb, and may be accomplished with administration of Fab fragments.For example, early work investigating the use of anti-EGFR mAbs in the treatment of cancer demonstrated that (Fab)2fragments were able to produce a dose-dependent reduction in tumor growth in a xenograft model, supporting the hypothesis that the activity of this mAb did not require Fc-dependent effector mechanisms27.

Antibody-dependent cellular cytotoxicity(ADCC)

Figure 1 Pharmacologic Mechanisms of Action for mAbs.Panel A, Inhibition of Cell Signaling via Binding to Soluble Targe; Panel B, Inhibition of Cell Signaling via Binding to Membrane-Bound Receptor; Panel C, Direct Induction of Apoptosis; Panel D, Antibody-Dependent Cellular Cytotoxicity; Panel E, Complement-Dependent Cytotoxicity; Panel F, CD28 Superagonist; Panel G, Delivery of Toxic Payload (Antibody-Drug Conjugate, Immunotoxin, Radioimmunoconjugate).

Early studies performed using cultured human monocytes in the presence and absence of murine IgG2a demonstrated that the ability of monocytes to kill tumor cells was significantly increased in the presence of IgG28.This phenomenon, which has been dubbed Antibody-Dependent Cellular Cytotoxicity(ADCC), is mediated by the interaction between the Fc region of an antibody and FcγRIIIa receptors present on the surface of immune cells.Brie fly, mAb may bind a cell surface target via its Fab region, and then engage leukocytes expressing FcγRIIIa via the Fc region of the mAb, leading to subsequent cell killing.

One example demonstrating the importance of the FcγR-mAb interaction has been provided by Cartron et al.29, who investigated the efficacy of rituximab in a panel of lymphoma patients.This team demonstrated that patients expressing a low affinity variant of FcγRIIIa, which contains a phenylalanine residue at position 158,received less bene fit (i.e., a shorter survival time) from rituximab treatment than patients who possess a high affinity variant of the receptor, with valine at position 158.This work strongly suggests that a significant fraction of the bene fit provided by rituximab is resultant from ADCC.

Complement-dependent cytotoxicity (CDC)

The ability of immunoglobulin molecules to recruit complement to mediate cell killing has been appreciated for several decades.Briefly, following mAb binding to a cell-surface target, Fc domains of the mAb may bind to soluble C1q (i.e., complement fixation), leading to stimulation of the complement pathway, and ultimately cell death.Complement fixation requires relatively high densities of mAb on the cell surface, as it has been estimated that C1q fixation requires separation of Fc domains by no more than ~40 nm30.In one example of the significance of CDC for mAb treatment of cancer, Capone and colleagues generated two monoclonal antibodies against breast cancer targets,and investigated possible mechanisms to explain their in vivo tumor cell killing.In vitro cell killing, for each mAb, required complement, implicating CDC as the primary mechanism of cell killing31.In vitro studies performed with matched chimeric mAbs of various subclasses have demonstrated that the IgG1 subclass has the greatest ability to induce cell death via CDC32.

Induction of apoptosis

Monoclonal antibody binding to cell surface receptors may lead to the induction of cell death via apoptotic pathways.For example, work by Trauth et al.33showed that anti-APO-1 mAbs induce apoptosis in tumor cells, in vitro and in vivo, in a manner distinct from ADCC and CDC.Briefly, they noted that cell death could occur after mAb binding under complement-free and serum-free conditions, and that the pattern of cell death was consistent with apoptosis and not necrosis, suggesting that the mAb-target interaction directly led to the induction of apoptosis.

Superagonists

In addition to the mechanisms of action discussed above, there has been an interest in developing immunomodulatory mAbs, which function as “superagonists”.Briefly, T cell stimulation typically requires a signal from the T Cell Receptor (TCR) and a costimulatory signal from CD2834.However, superagonist antibodies,such as the anti-CD28 mAb TGN-1412, have demonstrated an ability to stimulate T cell proliferation without TCR engagement35,thereby creating a possible mechanism for mAbs to increase immune-mediated clearance of cancers.However, superagonist mAbs may bring significant risks, as evidenced by the first-inman investigation of TGN-1412.Brie fly, the anti-CD28 mAb was dosed to six healthy volunteers, who all experienced a cytokine storm shortly after administration of the mAb, leading to severe side effects in all of the subjects36.It is possible that revised dosing schemes, possibly targeted low levels of mAb binding to stimulatory receptors, may allow for the desired anti-cancer effect without risk for toxicities associated with hyper-stimulation.

Delivery of toxic payloads (immunoconjugates)

The use of mAbs to deliver a highly potent payload to tumor sites is perhaps the epitome of Ehrlich’s ‘magic bullet’hypothesis.There are three major types of constructs which fall into this broad category: antibody-drug conjugates (ADCs),immunotoxins, and radioimmunoconjugates.Briefly, each construct is designed to employ the target specificity of a mAb to deliver a toxic payload selectively to tumor cells, potentially delivering high potency and low toxicity.Due to the complexity of these delivery systems, there are many potential issues which must be considered when developing an immunoconjugate,which have been outlined in detail in other reviews37,38.

The selective delivery of a small molecule chemotherapeutic agent to tumors using mAbs has been tested preclinically and clinically for several decades.The earliest examples of ADCs used mAbs to target clinically approved chemotherapeutics to tumors.However, these constructs often failed in clinical trials due to insufficient potency of the payload.A notable example of this failure was BR96-DOX, which showed remarkable antitumor activity in preclinical models39, but had unacceptable efficacy and toxicity pro files in clinical trials40.

Currently, ADC payloads are often selected from chemotherapeutic agents that have demonstrated unacceptable pharmacokinetic or toxicity profiles for clinical development, thereby ‘resurrecting’drug molecules that have been previously discarded.The most commonly clinically utilized agents at this time are maytansinoids,calicheamicins, and monomethyl auristatin E (MMAE).

While ADCs utilize small molecule chemotherapeutics as the cytotoxic agent, immunotoxins use highly potent bacterial and plant toxins to exert their antitumor effect.Currently, there are two anti-CD22 immunotoxins in clinical development41.Other payloads which have been investigated in the literature include ricin-like toxins42, as these molecules are extremely potent and are believed to have the capacity to induce apoptosis with only a single molecule entering the cytosol.

Radioimmunoconjugates, the final class of immunoconjugates,employ mAbs as targeting agents for selective delivery radionuclides to tumor cells.Several radionuclides have been investigated in preclinical and clinical trials; the two clinically approved agents employ90Y (β emitter) and131I (γ emitter).In many therapeutic protocols utilizing radioimmunotherapy,patients are pre-dosed with unlabeled (i.e., ‘cold’) mAb, followed by the administration of the radioimmunoconjugate.This strategy often allows minimization of off-target toxicities, via saturation and/or depletion of the target protein on healthy cells that may be associated with low density target expression.As such, pre-dosing often allows more selective uptake of radioimmunoconjugate in tumor cells, and improved pharmacokinetics of the conjugate43.

Currently marketed mAbs in oncology

FDA-approved mAbs used in oncology are summarized in Table 1.Important PK/PD considerations for each mAb have been detailed below.

Alemtuzumab

Alemtuzumab (Campath) is an anti-CD52 mAb approved for use in B-cell chronic lymphocytic leukemia (B-CLL).The PK of alemtuzumab has been described as being non-linear, with the maximal rate of elimination demonstrating co-variation with white blood cell counts, consistent with TMDD (note that CD52 is expressed on leukocytes)44.In addition to non-linear PK, alemtuzumab may display time- or treatment-dependent kinetics, where half-life increases after initial elimination of target-expressing cells.Key mechanisms of action of alemtuzumab include induction of apoptosis and CDC6.

In Phase III clinical trials, alemtuzumab was compared to chlorambucil, in previously untreated B-CLL patients.Those subjects who received alemtuzumab showed a 42%reduction in the risk of progression-free survival, as well as a significant (P<0.0001) improvement in overall response rates45.Additionally, other trials have demonstrated that this mAb is effective in treating patients with p53 mutations and deletions,which render chlorambucil treatment ineffective46.Alemtuzumab was withdrawn from the market in 2012, but its sponsors are seeking to rebrand it as a treatment for multiple sclerosis.

Bevacizumab

Bevacizumab (Avastin) is an anti-VEGF mAb that is approved for use in several cancers, including metastatic colorectal cancer(mCRC), non-squamous, non-small cell lung cancer (NSCLC),glioblastoma, and metastatic renal cell carcinoma.This mAb functions by binding to circulating VEGF, blocking its ability to bind to its target receptor, and blocking VEGF-mediated stimulation of pro-angiogenic signaling pathways.As is typical for mAbs which bind soluble ligands, bevacizumab displays linear PK, with a terminal half-life of ~20 days47.In trials, this mAb was added on to a standard chemotherapy combination of irinotecan, bolus fluorouracil, and leucovorin (IFL) to determine its efficacy in mCRC.Patients which were on the bevacizumab arm of the treatment showed improvements in all clinical efficacy endpoints tested in the study48.

Brentuximab vedotin

Brentuximab vedotin (Adcetris) is an anti-CD30 ADC approved as a second or third line therapy for the treatment of Hodgkin’s lymphoma (HL) and systemic anaplastic large cell lymphoma(ALCL).This construct consists of an anti-CD30 mAb linked to MMAE, which is a highly potent anti-mitotic agent.The clinical PK of brentuximab vedotin has been reported as ‘approximately proportional to dose’, with a half-life of 4-6 days for the intact ADC and 3-4 days for MMAE49.Due to overwhelmingly positive results in two Phase II clinical trials50,51, this ADC was granted an accelerated approval.Results from these studies demonstrated a 75% response rate in HL51and an 86% response rate in ALCL50,indicating that this drug has great potential in the treatment of these cancers.

Cetuximab

Cetuximab (Erbitux) is an anti-EGFR mAb which is approved for the treatment of squamous cell carcinoma of the head and neck(SCCHN) and K-Ras mutation negative, EGFR positive, mCRC.In a dose-ranging (50-500 mg/m2) study, cetuximab clearance was found to range from 20.0-83.7 mL/h/m2, indicating the presence of a saturable elimination pathway for this mAb, likely consistent with TMDD52.Cetuximab has been reported to exert its anti-tumor properties via signal inhibition, ADCC, and CDC8.When added to standard radiotherapy in patients diagnosed with SCCHN, cetuximab increased overall survival by 19.7 months and progression-free survival by 9.5 months, indicating bene fit compared to the standard of care53.K-Ras status has been investigated as a predictor of response to cetuximab, and trials have shown that patients positive for mutations in K-Ras have significantly lower responses when treated with cetuximab, likely due to the constitutive activation status of the variant protein54.

Denosumab

Denosumab (Xgeva) is an anti-RANKL mAb approved for the treatment of bone metastases from solid tumors and for unresectable giant cell bone tumors.Binding of denosumab to RANKL prevents interaction with RANK, thereby preventing osteoclasts from resorbing bone.The pharmacokinetics of denosumab have been reported as non-linear, with a maximal clearance value of 85 mL/h, and with saturation of the targetmediated pathway being achieved with doses of 120 mg/month55.A study in patients with breast cancer bone metastases demonstrated that denosumab was superior to the bisphosphonate zoledronic acid in the prevention of skeletal-related events such as pathological fractures, spinal cord compression, and bone surgery/radiation56.This indicates that use of this mAb may help to reduce some of the consequences of bone metastases in patients,improving their quality of life.

Ibritumomab tiuxetan

Ibritumomab tiuxetan (Zevalin) is an anti-CD20 radioimmunoconjugate indicated in the treatment of non-Hodgkin’s lymphoma (NHL).Administration of this drug is performed by first infusing rituximab followed by ibritumomab tiuxetan conjugated with either111In (imaging) or90Y(treatment).Clinical trials showed an increase in progressionfree survival of 1.1 months and an increased complete response rate when treating patients with90Y-ibritumomab tiuxetan compared to rituximab treatment, which indicates that delivery of the radioisotope allows for improved outcomes compared to a‘naked’ mAb delivered to the same target57.

Ipilimumab

Ipilimumab (Yervoy) is an anti-CTLA-4 mAb indicated for the treatment of unresectable or metastatic melanoma.In metastatic melanoma patients, ipilimumab pharmacokinetics were found to be linear over a dose range of 3-10 mg/kg, with an average clearance value of 14.9 mL/h58.Because this mAb targets an antigen expressed on T-cells, distributional challenges are not likely to be a significant determinant of its efficacy.Binding of ipilimumab to CTLA-4 relieves inhibitory signals on T-cell proliferation, thereby improving immune function in patients.Effectively, ipilimumab treatment serves to counteract the immune evasion mechanisms utilized by tumors to ensure their continued survival.

Patients with unresectable stage III or IV melanoma were treated with ipilimumab and/or a gp100 peptide vaccine, and it was observed that ipilimumab alone improved overall survival by 3.6 months compared to vaccine alone (6.4-10.0 months)59.Additionally, early clinical trial results indicated that treatment with ipilimumab led to an increase in lymphocyte activation markers, indicating improved immune functions in patients receiving mAb therapy60.

Ofatumumab

Ofatumumab (Arzerra) is an anti-CD20 mAb currently approved for use in treatment of chronic lymphocytic leukemia (CLL).In patients, ofatumumab displays both dose- and treatmentdependent pharmacokinetics over a dose range of 500-2,000 mg.On the first dose, clearance ranged from 65-215 mL/h, while after the fourth dose, clearance decreased to 10-28 mL/h61.Brie fly, this suggests that the elimination of ofatumumab is target-mediated,and that wipeout of CD20-positive cells after early doses contributes to a slower clearance on subsequent doses.It has been suggested that the primary mechanisms by which ofatumumab kills cancer cells are ADCC and CDC11.In trials as a single agent in CLL patients refractory to standard treatments (fludarabine),ofatumumab improved response rates from 23% to 47%-58%along with a median progression-free survival time of six months62.Additionally, trial results in follicular lymphoma have indicated that ofatumumab has some activity in rituximab-refractory patients63.

Panitumumab

Panitumumab (Vectibix) is an anti-EGFR mAb indicated for use in the treatment of mCRC.Clearance of panitumumab is markedly non-linear, approaching values of ~75 mL/d/kg at low doses (0.75 mg/kg), and decreases to ~4 mL/d/kg at higher doses (>2 mg/kg)64.Interestingly, panitumumab is eliminated more slowly than cetuximab, indicating that the target-mediated pathway may be less relevant for this mAb64.Additionally, the binding site barrier has been reported as relevant in preclinical models, with deeper penetration occurring into tumors at higher doses (500 µg) and later time points (96 h)65.The reported mechanisms of action for panitumumab include signal inhibition, ADCC, and CDC10.

Clinical trials in mCRC patients who had progressed after standard therapy, compared to best supportive care alone, indicated that panitumumab improved median progression-free survival from 7.3 to 8.0 weeks66.Additionally, as with other anti-EGFR mAbs,wild type K-Ras is necessary for response to treatment67.

Pertuzumab

Pertuzumab (Perjeta) is an anti-HER2 mAb indicated for the treatment of metastatic breast cancer.The PK of pertuzumab was found to be linear in the dose range of 0.5-15 mg/kg (mean clearance has been reported to be 0.214 L/d)68.The interaction of pertuzumab with HER2 is such that it blocks the interaction of HER2 and HER3, preventing dimerization and subsequent intracellular signaling13.In addition to this direct, Fab-mediated mechanism, pertuzumab also may induce cell death via ADCC13.Because pertuzumab targets a different motif in HER2 than trastuzumab, combination therapy was investigated to determine if there could be synergistic benefits.Addition of pertuzumab to trastuzumab and docetaxel therapy led to an increase in progression-free survival by 6.1 months (12.4-18.5 months),producing a clear clinical bene fit when added to standard therapy69.

Rituximab

Rituximab (Rituxan) is an anti-CD20 mAb indicated as a therapy for treatment of NHL and CLL and was the first mAb approved by the FDA for use in oncology.In clinical trials for NHL,rituximab was found to have non-stationary pharmacokinetics,with clearance decreasing from 38.2 mL/h after the first dose to 9.2 mL/h after the fourth dose70.This observation may be due to a reduction in TMDD caused by wipeout of CD20-positive cells after the initial infusion.In clinical trials, addition of rituximab to the standard CHOP-21 chemotherapy regimen was associated with an improvement in 3-year progression-free survival (85%vs.68%)17.In a different study, lymphoma patients treated with rituximab alone had an overall response rate of 50%, with a median duration of response of 8.6 months71.

Tositumomab

Tositumomab (Bexxar) is an anti-CD20 mAb indicated for the treatment of relapsed or refractory NHL, and is administered first as a ‘cold’ mAb, followed by administration of a ‘hot’131I-labeled mAb.It was noted in clinical trials that patients with a greater tumor burden were associated with increased volume of distribution, faster clearance, and shorter half-life of tositumomab72, indicating that TMDD is likely relevant in the pharmacokinetics of this drug.As this radioimmunotherapy regimen is not intended for first-line treatment of NHL, the pivotal clinical trial evaluated tositumomab compared to standard last qualifying chemotherapy regimens.In this trial,patients receiving tositumomab had a median duration of response of 6.4 months, compared to 3.4 months in the control group, with 3% of patients achieving a complete response73.As of February 2014, tositumomab will be withdrawn from the market in the U.S.and Canada, due to a manufacturer’s decision74.

Trastuzumab

Trastuzumab (Herceptin) is an anti-HER2 mAb approved for the treatment of breast cancer, metastatic gastric cancer,and metastatic gastroesophageal junction adenocarcinoma.The half-life of trastuzumab has been observed to range from 1.1 days (10 mg dose) to 23 days (500 mg dose) in clinical trials75.Additionally, population pharmacokinetic modeling has suggested that clearance of trastuzumab is directly related to shed extracellular domain of HER2 and has a weaker association with the number of tumor metastases75.In mice, tumor distribution was found to be more uniform at higher doses and at later time points, suggesting that saturation of the binding site barrier may be crucial in optimizing the efficacy of trastuzumab76.

Phase III clinical trials investigated the potential benefits of adding trastuzumab to standard chemotherapy in previously untreated breast cancer patients with HER2-overexpressing tumors.The trial results indicated that addition of trastuzumab was associated with a 4.8-month increase in overall survival(20.3-25.1 months) and a 2.8-month increase in progression-free survival (4.6-7.4 months)77.

Trastuzumab emtansine

Trastuzumab emtansine (Kadcyla) is an anti-HER2 ADC indicated for treatment of metastatic breast cancer.This ADC consists of the anti-HER2 mAb trastuzumab linked to mertansine, a maytansinoid which exerts its cytotoxic effect via tubulin binding.In addition to delivery of mertansine, this ADC retains the mechanisms of action associated with the ‘naked’ mAb, trastuzumab (signal inhibition and ADCC)14.In dose-escalation studies, there was an observed trend towards faster clearance at doses less than 1.2 mg/kg/3 weeks(CL=21.1-27.8 mL/d/kg); however, linear PK was observed at higher doses (CL=7.13-12.7 mL/d/kg), indicating a saturable clearance pathway78.Additionally, the observed free DM-1(payload) concentrations did not exceed 25 ng/mL, indicating that the conjugate is stable in plasma78.

In trials with patients diagnosed with advanced breast cancer,trastuzumab emtansine increased progression free survival relative to standard of care (lapatinib plus capecitabine) by 3.2 months(6.4-9.6 months) and median overall survival by 5.8 months (25.1-30.9 months)79.Additionally, the ADC was shown to have efficacy in patients whose disease had progressed after prior HER2-targeted therapy (progression-free survival =4.6 months)80.

mAbs in late-stage clinical trials

In addition to the currently marketed mAb products, there is a rich pipeline of products that are currently being investigated in clinical trials.Here we summarize mAb-based products that are in late-stage (Phase II and Phase III) clinical trials for cancer indications to give an overview of products that may be clinically available in the next few years.

Elotuzumab

Currently in Phase III clinical trials, elotuzumab is an anti-CS1(CD2 subset 1) mAb being investigated as a treatment option for multiple myeloma.Results of a dose-escalation study indicate that elotuzumab displays clear non-linear pharmacokinetics,with clearance decreasing from 71.4 to 15.7 mL/h over the dose range of 0.5-20 mg/kg81.Phase I clinical trial results indicated that this mAb has efficacy in treatment of multiple myeloma in combination with lenalidomide and dexamethasone (objective response rate of 82%)82.

Farletuzumab

Farletuzumab is an anti-folate receptor α (FRA) mAb that is being investigated for use in ovarian cancer, along with other epithelial cancers.Early clinical trial results in relapsed platinum sensitive ovarian cancer indicated that as a single agent farletuzumab induced stable disease at best in 30% of patients, while in combination with carboplatin and taxane 95%of patients achieved stable disease or better83.However, when this drug progressed into Phase III clinical trials, patients did not show a statistically significant improvement in progressionfree survival relative to the control arm84, leaving the future of farletuzumab in ovarian cancer treatment uncertain.

Inotuzumab ozogamicin

Inotuzumab ozogamicin is an ADC directed against CD22 which has progressed into Phase III clinical trials for the treatment of NHL and acute lymphocytic leukemia (ALL).In this construct,ozogamicin (a calicheamicin derivative) is the toxic payload used to destroy tumor cells.Phase I clinical trial data indicates that this ADC displays non-stationary PK, with decreased clearance after multiple dosing, relative to the first dose, indicative of the modulation of a target-mediated pathway with initial doses85,86.

Trials for the ADC in the treatment of NHL have been halted as of May 2013, as the drug in combination with rituximab was not likely to result in a significant improvement in overall survival, based on the planned interim analysis87.However, trials for other conditions, such as ALL, are ongoing and have shown promising results, with 58% of patients achieving a bone marrow complete response in a published study88.

Moxetumomab pasudotox

Moxetumomab pasudotox is an anti-CD22 immunotoxin consisting of an Fv as the targeting moiety fused to Pseudomonas exotoxin-A, which is being investigated in Phase III clinical trials for hairy cell leukemia (HCL).Published Phase I trial data is promising with an overall response rate of 86%89.Pharmacokinetic analysis of data has suggested that tumor burden is a significant covariate on clearance, demonstrating that TMDD may be important in the in vivo behavior of this drug90.

Naptumomab estafenatox

Naptumomab estafenatox is an anti-5T4 fusion protein consisting of a Fab fragment fused to staphylococcal enterotoxin E, which is being studied for use in renal cell carcinoma.This construct is proposed to function as a superantigen, recruiting immune effectors to the target site91.Within individual cycles of therapy, it was noted that the PK of naptumomab estafenatox was linear92.When comparing the first and second cycles of therapy,clearance was dramatically increased after the second dose of the drug (increased from 0.11 to 6.39 L/h/kg), which the investigators suggest was due to formation of antibodies against the construct92.Phase I trials support this mechanism as posttreatment tumor biopsies had significant T cell in filtration, and the construct has measurable anti-tumor activity in the clinic92.

Necitumumab

Necitumumab is an anti-EGFR mAb being developed for use in the treatment of NSCLC.In clinical trials, necitumumab displayed both dose- and treatment-dependent PK, with clearance values after the first dose (100-1,000 mg/week)ranging 13.9-53.2 mL/h, whereas after the final dose, clearance ranged from 1.45-40.2 mL/h93.Eli Lilly has recently announced that Phase III trials in stage IV NSCLC, where necitumumab was added to standard chemotherapy, have met the primary endpoint of increased overall survival, and they intend to submit data for regulatory approval in 201494.

Nivolumab

Nivolumab is an anti-programmed cell death receptor 1 (PD1)mAb which is currently in clinical trials for the treatment of NSCLC, renal cell carcinoma, and melanoma.The PK of nivolumab shows modest non-linearity, with a terminal halflife of 12 days at dose levels less than 3 mg/kg and a half-life of 20 days at a dose of 10 mg/kg, indicating that there is a saturable clearance pathway for this mAb95.To date, the most striking results have been observed when nivolumab was administered in combination with ipilimumab in stage III or IV melanoma patients.Clinical activity was observed in 65% of patients receiving the combination, and 53% of patients who received the maximum dose had a tumor reduction of greater than 80%96.

Onartuzumab

Onartuzumab is an anti-hepatocyte growth factor receptor(c-Met) monovalent mAb in trials for use in NSCLC and gastric cancer.At dose levels greater than 4 mg/kg, the PK appears to be linear; however, at a low dose (1 mg/kg),clearance is approximately two-fold greater than at the higher doses, indicating that there may be a readily saturated TMDD pathway97.However, population PK modeling based on Phase I and II clinical trials indicates that a 15 mg/kg dose every three weeks is adequate for the desired exposure, thereby minimizing the influence of the target-mediated clearance pathway97.

Racotumomab

Racotumomab is an anti-GM3 mAb in Phase III clinical trials as a cancer vaccine for advanced NSCLC.Phase I clinical trial data in patients with NSCLC showed that treatment with racotumomab (4+ doses) produced a specific antibody response against both the mAb and against the specific target, along with generating a favorable survival pro file98.

Rilotumumab

Rilotumumab is an anti-hepatocyte growth factor (HGF)mAb being investigated for the treatment of gastric and gastroesophageal cancers.Results from dose escalation studies(0.5-20 mg/kg) indicate that rilotumumab displays linear PK in man, with the average clearance being 0.141 mL/h/kg and no clear dose-dependent changes observed99.

Conclusion

In this review, we have summarized the pharmacokinetics and pharmacodynamics of monoclonal antibodies used for oncologic indications, including mechanisms of action.Monoclonal antibodies may be considered to be the most important class of anti-cancer agents, with 14 mAbs in current clinical use, and with many more in development.This drug class, which achieves effects through a variety of mechanisms, provides several bene fits over traditional small-molecule chemotherapeutic agents,including slow rates of elimination (thus allowing infrequent dosing), high efficacy, and low off-target toxicity.Based on the promise of agents in development, it is anticipated that anticancer mAbs will continue to grow in importance over the next 5-10 years.

Conflict of interest

No conflicts of interest are disclosed.

1.Hericourt J, Richet C.Physologie Pathologique-de la serotherapie dans la traitement du cancer.Comptes Rendus Hebd.Seanc Acad Sci 1895;121:567-569.

2.Ehrlich P, eds.Collected studies on immunity.New York: John Wiley & Sons; 1906.

3.Köhler G, Milstein C.Continuous cultures of fused cells secreting antibody of prede fined specificity.Nature 1975;256:495-497.

4.Smith MR.Rituximab (monoclonal anti-CD20 antibody):mechanisms of action and resistance.Oncogene 2003;22:7359-7368.5.Hudis CA.Trastuzumab--mechanism of action and use in clinical practice.N Engl J Med 2007;357:39-51.

6.Alinari L, Lapalombella R, Andritsos L, Baiocchi RA, Lin TS, Byrd JC.Alemtuzumab (Campath-1H) in the treatment of chronic lymphocytic leukemia.Oncogene 2007;26:3644-3653.

7.Srinivasan A, Mukherji SK.Tositumomab and iodine I 131 tositumomab (Bexaar).AJNR Am J Neuroradiol 2011;32:637-638.

8.Zhuang H, Xue ZY, Wang L, Li XY, Zhang N, Zhang RX.Efficacy and Immune Mechanisms of Cetuximab for the Treatment of Metastatic Colorectal Cancer.Clin Oncol Cancer Res 2011;8:207-214.

9.Wang Y, Fei D, Vanderlaan M, Song A.Biological activity of bevacizumab, a humanized anti-VEGF antibody in vitro.Angiogenesis 2004;7:335-345.

10.Schneider-Merck T, Lammerts van Bueren JJ, Berger S, Rossen K, van Berkel PH, Derer S, et al.Human IgG2 antibodies against epidermal growth factor receptor effectively trigger antibodydependent cellular cytotoxicity but, in contrast to IgG1, only by cells of myeloid lineage.J Immunol 2010;184:512-520.

11.Nightingale G.Ofatumumab: a novel anti-CD20 monoclonal antibody for treatment of refractory chronic lymphocytic leukemia.Ann Pharmacother 2011;45:1248-1255.

12.Lipson EJ, Drake CG.Ipilimumab: an anti-CTLA-4 antibody for metastatic melanoma.Clin Cancer Res 2011;17:6958-6962.

13.Capelan M, Pugliano L, De Azambuja E, Bozovic I, Saini KS,Sotiriou C, et al.Pertuzumab: new hope for patients with HER2-positive breast cancer.Ann Oncol 2013;24:273-282.

14.Junttila TT, Li G, Parsons K, Phillips GL, Sliwkowski MX.Trastuzumab-DM1 (T-DM1) retains all the mechanisms of action of trastuzumab and efficiently inhibits growth of lapatinib insensitive breast cancer.Breast Cancer Res Treat 2011;128:347-356.

15.Reichert JM.Antibodies to watch in 2013: Mid-year update.Mabs 2013;5:513-517.

16.van Rhee F, Szmania SM, Dillon M, van Abbema AM, Li X,Stone MK, et al.Combinatorial efficacy of anti-CS1 monoclonal antibody elotuzumab (HuLuc63) and bortezomib against multiple myeloma.Mol Cancer Ther 2009;8:2616-2624.

17.Pfreundschuh M, Trümper L, Osterborg A, Pettengell R, Trneny M, Imrie K, et al.CHOP-like chemotherapy plus rituximab versus CHOP-like chemotherapy alone in young patients with goodprognosis diffuse large-B-cell lymphoma: a randomised controlled trial by the MabThera International Trial (MInT) Group.Lancet Oncol 2006;7:379-391.

18.Habermann TM, Weller EA, Morrison VA, Gascoyne RD,Cassileth PA, Cohn JB, et al.Rituximab-CHOP versus CHOP alone or with maintenance rituximab in older patients with diffuse large B-cell lymphoma.J Clin Oncol 2006;24:3121-3127.

19.Waldmann TA, Strober W.Metabolism of immunoglobulins.Prog Allergy 1969;13:1-110.

20.Wang W, Wang EQ, Balthasar JP.Monoclonal antibody pharmacokinetics and pharmacodynamics.Clin Pharmacol Ther 2008;84:548-558.

21.Levy G.Pharmacologic target-mediated drug disposition.Clin Pharmacol Ther 1994;56:248-252.

22.Mager DE, Jusko WJ.General pharmacokinetic model for drugs exhibiting target-mediated drug disposition.J Pharmacokinet Pharmacodyn 2001;28:507-532.

23.Meijer RT, Koopmans RP, ten Berge IJ, Schellekens PT.Pharmacokinetics of murine anti-human CD3 antibodies in man are determined by the disappearance of target antigen.J Pharmacol Exp Ther 2002;300:346-353.

24.Jain RK.Physiological barriers to delivery of monoclonal antibodies and other macromolecules in tumors.Cancer Res 1990;50:814s-819s.

25.Fujimori K, Covell DG, Fletcher JE, Weinstein JN.A modeling analysis of monoclonal antibody percolation through tumors: a binding-site barrier.J Nucl Med 1990;31:1191-1198.

26.Juweid M, Neumann R, Paik C, Perez-Bacete MJ, Sato J, van Osdol W, et al.Micropharmacology of monoclonal antibodies in solid tumors: direct experimental evidence for a binding site barrier.Cancer Res 1992;52:5144-5153.

27.Fan Z, Masui H, Altas I, Mendelsohn J.Blockade of epidermal growth factor receptor function by bivalent and monovalent fragments of 225 anti-epidermal growth factor receptor monoclonal antibodies.Cancer Res 1993;53:4322-4328.

28.Steplewski Z, Lubeck MD, Koprowski H.Human macrophages armed with murine immunoglobulin G2a antibodies to tumors destroy human cancer cells.Science 1983;221:865-867.

29.Cartron G, Dacheux L, Salles G, Solal-Celigny P, Bardos P,Colombat P, et al.Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcgammaRIIIa gene.Blood 2002;99:754-758.

30.Paul WE, eds.Fundamental immunology.Philadelphia: LippincottWilliams & Wilkins; 2003.

31.Capone PM, Papsidero LD, Croghan GA, Chu TM.Experimental tumoricidal effects of monoclonal antibody against solid breast tumors.Proc Natl Acad Sci U S A 1983;80:7328-7332.

32.Brüggemann M, Williams GT, Bindon CI, Clark MR, Walker MR,Jefferis R, et al.Comparison of the effector functions of human immunoglobulins using a matched set of chimeric antibodies.J Exp Med 1987;166:1351-1361.

33.Trauth BC, Klas C, Peters AM, Matzku S, Möller P, Falk W, et al.Monoclonal antibody-mediated tumor regression by induction of apoptosis.Science 1989;245:301-305.

34.Acuto O, Michel F.CD28-mediated co-stimulation: a quantitative support for TCR signalling.Nat Rev Immunol 2003;3:939-951.

35.Tacke M, Hanke G, Hanke T, Hünig T.CD28-mediated induction of proliferation in resting T cells in vitro and in vivo without engagement of the T cell receptor: evidence for functionally distinct forms of CD28.Eur J Immunol 1997;27:239-247.

36.Suntharalingam G, Perry MR, Ward S, Brett SJ, Castello-Cortes A, Brunner MD, et al.Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412.N Engl J Med 2006;355:1018-1028.

37.Gerber HP, Koehn FE, Abraham RT.The antibody-drug conjugate: an enabling modality for natural product-based cancer therapeutics.Nat Prod Rep 2013;30:625-639.

38.Lin K, Tibbitts J.Pharmacokinetic considerations for antibody drug conjugates.Pharm Res 2012;29:2354-2366.

39.Trail PA, Willner D, Lasch SJ, Henderson AJ, Hofstead S, Casazza AM, et al.Cure of xenografted human carcinomas by BR96-doxorubicin immunoconjugates.Science 1993;261:212-215.

40.Tolcher AW, Sugarman S, Gelmon KA, Cohen R, Saleh M,Isaacs C, et al.Randomized phase II study of BR96-doxorubicin conjugate in patients with metastatic breast cancer.J Clin Oncol 1999;17:478-484.

41.Shah NN, Bhojwani D, Silverman LB, Whitlock JA, Richards K,Stetler-Stevenson M, et al.A novel anti-CD22 immunotoxin,moxetumomab pasudotox (Ha22, Cat-8015): activity in pediatric patients with relapsed acute lymphoblastic leukemia (All) after allogeneic hematopoietic stem cell transplantation (Sct).Biol Blood Marrow Tr 2012;18:S234-S234.

42.Vitetta ES, Stone M, Amlot P, Fay J, May R, Till M, et al.Phase I immunotoxin trial in patients with B-cell lymphoma.Cancer Res 1991;51:4052-4058.

43.Sharkey RM, Press OW, Goldenberg DM.A re-examination of radioimmunotherapy in the treatment of non-Hodgkin lymphoma:prospects for dual-targeted antibody/radioantibody therapy.Blood 2009;113:3891-3895.

44.Mould DR, Baumann A, Kuhlmann J, Keating MJ, Weitman S,Hillmen P, et al.Population pharmacokinetics-pharmacodynamics of alemtuzumab (Campath) in patients with chronic lymphocytic leukaemia and its link to treatment response.Br J Clin Pharmacol 2007;64:278-291.

45.Hillmen P, Skotnicki AB, Robak T, Jaksic B, Dmoszynska A,Wu J, et al.Alemtuzumab compared with chlorambucil as firstline therapy for chronic lymphocytic leukemia.J Clin Oncol 2007;25:5616-5623.

46.Lozanski G, Heerema NA, Flinn IW, Smith L, Harbison J,Webb J, et al.Alemtuzumab is an effective therapy for chronic lymphocytic leukemia with p53 mutations and deletions.Blood 2004;103:3278-3281.

47.Lu JF, Bruno R, Eppler S, Novotny W, Lum B, Gaudreault J.Clinical pharmacokinetics of bevacizumab in patients with solid tumors.Cancer Chemother Pharmacol 2008;62:779-786.

48.Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T,Hainsworth J, Heim W, et al.Bevacizumab plus irinotecan,fluorouracil, and leucovorin for metastatic colorectal cancer.N Engl J Med 2004;350:2335-2342.

49.Younes A, Bartlett NL, Leonard JP, Kennedy DA, Lynch CM,Sievers EL, et al.Brentuximab vedotin (SGN-35) for relapsed CD30-positive lymphomas.N Engl J Med 2010;363:1812-1821.

50.Pro B, Advani R, Brice P, Bartlett NL, Rosenblatt JD, Illidge T, et al.Brentuximab vedotin (SGN-35) in patients with relapsed or refractory systemic anaplastic large-cell lymphoma: results of a phase II study.J Clin Oncol 2012;30:2190-2196.

51.Younes A, Gopal AK, Smith SE, Ansell SM, Rosenblatt JD, Savage KJ, et al.Results of a pivotal phase II study of brentuximab vedotin for patients with relapsed or refractory Hodgkin’s lymphoma.J Clin Oncol 2012;30:2183-2189.

52.Tan AR, Moore DF, Hidalgo M, Doroshow JH, Poplin EA, Goodin S, et al.Pharmacokinetics of cetuximab after administration of escalating single dosing and weekly fixed dosing in patients with solid tumors.Clin Cancer Res 2006;12:6517-6522.

53.Bonner JA, Harari PM, Giralt J, Azarnia N, Shin DM, Cohen RB,et al.Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck.N Engl J Med 2006;354:567-578.

54.Karapetis CS, Khambata-Ford S, Jonker DJ, O’Callaghan CJ, Tu D,Tebbutt NC, et al.K-ras mutations and bene fit from cetuximab in advanced colorectal cancer.N Engl J Med 2008;359:1757-1765.

55.Gibiansky L, Sutjandra L, Doshi S, Zheng J, Sohn W, Peterson MC, et al.Population pharmacokinetic analysis of denosumab in patients with bone metastases from solid tumours.Clin Pharmacokinet 2012;51:247-260.

56.Stopeck AT, Lipton A, Body JJ, Steger GG, Tonkin K, de Boer RH,et al.Denosumab compared with zoledronic acid for the treatment of bone metastases in patients with advanced breast cancer: a randomized, double-blind study.J Clin Oncol 2010;28:5132-5139.

57.Witzig TE, Gordon LI, Cabanillas F, Czuczman MS,Emmanouilides C, Joyce R, et al.Randomized controlled trial of yttrium-90-labeled ibritumomab tiuxetan radioimmunotherapy versus rituximab immunotherapy for patients with relapsed or refractory low-grade, follicular, or transformed B-cell non-Hodgkin’s lymphoma.J Clin Oncol 2002;20:2453-2463.

58.Cameron F, Whiteside G, Perry C.Ipilimumab: first global approval.Drugs 2011;71:1093-1104.

59.Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA,Haanen JB, et al.Improved survival with ipilimumab in patients with metastatic melanoma.N Engl J Med 2010;363:711-723.

60.Phan GQ, Yang JC, Sherry RM, Hwu P, Topalian SL,Schwartzentruber DJ, et al.Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma.Proc Natl Acad Sci U S A 2003;100:8372-8377.

61.Coiffier B, Losic N, Rønn BB, Lepretre S, Pedersen LM,Gadeberg O, et al.Pharmacokinetics and pharmacokinetic/pharmacodynamic associations of ofatumumab, a human monoclonal CD20 antibody, in patients with relapsed or refractory chronic lymphocytic leukaemia: a phase 1-2 study.Br J Haematol 2010;150:58-71.

62.Wierda WG, Kipps TJ, Mayer J, Stilgenbauer S, Williams CD, Hellmann A, et al.Ofatumumab as single-agent CD20 immunotherapy infludarabine-refractory chronic lymphocytic leukemia.J Clin Oncol 2010;28:1749-1755.

63.Czuczman MS, Fayad L, Delwail V, Cartron G, Jacobsen E,Kuliczkowski K, et al.Ofatumumab monotherapy in rituximabrefractory follicular lymphoma: results from a multicenter study.Blood 2012;119:3698-3704.

64.Yang BB, Lum P, Chen A, Arends R, Roskos L, Smith B, et al.Pharmacokinetic and pharmacodynamic perspectives on the clinical drug development of panitumumab.Clin Pharmacokinet 2010;49:729-740.

65.Freeman DJ, McDorman K, Ogbagabriel S, Kozlosky C, Yang BB,Doshi S, et al.Tumor penetration and epidermal growth factor receptor saturation by panitumumab correlate with antitumor activity in a preclinical model of human cancer.Mol Cancer 2012;11:47.

66.Van Cutsem E, Peeters M, Siena S, Humblet Y, Hendlisz A,Neyns B, et al.Open-label phase III trial of panitumumab plus best supportive care compared with best supportive care alone in patients with chemotherapy-refractory metastatic colorectal cancer.J Clin Oncol 2007;25:1658-1664.

67.Amado RG, Wolf M, Peeters M, Van Cutsem E, Siena S, Freeman DJ, et al.Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer.J Clin Oncol 2008;26:1626-1634.

68.Ng CM, Lum BL, Gimenez V, Kelsey S, Allison D.Rationale for fixed dosing of pertuzumab in cancer patients based on population pharmacokinetic analysis.Pharm Res 2006;23:1275-1284.

69.Baselga J, Cortés J, Kim SB, Im SA, Hegg R, Im YH, et al.Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer N Engl J Med 2012;366:109-119.

70.Berinstein NL, Grillo-López AJ, White CA, Bence-Bruckler I,Maloney D, Czuczman M, et al.Association of serum Rituximab(IDEC-C2B8) concentration and anti-tumor response in the treatment of recurrent low-grade or follicular non-Hodgkin’s lymphoma.Ann Oncol 1998;9:995-1001.

71.Maloney DG, Grillo-López AJ, White CA, Bodkin D, Schilder RJ, Neidhart JA, et al.IDEC-C2B8 (Rituximab) anti-CD20 monoclonal antibody therapy in patients with relapsed low-grade non-Hodgkin’s lymphoma.Blood 1997;90:2188-2195.

72.BEXXAR (package insert) (2013) Glaxo-Smith-Kline, Research Triangle Park, NC.

73.Kaminski MS, Zelenetz AD, Press OW, Saleh M, Leonard J,Fehrenbacher L, et al.Pivotal study of iodine I 131 tositumomab for chemotherapy-refractory low-grade or transformed low-grade B-cell non-Hodgkin’s lymphomas.J Clin Oncol 2001;19:3918-3928.

74.GSK to discontinue manufacture and sale of the BEXXAR®therapeutic regimen (tositumomab and iodine I 131 tositumomab).Press Release.August 7, 2013.

75.Bruno R, Washington CB, Lu JF, Lieberman G, Banken L, Klein P.Population pharmacokinetics of trastuzumab in patients with HER2+ metastatic breast cancer.Cancer Chemother Pharmacol 2005;56:361-369.

76.Lee CM, Tannock IF.The distribution of the therapeutic monoclonal antibodies cetuximab and trastuzumab within solid tumors.BMC Cancer 2010;10:255.

77.Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, et al.Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2.N Engl J Med 2001;344:783-792.

78.Girish S, Gupta M, Wang B, Lu D, Krop IE, Vogel CL, et al.Clinical pharmacology of trastuzumab emtansine (T-DM1): an antibodydrug conjugate in development for the treatment of HER2-positive cancer.Cancer Chemother Pharmacol 2012;69:1229-1240.

79.Verma S, Miles D, Gianni L, Krop IE, Welslau M, Baselga J, et al.Trastuzumab emtansine for HER2-positive advanced breast cancer.N Engl J Med 2012;367:1783-1791.

80.Burris HA 3rd, Rugo HS, Vukelja SJ, Vogel CL, Borson RA,Limentani S, et al.Phase II study of the antibody drug conjugate trastuzumab-DM1 for the treatment of human epidermal growth factor receptor 2 (HER2)-positive breast cancer after prior HER2-directed therapy.J Clin Oncol 2011;29:398-405.

81.Zonder JA, Mohrbacher AF, Singhal S, van Rhee F, Bensinger WI,Ding H, et al.A phase 1, multicenter, open-label, dose escalation study of elotuzumab in patients with advanced multiple myeloma.Blood 2012;120:552-559.

82.Lonial S, Vij R, Harousseau JL, Facon T, Moreau P, Mazumder A,et al.Elotuzumab in combination with lenalidomide and low-dose dexamethasone in relapsed or refractory multiple myeloma.J Clin Oncol 2012;30:1953-1959.

83.Armstrong DK, White AJ, Weil SC, Phillips M, Coleman RL.Farletuzumab (a monoclonal antibody against folate receptor alpha) in relapsed platinum-sensitive ovarian cancer.Gynecol Oncol 2013;129:452-458.

84.Eisai announces results of phase III study of anticancer agent farletuzumab in patients with relapsed platinum-sensitive ovarian cancer.Press Release.January 11, 2013.

85.Ogura M, Hatake K, Ando K, Tobinai K, Tokushige K, Ono C,et al.Phase I study of anti-CD22 immunoconjugate inotuzumab ozogamicin plus rituximab in relapsed/refractory B-cell non-Hodgkin lymphoma.Cancer Sci 2012;103:933-938.

86.Ogura M, Tobinai K, Hatake K, Uchida T, Kasai M, Oyama T, et al.Phase I study of inotuzumab ozogamicin (CMC-544) in Japanese patients with follicular lymphoma pretreated with rituximab-based therapy.Cancer Sci 2010;101:1840-1845.

87.P fizer discontinues phase 3 study of inotuzumab ozogamicin in relapsed or refractory aggressive non-Hodgkin lymphoma (NHL)due to futility.Press Release.May 20, 2013.

88.Kantarjian H, Thomas D, Jorgensen J, Kebriaei P, Jabbour E,Rytting M, et al.Results of inotuzumab ozogamicin, a CD22 monoclonal antibody, in refractory and relapsed acute lymphocytic leukemia.Cancer 2013;119:2728-2736.

89.Kreitman RJ, Tallman MS, Robak T, Coutre S, Wilson WH,Stetler-Stevenson M, et al.Phase I trial of anti-CD22 recombinant immunotoxin moxetumomab pasudotox (CAT-8015 or HA22) in patients with hairy cell leukemia.J Clin Oncol 2012;30:1822-1828.

90.Kreitman RJ, Arons E, Stetler-Stevenson M, Wilson WH,FitzGerald DJ, Pastan I.Pharmacokinetic analysis of response in hairy cell leukemia treated by anti-CD22 recombinant immunotoxin moxetumomab pasudotox.Blood 2013;122:2871.

91.Forsberg G, Skartved NJ, Wallén-Ohman M, Nyhlén HC, Behm K, Hedlund G, et al.Naptumomab estafenatox, an engineered antibody-superantigen fusion protein with low toxicity and reduced antigenicity.J Immunother 2010;33:492-499.

92.Borghaei H, Alpaugh K, Hedlund G, Forsberg G, Langer C,Rogatko A, et al.Phase I dose escalation, pharmacokinetic and pharmacodynamic study of naptumomab estafenatox alone in patients with advanced cancer and with docetaxel in patients with advanced non-small-cell lung cancer.J Clin Oncol 2009;27:4116-4123.

93.Kuenen B, Witteveen PO, Ruijter R, Giaccone G, Dontabhaktuni A, Fox F, et al.A phase I pharmacologic study of necitumumab(IMC-11F8), a fully human IgG1 monoclonal antibody directed against EGFR in patients with advanced solid malignancies.Clin Cancer Res 2010;16:1915-1923.

94.Lilly announces phase III necitumumab study meets primary endpoint of overall survival.Press Release.August 13, 2013.

95.Kuenen B, Witteveen PO, Ruijter R, Giaccone G, Dontabhaktuni A, Fox F, et al.A phase I pharmacologic study of necitumumab(IMC-11F8), a fully human IgG1 monoclonal antibody directed against EGFR in patients with advanced solid malignancies.Clin Cancer Res 2010;16:1915-1923.

96.Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA,Lesokhin AM, et al.Nivolumab plus ipilimumab in advanced melanoma.N Engl J Med 2013;369:122-133.

97.Xin Y, Jin D, Eppler S, Damico-Beyer LA, Joshi A, Davis JD, et al.Population pharmacokinetic analysis from phase I and phase II studies of the humanized monovalent antibody, onartuzumab(MetMAb), in patients with advanced solid tumors.J Clin Pharmacol 2013;53:1103-1111.

98.Neninger E, Díaz RM, de la Torre A, Rives R, Díaz A, Saurez G,et al.Active immunotherapy with 1E10 anti-idiotype vaccine in patients with small cell lung cancer: report of a phase I trial.Cancer Biol Ther 2007;6:145-150.

99.Gordon MS, Sweeney CS, Mendelson DS, Eckhardt SG,Anderson A, Beaupre DM, et al.Safety, pharmacokinetics, and pharmacodynamics of AMG 102, a fully human hepatocyte growth factor-neutralizing monoclonal antibody, in a first-in-human study of patients with advanced solid tumors.Clin Cancer Res 2010;16:699-710.