12
Research Article Tumor Static Concentration Curves in Combination Therapy Tim Cardilin, 1,2,8 Joachim Almquist, 1,3 Mats Jirstrand, 1 Alexandre Sostelly, 4,5 Christiane Amendt, 6 Samer El Bawab, 4 and Johan Gabrielsson 7 Received 24 June 2016; accepted 9 September 2016; published online 28 September 2016 Abstract. Combination therapies are widely accepted as a cornerstone for treatment of different cancer types. A tumor growth inhibition (TGI) model is developed for combinations of cetuximab and cisplatin obtained from xenograft mice. Unlike traditional TGI models, both natural cell growth and cell death are considered explicitly. The growth rate was estimated to 0.006 h 1 and the natural cell death to 0.0039 h 1 resulting in a tumor doubling time of 14 days. The tumor static concentrations (TSC) are predicted for each individual compound. When the compounds are given as single-agents, the required concentrations were computed to be 506 μg · mL 1 and 56 ng · mL 1 for cetuximab and cisplatin, respectively. A TSC curve is constructed for different combinations of the two drugs, which separates concentration combinations into regions of tumor shrinkage and tumor growth. The more concave the TSC curve is, the lower is the total exposure to test compounds necessary to achieve tumor regression. The TSC curve for cetuximab and cisplatin showed weak concavity. TSC values and TSC curves were estimated that predict tumor regression for 95% of the population by taking between-subject variability into account. The TSC concept is further discussed for different concentration-effect relationships and for combinations of three or more compounds. KEY WORDS: mixture dynamics; model-based drug development; oncology; pharmacokinetic- pharmacodynamic modeling; tumor xenograft. INTRODUCTION Combination therapy plays a signicant role in pharma- cotherapy (1). One important aspect of combination therapy is the potential for synergistic effects. Other benets com- pared to single-agent treatment include higher overall ef- cacy, lower exposure to the drugs and thereby improved safety, reduced toxicity, and lower risk of developing drug resistances (25). Establishing synergies, nding the most effective anti- cancer drug combinations, and optimizing dosing schedules are all challenging tasks. Mathematical modeling has proved to be a valuable tool that addresses these challenges and can be used to quantify and predict the impact of drug combina- tions on tumor growth dynamics (6,7). Different data-driven tumor models have been suggested (8,9). One of the most commonly applied experimental models is the tumor growth inhibition (TGI) model (1013). The TGI model balances model complexity with data availability. In particular, the TGI model maintains a semi- mechanistic foundation while having few enough system and drug parameters for the model to still be calibrated using experimental data. A theoretical foundation for the TGI model has also been established by deriving the model from basic probabilistic assumptions in both single-agent and combination therapy settings (14,15). The TGI model divides cancer cells into two categories: proliferating or damaged. The model consists of a main compartment representing the proliferating cancer cells and a number of damage compartments which cells irreversibly traverse before dying. The drug action of an anticancer compound is said to be cytostatic and/or cytotoxic. Cytostatic drug action acts on the proliferating cells by inhibiting proliferation (cell growth), whereas cytotoxic drug action stimulates cell death by triggering apoptosis. The TGI model 1 Fraunhofer-Chalmers Centre, Chalmers Science Park, Gothenburg, Sweden. 2 Department of Mathematical Sciences, Chalmers University of Technology and University of Gothenburg, Gothenburg, Sweden. 3 Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden. 4 Global Early Development-Quantitative Pharmacology and Drug Disposition, Quantitative Pharmacology, Merck, Darmstadt, Germany. 5 Present Address: Pharmaceutical Research and Early Development, Hoffmann-Le Roche, Basel, Switzerland. 6 Translation Innovation Platform Oncology, Merck, Darmstadt, Germany. 7 Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden. 8 To whom correspondence should be addressed. (e-mail: [email protected]) The AAPS Journal, Vol. 19, No. 2, March 2017 ( # 2016) DOI: 10.1208/s12248-016-9991-1 1550-7416/17/0200-0456/0 # 2016 The Author(s). This article is published with open access at Springerlink.com 456

Tumor Static Concentration Curves in Combination Therapy · Research Article Tumor Static Concentration Curves in Combination Therapy Tim Cardilin,1,2,8 Joachim Almquist,1,3 Mats

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Tumor Static Concentration Curves in Combination Therapy · Research Article Tumor Static Concentration Curves in Combination Therapy Tim Cardilin,1,2,8 Joachim Almquist,1,3 Mats

Research Article

Tumor Static Concentration Curves in Combination Therapy

Tim Cardilin,1,2,8 Joachim Almquist,1,3 Mats Jirstrand,1 Alexandre Sostelly,4,5 Christiane Amendt,6

Samer El Bawab,4 and Johan Gabrielsson7

Received 24 June 2016; accepted 9 September 2016; published online 28 September 2016

Abstract. Combination therapies are widely accepted as a cornerstone for treatment ofdifferent cancer types. A tumor growth inhibition (TGI) model is developed for combinationsof cetuximab and cisplatin obtained from xenograft mice. Unlike traditional TGI models,both natural cell growth and cell death are considered explicitly. The growth rate wasestimated to 0.006 h−1 and the natural cell death to 0.0039 h−1 resulting in a tumor doublingtime of 14 days. The tumor static concentrations (TSC) are predicted for each individualcompound. When the compounds are given as single-agents, the required concentrationswere computed to be 506 μg · mL−1 and 56 ng · mL−1 for cetuximab and cisplatin,respectively. A TSC curve is constructed for different combinations of the two drugs, whichseparates concentration combinations into regions of tumor shrinkage and tumor growth.The more concave the TSC curve is, the lower is the total exposure to test compoundsnecessary to achieve tumor regression. The TSC curve for cetuximab and cisplatin showedweak concavity. TSC values and TSC curves were estimated that predict tumor regression for95% of the population by taking between-subject variability into account. The TSC conceptis further discussed for different concentration-effect relationships and for combinations ofthree or more compounds.

KEY WORDS: mixture dynamics; model-based drug development; oncology; pharmacokinetic-pharmacodynamic modeling; tumor xenograft.

INTRODUCTION

Combination therapy plays a significant role in pharma-cotherapy (1). One important aspect of combination therapyis the potential for synergistic effects. Other benefits com-pared to single-agent treatment include higher overall effi-cacy, lower exposure to the drugs and thereby improvedsafety, reduced toxicity, and lower risk of developing drugresistances (2–5).

Establishing synergies, finding the most effective anti-cancer drug combinations, and optimizing dosing schedulesare all challenging tasks. Mathematical modeling has provedto be a valuable tool that addresses these challenges and canbe used to quantify and predict the impact of drug combina-tions on tumor growth dynamics (6,7).

Different data-driven tumor models have been suggested(8,9). One of the most commonly applied experimentalmodels is the tumor growth inhibition (TGI) model (10–13).The TGI model balances model complexity with dataavailability. In particular, the TGI model maintains a semi-mechanistic foundation while having few enough system anddrug parameters for the model to still be calibrated usingexperimental data. A theoretical foundation for the TGImodel has also been established by deriving the model frombasic probabilistic assumptions in both single-agent andcombination therapy settings (14,15).

The TGI model divides cancer cells into two categories:proliferating or damaged. The model consists of a maincompartment representing the proliferating cancer cells and anumber of damage compartments which cells irreversiblytraverse before dying. The drug action of an anticancercompound is said to be cytostatic and/or cytotoxic. Cytostaticdrug action acts on the proliferating cells by inhibitingproliferation (cell growth), whereas cytotoxic drug actionstimulates cell death by triggering apoptosis. The TGI model

1 Fraunhofer-Chalmers Centre, Chalmers Science Park, Gothenburg,Sweden.

2 Department of Mathematical Sciences, Chalmers University ofTechnology and University of Gothenburg, Gothenburg, Sweden.

3 Department of Biology and Biological Engineering, ChalmersUniversity of Technology, Gothenburg, Sweden.

4 Global Early Development-Quantitative Pharmacology and DrugDisposition, Quantitative Pharmacology, Merck, Darmstadt,Germany.

5Present Address: Pharmaceutical Research and Early Development,Hoffmann-Le Roche, Basel, Switzerland.

6 Translation Innovation Platform Oncology, Merck, Darmstadt,Germany.

7 Department of Biomedical Sciences and Veterinary Public Health,Swedish University of Agricultural Sciences, Uppsala, Sweden.

8 To whom correspondence should be addressed. (e-mail:[email protected])

The AAPS Journal, Vol. 19, No. 2, March 2017 (# 2016)DOI: 10.1208/s12248-016-9991-1

1550-7416/17/0200-0456/0 # 2016 The Author(s). This article is published with open access at Springerlink.com 456

Page 2: Tumor Static Concentration Curves in Combination Therapy · Research Article Tumor Static Concentration Curves in Combination Therapy Tim Cardilin,1,2,8 Joachim Almquist,1,3 Mats

can be generalized to combination therapy with two or morecytostatic and/or cytotoxic compounds. There have beenseveral reported successes of this (16–20).

Several general methods have been established toevaluate drug combinations (21–24). A graphical tool is theisobologram (25–28). The isobologram is based on thesimplified model of a direct relationship between drug doseand a biomarker response and generates a Bcurve ofadditivity^ which can be used to distinguish additivity fromsynergy. A typical isobologram is shown in Fig. 1. Althoughdepicted as such here, isobolographic curve of additivity neednot be a straight line (29). The isobologram does not take intoaccount the drug response, nor cancer-specific informationthat a TGI model can provide.

A TGI model is developed for individual exposure tocombinations of cetuximab and cisplatin for the treatment of non-small cell lung cancer. These compounds are clinically relevant andhave been tested together on various cell lines aswell as in the clinic(30,31). Themodel assumes that the compounds act independentlyof each another. The model is an extension of the standard TGImodels in that it incorporates natural cell death. A consequence ofthis is that the initial tumor volume should be distributedappropriately among the main and damage compartments. Themodel is mechanistically attractive, since it discriminates betweennatural cell death and increased kill rate by means of chemicalintervention. We further investigate conditions for tumor stasisbased on the model equations for both the single-agent treatmentsand the combination therapy. This results in the derivation of whatis defined as the tumor static concentration (TSC) value and, forcombinations of two compounds, the tumor static concentrationcurve. The TSC value of a single compound is the plasmaconcentration of that compound required to maintain the tumorin stasis. Such values have been obtained for TGI models (12,32).The TSC curve is a generalization of the TSC value when twocompounds are given together (33,34). Instead of a single TSC

value, the TSC curve consists of all concentration pairs which givetumor stasis when present simultaneously.Moreover, we showhowthe curvature of the TSC profile has a large impact on the resultingtumorgrowth and total exposure to test compounds. In particular, ahighly concave1 (i.e., curving downward) TSC curve will allow fortumor regression even with a reduced total drug exposure.Additionally, by taking between-subject variability into account,we show that it is possible to obtain TSC curves for specificindividuals from the experimental data. This also makes it possibleto construct a TSC curve of concentration pairs for which a largepercentage (e.g., 95%)of the population is predicted to show tumorregression.

METHODS

Experimental Data

Patient-derived xenograft data consisted of 40 tumor-bearing female nude-nu mice used in a combination therapyexperiment with the anticancer compounds cetuximab andcisplatin to treat non-small cell lung cancer. These data havepreviously been published by Amendt et al. (35).

The 40 mice were divided evenly into four treatment arms:vehicle, single-agent treatment with cetuximab, single-agent treat-ment with cisplatin, and combination treatment with bothcetuximab and cisplatin. Treatment was started when tumorsbecame palpable (with sizes around 80–200mm3). All treatmentarms were given intraperitoneal doses on days 0, 3, 7, 10, and 14and the doses were always 30mg/kg for cetuximab and 5mg/kg forcisplatin. The mice in the combination arm were given doses ofboth drugs on each day of dosing. Tumors were measured bycaliper on days 0, 3, 7, 10, 14, 17, 21, 24, and 28.

Drug Exposure Models

Since there were no available exposure data for eithercetuximab or cisplatin, pharmacokinetic models were taken fromliterature for both compounds. To describe cetuximab exposure, aone-compartment pharmacokinetic model with first-order absorp-tion kinetics was used, with the explicit solution

Ccetuximab ¼ kaFDV ka−keð Þ e−ket−e−kat

� � ð1Þ

where ka is the absorption rate, ke the elimination rate, D thedose of cetuximab, F the bioavailability, and V the distribu-tion volume (36). This model is shown in Fig. 2 (left).

To describe total cisplatin exposure, a two-compartmentpharmacokinetic model with first-order absorption kinetics wasused. Themodel is described by the following systemof differentialequations (37). The gut compartment is

dAg

dt¼ −kaAg ð2Þ

Drug in the central (plasma) and peripheral (tissue)compartments then become

Antagonism

Synergy

0.0 0.2 0.4 0.6 0.8 1.00.0

0.2

0.4

0.6

0.8

1.0

DoseA (mg/kg)

Dos

e B(m

g/kg

)

Fig. 1. The isobolographic curve of additivity (blue). An experimen-tally observed effect above or below this curve is indicative of asynergistic or antagonistic relationship between the two drugs,respectively 1 Although mathematically the curve is said to be convex

457Tumor Static Concentration Curves

Page 3: Tumor Static Concentration Curves in Combination Therapy · Research Article Tumor Static Concentration Curves in Combination Therapy Tim Cardilin,1,2,8 Joachim Almquist,1,3 Mats

dAp

dt¼ kaFAg− k10 þ k12ð ÞAp þ k21At

dAt

dt¼ k12Ap−k21At

ð3Þ

The initial conditions of the gut, central, and peripheralcompartments are

Ag 0ð Þ ¼ D; Ap 0ð Þ ¼ 0; At 0ð Þ ¼ 0 ð4Þ

where ka is the absorption rate, D the dose, F the bioavailability,and k10, k12, and k21, the microconstants. Here, Ag, Ap, and At

denote the amount of cisplatin in the gut, plasma and tissue,respectively. An illustration of this model is shown in Fig. 2 (right).The total plasma concentration of cisplatin is then given by

Ccisplatin ¼ Ap

VCð5Þ

where VC denotes the volume of the central compartment.

Tumor Growth Inhibition Model

A TGI model with cetuximab as a cytostatic andcisplatin as a cytotoxic drug was used to describe the datain the final model. Modeling of each drug as cytostatic/

cytotoxic was tested using both linear and nonlinearfunctions. The final selection was made based on AIC.Drug action was assumed to be independent. An illustra-tion of this model is given in Fig. 3. The correspondingsystem of differential equations is

dV1

dt¼ I Ccetuximabð ÞkgV1−S Ccisplatin

� �kkV1

dV4

dt¼ kkV3−kkV4

ð6Þ

Here V1 is the main compartment of proliferatingcancer cells, V2, V3, and V4 are the damage compartmentsthat cells must go through before dying, kg is the growthrate, and kk is the (natural) death rate. The inhibitory drugaction, I, of cetuximab was described using an Emax-functionand the stimulatory drug action, S, of cisplatin wasdescribed using a linear function

I Ccetuximabð Þ ¼ 1−ImaxCcetuximab

IC50 þ CcetuximabS Ccisplatin� � ¼ 1 þ b Ccisplatin

ð7Þ

where Imax is the maximum inhibition cetuximab can accom-plish, IC50 is the concentration of cetuximab required toobtain 50% of the maximum inhibition, and b is a pharma-codynamic parameter of cisplatin. The total tumor volume

Fig. 2. (left) Pharmacokinetic model for cetuximab with gut and plasma compartments. The parameters ka and ke represent the drugabsorption and elimination rates, respectively. (right) pharmacokinetic model for cisplatin with gut, plasma, and tissue compartments. Theparameter ka represents the absorption rate and k12, k21, and k10 are the microconstants

ExposureA

Drug A Drug B

kk kkkkkkDamagedCells

CyclingCells

DamagedCells

DamagedCells

kg

ExposureB

Fig. 3. TGI model for combination therapy with independent drug action. Cetuximab (A) inhibits cell proliferation, whereas cisplatin(B) stimulates cell apoptosis. The kg and kk parameters represent the natural cell kill and death rates, respectively

458 Cardilin et al.

Page 4: Tumor Static Concentration Curves in Combination Therapy · Research Article Tumor Static Concentration Curves in Combination Therapy Tim Cardilin,1,2,8 Joachim Almquist,1,3 Mats

Vtotal comprises both V1 and the compartments representingdying cells.

Vtotal ¼ V1 þ V2 þ V3 þ V4 ð8Þ

Initial Conditions for the TGI Model

An important question is how to distribute the initial tumorvolume among the different compartments V1, V2,V3, and V4.Without natural cell death, the usual thing to do is to put all of theinitial volume into the main compartment V1. However, withnatural cell death included, this would neglect that cells have beendying since before treatment was started and one would thereforeexpect some cells to be in V2 to V4.

It is possible to show that, starting from any distribution ofthe total initial tumor volume into V1 and V2 to V4, the ratiobetween two neighboring compartments, (i.e., Vi/Vi − 1) willapproach a constant. If one assumes that the tumor has alreadyexisted for a long time before treatment started, it would makesense to also assume that this stage has been reached. Therefore,a reasonable choice of initial conditions is given by

V1 0ð Þ ¼ V0; V2 0ð Þ ¼ V0 kkkg

� �; V3 0ð Þ ¼ V0 kk

kg

� �2

; V4 0ð Þ

¼ V0 kkkg

� �3

ð9Þ

where V0 is the initial volume of V1. For a detailed derivationof the initial conditions, see Appendix 1.

Deriving Tumor Static Concentrations

Consider the individuals treated with cetuximab alone.Since tumor regression is often a treatment goal, we wouldlike to determine plasma concentrations resulting in tumorshrinkage. This is done by determining the drug concentra-tion for which the input and output of each compartment is inbalance. Plasma concentrations above this level may then beassumed to give tumor shrinkage.2 For the cetuximabtreatment, this means that each right-hand side in Eq. 6should be zero, but since V2 to V4 only act to delay the deathprocess, we only need to consider V1. For single-agenttreatment with cetuximab it must hold

kg 1−ImaxCcetuximab

IC50 þ Ccetuximab

� �−kk

� �V1 ¼ 0 ð10Þ

which implies that

kg 1−ImaxCcetuximab

IC50 þ Ccetuximab

� �−kk ¼ 0 ð11Þ

since the volume of the main compartment V1 will not be zerounless the tumor is already eradicated. Equation 11 can be solvedfor Ccetuximab to obtain

Ccetuximab ¼ IC50 kg−kk� �

kk− 1−Imaxð Þkg ð12Þ

This concentration is defined as the tumor static concentration(TSC) value of cetuximab. The TSC value holds for any tumorvolume and at any point in time. Therefore, to ensure tumorshrinkage over time, plasma exposure above TSC should beestablished. ATSC value for cisplatin can be derived in a similarmanner. When Ccetuximab = 0 the condition for stasis becomes

kg−kk 1þ bCcisplatin� � ¼ 0 ð13Þ

which can be simplified to

Ccisplatin ¼ kg−kkb kk

ð14Þ

Tumor Static Concentrations for Combinations

An analysis similar to that for the single-agent compounds canbe performed for the treatment arm that receives both compounds.For V1, it must therefore hold that

kg 1−ImaxCcetuximab

IC50 þ Ccetuximab

� �−kk 1þ bCcisplatin

� � ¼ 0 ð15Þ

This equation describes a curve in the concentration planewith Ccetuximab and Ccisplatin along the coordinate axes. In this caseone can solve for Ccisplatin to obtain

Ccisplatin ¼ kgb kk

1−ImaxCcetuximab

IC50 þ Ccetuximab

� �−1b

ð16Þ

Equation 16 describes a curve of concentration pairs(Ccetuximab, Ccisplatin). Any concentration pairs located abovethis curve will give tumor shrinkage, whereas concentrationpairs located below the curve will give tumor growth. Equa-tion 16 is therefore introduced as the TSC curve. The points ofintersection with the coordinate axes are the TSC values derivedfor the individual compounds. The TSC curve is valid for anytumor volume and at any time point. To ensure tumor shrinkageover time, dosing should be such that the concentration pair(Ccetuximab, Ccisplatin) is kept above the TSC curve at all times. Anillustration of what a TSC curve could look like is given in Fig. 4.How the TSC concept is generalized to three or morecompounds is discussed in Appendix 2.

Computational Methods

Nonlinear mixed-effects modeling of the cetuximab andcisplatin pharmacodynamic data was performed with a first-orderconditional estimation (FOCE) method, using a computational

2 The assumption that drug effect increases with concen-tration is true for most concentration-effect relationships

459Tumor Static Concentration Curves

Page 5: Tumor Static Concentration Curves in Combination Therapy · Research Article Tumor Static Concentration Curves in Combination Therapy Tim Cardilin,1,2,8 Joachim Almquist,1,3 Mats

framework developed at the Fraunhofer-Chalmers ResearchCentre for Industrial Mathematics (Gothenburg, Sweden) andimplemented in Mathematica 10 (Wolfram Research) (38). Themodel was evaluated based on individual fit, residual analysis,visualizations of Empirical Bayes Estimates, and the Akaikeinformation criterion.

To validate the assumption of independent drug action, aseparate set of parameter estimates were obtained by omitting thecombination arm. Then, a visual predictive check (VPC) wasperformed by using these estimates to simulate n = 5 000individuals given the combination treatment and comparing the5th and 95th quantiles to the actual observations.

To generate a TSC curve for which 95% of the population isexpected to show tumor regression, a Monte Carlo simulation wasperformed. The between-subject variability information containedin the covariance matrix Ω was used to simulate TSC curves for1000 hypothetical individuals. The 95% TSC curve was thenselected as the 95th percentile of the simulated curves.

RESULTS

Drug Exposure Models

Parameter estimates for the pharmacokinetic models ofcetuximab and cisplatin were taken from the literature (36,37).

The parameter values used for the cetuximab model (Eq. 1)were ke= 0.017 h− 1, ka= 0.44 h− 1, F= 1, and V=0.094 mL · kg−1.The corresponding exposure profile is shown in Fig. 5 (left).

The parameter values used for the cisplatin model (Eqs. 2–4)were ka= 42 h− 1, k10 = 1.3 h− 1, k12 = 3.3 h− 1, k21 = 1.12 h− 1, F= 1,and Vp= 377 mL ⋅ kg− 1. The corresponding exposure profile isshown in Fig. 5 (right).

Tumor Growth Inhibition Model

A population pharmacodynamic model was constructed byletting the parameters kg and V0 have lognormally distributedbetween-subject variability. A sample of the individual fit for eachtreatment arm is given in Fig. 6. Visual predictive checks for all fortreatment arms can be found in Appendix 3.

The parameter estimates for the tumor model are shown inTable I. The estimated net growth rate is given by kg− kk=0.0021 h− 1 corresponding to a doubling time of 330 h or 14 days.When the estimated volume of themain compartment of 60mm3 isused, the total initial volume is computed to be

Vtotal ¼ 1þ kkkg

� �þ kk

kg

� �2

þ kkkg

� �3 !

V0 ð17Þ

which gives an estimate of 140 mm3.The parameter value of Imax was set to 1 in the final model.

The estimated IC50 value for cetuximab of 994 μg ·mL−1 may becompared to the maximum exposure of 400 μg ·mL−1 whichcorresponds to roughly 30% inhibition of cell proliferation. Theestimated pharmacodynamics parameter of cisplatin is 0.0093mL ⋅ng− 1. and could be compared to themaximumcisplatin exposure of104 ng ·mL−1 which increases the kill rate by a factor of 100,although such levels of exposure only occurred during very shorttime periods (see Fig. 5). The model assumption of independentdrug action was validated by a visual predictive check for thecombination arm (see Appendix 4).

Tumor Shrinkage

Tumor Growth

0.0 0.2 0.4 0.6 0.8 1.00.0

0.2

0.4

0.6

0.8

1.0

CA (ng/ml)

CB

(ng/

ml)

Fig. 4. Schematic of a TSC curve (solid curve) for combinationtherapy with two compounds A and B. Reference straight line(dashed). Concentration pairs (CA,CB) above the curve (greenshaded area) give tumor shrinkage, whereas concentration pairsbelow (red shaded area) give tumor growth

C C

Fig. 5. (left) Drug exposure for cetuximab. The orange line indicates the IC50 estimate for cetuximab, and the green line the estimated TSCvalue for cetuximab. (right) Drug exposure for cisplatin. The orange line indicates the estimated plasma concentration required to double thedeath rate (given by 1/b), and the green curve the TSC value for cisplatin. Dosing was repeated for both compounds on days 0, 3, 7, 10, and 14

460 Cardilin et al.

Page 6: Tumor Static Concentration Curves in Combination Therapy · Research Article Tumor Static Concentration Curves in Combination Therapy Tim Cardilin,1,2,8 Joachim Almquist,1,3 Mats

TSC Curve of Cetuximab-Cisplatin

Recall that from the tumor model, we derived Eq. 16 forthe TSC curve. Using the parameter estimates from Table I,the TSC curve was computed, as depicted in Fig. 7. Theindividual TSC values were computed to be TSCCetuximab =506 μg · mL−1 and TSCcisplatin = 56 ng · mL−1. The TSC curveshows a slight deviation from a straight line. Individual TSCcurves were constructed by taking between-subject variabilityinto account. These are shown in blue in Fig. 8. The dashedred TSC curve, obtained by Monte Carlo simulation,indicates expected concentrations required for 95% of thepopulation to experience tumor shrinkage.

Four Cases of TSC Curves for Independent Drug Action

While there are many ways to incorporate drug interac-tion into a tumor growth model, the number of models withindependent action is considerably smaller and therefore

easier to investigate. Consider the tumor model in Eq. 6 withtwo generic compounds A and B. The correspondingequation for the TSC curve reads

I CAð Þkg−S CBð Þkk ¼ 0 ð18Þ

We shall consider four cases depending on how I(CA)and S(CB) are chosen. In all four cases, the TSC equation canbe written in the form

αCACB þ βCA þ γCB ¼ δ ð19Þ

where α, β, γ, and δ are constants which depend on both the drugand the system parameters. The individual TSC values can then beexpressed as

TSCA ¼ δβ; TSCB ¼ δ

γð20Þ

Linear Inhibition and Linear Stimulation

When both the cytostatic drug action and the cytotoxicdrug action are assumed linear

I CAð Þ ¼ 1−a CA; S CBð Þ ¼ 1þ bCB ð21Þ

the resulting TSC curve will also be linear and can berearranged to

kga CA þ kkb CB ¼ kg−kk ð22Þ

which is a straight line passing through the individual TSCvalues.

Nonlinear Inhibition and Linear Stimulation

In the second scenario, the cytostatic drug action is of Emax-type with unit sigmoidicity and the cytotoxic drug action is linear

Vehicle

Combination

Cetuximab

Cisplatin

0 5 10 15 20 250

200

400

600

800

1000

Time (days)

Tum

orvo

lum

e(m

m3)

Fig. 6. Representative time courses of observed (symbols) tumorvolumes (mm3) for each of the four treatment arms. The solid linesrepresent the tumor volumes fitted by the TGI model

Table I. Parameter Estimates for the Model Describing the Cetuximab and Cisplatin Combination Therapy

Parameter Population median (RSE%) Between-subject variabilitya (RSE%)

kg(h− 1) 0.0060 (11) 0.10 (23)

kk(h− 1) 0.0039 (17) –

V0(mm3) 60 (8) 0.33 (12)IC50(µg ⋅mL− 1) 994 (42) –Imax 1 (fixed) –b (mL ⋅ ng− 1) 0.0093 (30) –σb 27 (4) –cov(kg, V

0) 0.13 (26) –cor(kg, V

0) 0.48 –

aCalculated as 100�ffiffiffiffiffiffiω2ii

qb Intra-individual variability

461Tumor Static Concentration Curves

Page 7: Tumor Static Concentration Curves in Combination Therapy · Research Article Tumor Static Concentration Curves in Combination Therapy Tim Cardilin,1,2,8 Joachim Almquist,1,3 Mats

I CAð Þ ¼ 1−ImaxCA

IC50 þ CA; S CBð Þ ¼ 1þ b CB ð23Þ

The TSC equation can be shown to be

kkbCACB þ kk− 1−Imaxð Þkg� �

CA þ kkbIC50CB

¼ IC50 kg−kk� � ð24Þ

The first term is nonlinear in CA and CB and will providecurvature to the TSC curve. This case was illustrated in thecetuximab-cisplatin example above.

Linear Inhibition and Nonlinear Stimulation

The third case has the roles reversed—the cytostatic drugaction is now linear, while the cytotoxic drug action is ofEmax-type with unit sigmoidicity

S CAð Þ ¼ 1−a CA; S CBð Þ ¼ 1þ SmaxCB

SC50 þ CBð25Þ

The corresponding TSC equation becomes

kga CACB þ kgaSC50CA þ kg− 1þ Smaxð Þkk� �

CB

¼ SC50 kg−kk� � ð26Þ

Nonlinear Inhibition and Nonlinear Stimulation

Lastly, both the cytostatic drug action and the cytotoxic drugaction are assumed to be nonlinear of Emax-type with unitsigmoidicity

S CAð Þ ¼ 1−ImaxCA

IC50 þ CA; S CBð Þ ¼ 1þ SmaxCB

SC50 þ CBð27Þ

The corresponding TSC equation is similar to the previoustwo cases, albeit with slightly more complicated terms

1þ Smaxð Þkk− 1−Imaxð Þkg� �

CACB

þ SC50 kk− 1−Imaxð Þkg� �

CA þ IC50 1þ Smaxð Þkk−kg� �

CB

¼ IC50SC50 kg−kk� � ð28Þ

Impact of the TSC Curve on Tumor Growth

To illustrate the impact of different TSC curves, twohypothetical curves with the same individual TSC values butdifferent curvatureswere constructed based on the nonlinear-linearTSC equation (Eq. 24). The two curves are depicted in Fig. 9 (left)with their corresponding parameter values listed inTable II. Tumorvolume time courses were simulated assuming a constant plasmaconcentration of each drug at three different levels: (A) 0.3 μg ·mL−1, (B) 0.4 μg ·mL−1, and (C) 0.5 μg ·mL−1. The different timecourses are shown in Fig. 9 (right).As expected, the tumor volumesare much lower for the blue curves than for the red, since thecorresponding blue TSC curve exhibits much larger curvature thanits red counterpart.

Tumor Shrinkage

Tumor Growth

0 100 200 300 400 5000

10

20

30

40

50

60

70

CCetuximab ( g/mL)

CC

ispl

atin

(ng/

mL)

Fig. 7. TSC curve for cetuximab-cisplatin combinations (blue, bold)distinguishing between regions of tumor growth (light red) and tumorshrinkage (light green). Reference straight line connecting theindividual TSC values (black, dashed)

C

C

Fig. 8. Individual TSC curves (blue) for the cetuximab-cisplatin combina-tion therapy, taking into account between-subject variability in the growthrate kg. The TSC curve for the median individual from Fig. 7 is shown as asolid red curve. The dashed red line represents a region below which 95%ofthe population is expected to show tumor regression

462 Cardilin et al.

Page 8: Tumor Static Concentration Curves in Combination Therapy · Research Article Tumor Static Concentration Curves in Combination Therapy Tim Cardilin,1,2,8 Joachim Almquist,1,3 Mats

DISCUSSION

Drug Exposure Models

Since there were no available exposure data for eithercetuximab or cisplatin, models were taken from literature.For this reason, relatively simple models were chosen to givea rough estimate of what the exposure could have lookedlike. A disadvantage with this approach is that all individualsare assumed to have identical exposure profiles; there is nobetween-subject variation. This could lead to situations whereactual variability in kinetic parameters is falsely attributed tovariations in dynamic parameters, e.g., a large-growing tumorthat was due to low drug exposure, could instead be falselyexplained by the model with an unreasonably large growthrate parameter or low potency drug.

Tumor Growth Inhibition Model

A TGI model with independent drug action betweencetuximab and cisplatin proved sufficient to explain the combina-tion arm and no investigation into possible interaction terms wasnecessary. Since exposure profiles and models were taken solely

from the literature, an assumption of no interaction on exposurelevel was implied. All parameters in Table I except for IC50 wereestimated with acceptable precision and were located withinbiologically reasonable ranges. The relatively poor precision forIC50 is due to the estimated value of 994 μg ·mL−1 is well above themaximum drug exposure at around 400 μg ·mL−1 and themodel istherefore insensitive to changes in IC50. The between-individualvariation was large for both the growth rate and initial tumorvolume, which is due to the variability between subjects in thevehicle arm. It is also possible that some of the between-subjectvariability in growth rate is actually due to different levels ofepidermal growth factor receptor expression (EGFR), which hasbeen suggested to be associated with the drug effect of cetuximab(35). In the TGI model, individuals with a high EGFR expressionwould therefore be given a larger growth rate than thosewith a lowEGFRexpression. The parameter estimateswhen the combinationarmwas removed (Table III)were very similar to before, indicatingthat the combination arm is well-explained by the zero-interactionmodel. This is further validated from the visual predictive check inFig. 10 (Appendix 3)which shows that almost all observations fromthe combination arm fall within the estimated 90% confidenceintervals obtained using Table III.

The TGI model incorporates a natural cell death rate kkwhich was successfully estimated. However, it should be notedthat the vehicle model alone is not identifiable; one can onlydetermine the net growth rate kg − kk. However, with thepresence of a drug, these parameters can be separated andwere estimated with good precision. This is made possible by anunderlying assumption that the natural and drug-induced deathprocesses are equivalent in a modeling sense, which is consistentwith our viewpoint of cisplatin stimulating an already existingdeath process as opposed to induces a new one.

TSC Curve for Cetuximab-Cisplatin

The TSC curve is visually similar to the well-establishedisobologram (26,27), but with doses along the coordinate axes

Fig. 9. (left) Two different TSC curves (blue and red) and (right) the corresponding growth dynamics assuming a constant drug infusion ofeach drug at three concentration levels (A, B, and C)

Table II. Parameter Values Used to Construct the Two TSC Curvesin Fig. 9 (Blue and Red)

Parameter Value (blue) Value (red)

kg(h− 1) 0.0125 0.0125

kk(h− 1) 0.0042 0.0042

V0(mm3) 100 100IC50(µg ⋅mL− 1) 0.05 0.5Imax 0.7 1.0b (mL ⋅ ng− 1) 2.0 2.0

463Tumor Static Concentration Curves

Page 9: Tumor Static Concentration Curves in Combination Therapy · Research Article Tumor Static Concentration Curves in Combination Therapy Tim Cardilin,1,2,8 Joachim Almquist,1,3 Mats

replaced with drug plasma concentrations, and the consideredeffect is when the net growth rate is zero.While the isobologram istypically based on a dose-effect model ofEmax-type, the TSC curveis obtained from an exposure-driven growth model.

The TSC curve derived from cetuximab-cisplatin data servesas a practical example. The TSC curve exhibited a slight curvature,indicating that the drug combination effect is weakly synergistic.This is an interesting result given previous claims that EGFRinhibitors should act antagonistically with chemotherapy, althoughit is here limited to only one study of one specific combination (39).The certainty of the weak synergy is further explored inAppendix 5.

It is possible to derive TSC curves for more complicatedgrowth functions than the exponential one used here. TheGompertz, logistic, and Simeoni growth functions (9,12) all startwith an exponential growth phase which slows down as the tumor

grows large. ATSC curve can then be derived by assuming that thetumor maintains its initial exponential growth. This is a necessaryapproximation, since we want the concentration pairs above theTSCcurve to give tumor shrinkage for all tumor volumes, includingthose in the initial exponential phase.

The individual TSC curves in Fig. 8 show that there canbe significant variation in which concentrations will be neededfor tumor shrinkage. It is necessary to take between-subjectvariability into account and construct and simulate individualTSC curves. One can then target concentrations above theTSC curve for, e.g., 95% of the population (dashed, red).

In Fig. 8, the individual TSC curves are non-intersecting.This is not true in general and would typically only occur whenonly one parameter has population variability (in this case kg). Iftwo or more parameters have between-subject variability, someof the individual TSC curve would be expected to intersectunless the parameters are highly correlated.

Four Cases of TSC Curves for Independent Drug Action

We outlined four standard cases of independent drug actionbetween a cytotoxic drug and a cytostatic drug. The first importantobservation is that the case with linear inhibitory drug action andlinear stimulatory drug action is the only case where the TSC curvebecomes a straight line (signifiedbyα= 0) indicating additivity. Thisstraight line means that the concentration-effect relationship islinear even when the drugs are combined. There are no benefitsfrom combining the drugs. In contrast, when either the inhibition orstimulation is nonlinear, or both are nonlinear, the TSC curve willalso be a nonlinear curve falling below the straight line connectingthe individual TSC values. It will then be possible to decrease thetotal exposure levels of both compounds and still obtain tumorshrinkage.Evenwith twodrugswhichwhengiven individually eachrequire a concentration of 100μg ·mL−1 to be above theTSCvaluea sufficiently curved TSC curved may tell us that tumor shrinkagecan be obtained with a concentration of 25 μg ·mL−1 of each drugwhen given together. If the TSC curve were a straight line, therequired concentrations would instead have been 50 μg ·mL−1 ofeach drug.

None of the four cases studied allow for antagonism, or anoutward curving TSC curve. To allow for antagonism, the mostcommon way would be to include an explicit (negative) interactionterm added to the TGI model, which would contribute to making

Table III. Parameter Estimates for the Model Describing the Cetuximab and Cisplatin Combination Therapy, Using Only the Vehicle andSingle-Agent Treatment Arms

Parameter Population median (RSE%) Between-subject variability (RSE%)

kg(h− 1) 0.0065 (13) 0.10 (23)

kk(h− 1) 0.0044 (19) –

V0(mm3) 59 (8) 0.32 (9)IC50(µg ⋅mL− 1) 1139 (50) –Imax 1 (fixed) –b (mL ⋅ ng− 1) 0.0074 (31) –σa 30 (5) –cov(kg, V

0) 0.07 (27) –cor(kg, V

0) 0.50 –

a Intra-individual variability

0 5 10 15 20 250

50

100

150

200

250

300

Time (days)

Tum

orvo

lum

e(m

m3)

Fig. 10. A VPC for the combination arm using the parameterestimates from Table III. The solid lines indicate the simulated 5th,50th, and 95th percentiles. Colored dots represent different observedindividuals in the combination arm

464 Cardilin et al.

Page 10: Tumor Static Concentration Curves in Combination Therapy · Research Article Tumor Static Concentration Curves in Combination Therapy Tim Cardilin,1,2,8 Joachim Almquist,1,3 Mats

the parameter α in Eq. 19 negative and result in an antagonisticTSC curve.

Impact of the TSC Curve on Tumor Growth

We also investigated the relationship between the TSCcurve and tumor growth dynamics. We showed in Fig. 9 thateven if the individual TSC values are the same, the shape ofthe TSC curve is very important to how rapidly the tumorvolume changes. Since we have assumed that the drugs do notinteract, the different growth dynamics shown in Fig. 9 mustoriginate from the nonlinear concentration-response relation-ship of the inhibitory function I(CB). In particular, whatultimately decides why the blue TSC curve performs better ishow well the drugs perform at exposure levels well belowTSC. That the drugs have higher efficacy in the blue curvethan in the red curve at concentrations lower than the TSCvalue is translated into the blue TSC curve exhibiting largercurvature than the red.

Extensions and Future Work

Since we focused on introducing the TSC curve andproviding several examples under zero-interaction hypothe-ses, a natural next step would be to explore how the TSCcurve changes with the inclusion of (natural) synergy/antagonism terms. Synergy terms should increase the concav-ity of the TSC curve, while antagonism should have theopposite effect and make it more convex. Future work willalso need to explore the validity and utility of the TSCapproach, for example by using independent data sets.

CONCLUSIONS

The concept of a TSC curve for drug combinations intumor growth models is derived and applied to an in vivodataset. The TSC curve provides three important pieces ofinformation. First, it gives minimal concentration pairsneeded to achieve tumor shrinkage. Second, the shape ofthe TSC curve indicates how well the compounds work incombination, in the sense that a highly concave TSC curveindicates that it is possible to reduce the total drug exposureand still obtain tumor shrinkage. Third, the TSC curve can beused to optimize dosing regimens by maintaining concentra-tions above the curve as much and for as long as possible.

ACKNOWLEDGMENTS

Tim Cardilin was supported by an education Grant fromMerck.

Open Access This article is distributed under the termsof the Creative Commons Attribution 4.0 InternationalLicense (http://creativecommons.org/licenses/by/4.0/), whichpermits unrestricted use, distribution, and reproduction inany medium, provided you give appropriate credit to theoriginal author(s) and the source, provide a link to theCreative Commons license, and indicate if changes weremade.

APPENDIX 1

Consider the unperturbed tumor model incorporatingnatural cell death, with main compartment V1 and damagecompartments V1, …, Vn, described by the following systemof differential equations

dV1

dt¼kgV1−kkV1

dV2

dt¼kkV1−kkV2

dVi

dt¼ kkVi−1−kkVi; 3≤ i≤n

ð29Þ

This model can also be expressed in matrix form

dvdt

¼ Av ð30Þ

where v = (V1,…, Vn) is the n-dimensional vector of volumesand the n × n-matrix A is defined by

kg−kk 0 ⋯ 0 0kk −kk ⋯ 0 0⋮ ⋮0 0 ⋯ kk −kk

0BB@

1CCA ð31Þ

Since A is a triangular matrix, its eigenvalues can befound on the diagonal

λ1 ¼ kg−kk; λ2 ¼ λ3 ¼ … ¼ λn ¼ −kk ð32Þ

Fig. 11. A VPC for each of the four treatment arms using theparameter estimates from Table I. The solid lines indicate thesimulated 5th, 50th, and 95th percentiles. Colored dots representdifferent observed individuals in the combination arm

465Tumor Static Concentration Curves

Page 11: Tumor Static Concentration Curves in Combination Therapy · Research Article Tumor Static Concentration Curves in Combination Therapy Tim Cardilin,1,2,8 Joachim Almquist,1,3 Mats

The corresponding eigenvectors are given by

w1 ¼ 1;kkkg

;kkkg

� �2

;…;kkkg

� �3 !

; w2 ¼ 0;…; 0; 1ð Þ ð33Þ

The solution to Eq. 29 is then given by a linearcombination of the n independent solutions

w1eλ1t; w2eλ2t ð34Þand

eλ2 t tw2 þ w2;2� �

; eλ2tt2

2w2 þ tw2;2 þ w2;3

� �;…;

eλ2 ttn−1

n−1ð Þ! w2 þ…þ w2;n

� � ð35Þ

where the vectors w2,k are so-called generalized eigenvectorsof the matrix A, which for this particular problem can beshown to be

w2;2 ¼ 0; 1; 0; …; 0ð Þ; w2;3 ¼ 0; 0; 1; 0; …; 0ð Þ; …;w2;n

¼ 0; 0; 0; …; 1ð Þ ð36Þ

As t grows large, eλ2 t ¼ e−kkt goes to zero. Therefore, forsufficiently large t, the solution to Eq. 29 is described only by thefirst solution

v tð Þ ¼ w1e kg−kkð Þt ð37Þ

Hence, the tumor will grow exponentially with rate param-eter λ1 and a constant proportion between the compartmentsgoverned by w1.

APPENDIX 2

It is in theory possible to generalize the TSCconstruction to treatments with three or more compoundsby following the same steps. We know that a treatmentwith a single compound gives a point (the TSC value) inℝ and a combination of two compounds gives a curve (theTSC curve) in ℝ2. Likewise, a combination of threecompounds will give a surface (the TSC surface) in ℝ3,and, in general, a combination of n compounds will givean (n − 1)-dimensional hypersurface in ℝn.

To show one higher-dimensional example, imagine atreatment with n independent cytotoxic compounds withconcentrations C1,…, Cn and each with a linear stimulatoryfunction

Si Cið Þ ¼ aiCi; 1≤ i≤n ð38Þ

with different potency parameters ai. The differentialequation for the proliferating state V1 would in such asituation be

dV1

dt¼ kgV1−

Xni¼1

kkSi Cið ÞV1 ð39Þ

The condition for tumor stasis is therefore

kg−Xni¼1

kkSi Cið Þ ¼ 0 ð40Þ

In particular, for n = 3 this gives an equation on the form

a1C1 þ a2C2 þ a3C3 ¼ kgkk

ð41Þ

which can be recognized as the equation of a plane in ℝ3.

APPENDIX 3

Using the parameter estimates in Table I, a VPC wasperformed for each of the four treatment arms as shown inFig. 11. The figure indicates that all treatment arms wereadequately predicted by the model.

APPENDIX 4

To validate the assumption of independent drugaction, a separate set of parameter estimates wereobtained by omitting the combination arm (see Table III).Then, a VPC was performed by using these estimates tosimulate n = 5000 individuals given the combination treat-ment and comparing the 5th and 95th quantiles to theactual observations. The resulting VPC is shown in Fig. 10.With approximately 10% of the observations outside theshaded region, the assumption of independent drug actioncannot be rejected.

APPENDIX 5

To study the certainty of the weak synergy betweencetuximab and cisplatin, a Monte Carlo simulation wasperformed to construct TSC curves consistent with therelative standard errors reported for the parameters inTable I. To evaluate whether such a curve was sufficientlycurved, the ratio between the areas under the TSC curve andthe reference line was computed, yielding values ranging fromzero to one. If the ratio was below 0.9, the TSC curve wasregarded as sufficiently curved to indicate (at least) weaksynergy. A total of 99.4% of the simulated TSC curvesshowed weak synergy, indicating that the claim of weaksynergy is well-founded.

REFERENCES

1. DeVita Jr VT, Young RC, Canellos GP. Combination versussingle agent chemotherapy: a review of the basis for selection ofdrug treatment of cancer. Cancer. 1975;35(1):98–110.

2. Al-Lazikani B, Banerji U, Workman P. Combinatorial drugtherapy for cancer in the post-genomic era. Nat Biotechnol.2012;30(7):679–92. doi:10.1038/nbt.2284.

3. Komarova NL, Boland CR. Cancer: calculated treatment.Nature. 2013;499(7458):291–2. doi:10.1038/499291a.

466 Cardilin et al.

Page 12: Tumor Static Concentration Curves in Combination Therapy · Research Article Tumor Static Concentration Curves in Combination Therapy Tim Cardilin,1,2,8 Joachim Almquist,1,3 Mats

4. Nijenhuis CM, Haanen JB, Schellens JH, Beijnen JH. Iscombination therapy the next step to overcome resistance andreduce toxicities in melanoma? Cancer Treat Rev.2012;39(4):305–12. doi:10.1016/j.ctrv.2012.10.006.

5. Sharma P, Allison JP. Immune checkpoint targeting in cancertherapy: toward combination strategies with curative potential.Cell. 2015;161(2):205–14. doi:10.1016/j.cell.2015.03.030.

6. Anderson AR, Quaranta V. Integrative mathematical oncology.Nat Rev Cancer. 2008;8(3):227–34. doi:10.1038/nrc2329.

7. Bozic I, Reiter JG, Allen B, Antal T, Chatterjee K, Shah P, et al.Evolutionary dynamics of cancer in response to targetedcombination therapy. eLife. 2013;2:e00747. doi:10.7554/eLife.00747.

8. Ribba B, Holford NH, Magni P, Troconiz I, Gueorguieva I,Girard P, et al. A review of mixed-effects models of tumorgrowth and effects of anticancer drug treatment used inpopulation analysis. CPT Pharmacometrics Syst Pharmacol.2014;3:e113. doi:10.1038/psp.2014.12.

9. Sachs RK, Hlatky LR, Hahnfeldt P. Simple ODE models of tumorgrowth and anti-angiogenic or radiation treatment. Math ComputModel. 2001;33:1297–305. doi:10.1016/S0895-7177(00)00316-2.

10. Choo EF, Ng CM, Berry L, Belvin M, Lewin-Koh N, MerchantM, et al. PK-PD modeling of combination efficacy effect fromadministration of the MEK inhibitor GDC-0973 and PI3Kinhibitor GDC-0941 in A2058 xenografts. Cancer ChemotherPharmacol. 2013;71(1):133–43. doi:10.1007/s00280-012-1988-6.

11. Evans ND, Dimelow RJ, Yates JW. Modelling of tumour growthand cytotoxic effect of docetaxel in xenografts. Comput MethodsProg Biomed. 2014;114(3):e3–e13. doi:10.1016/j.cmpb.2013.06.014.

12. Simeoni M, Magni P, Cammia C, De Nicolao G, Croci V, PesentiE, et al. Predictive pharmacokinetic-pharmacodynamic modelingof tumor growth kinetics in xenograft models after administra-tion of anticancer agents. Cancer Res. 2004;64(3):1094–101.

13. Tate SC, Cai S, Ajamie RT, Burke T, Beckmann RP, Chan EM, et al.Semi-mechanistic pharmacokinetic/pharmacodynamic modeling ofantitumor activity of LY2835219, a new cyclin-dependent kinase 4/6inhibitor, in mice bearing human tumor xenografts. Clin Cancer Res.2014;20(14):3763–74. doi:10.1158/1078-0432.CCR-13-2846.

14. Magni P, GermaniM,DeNicolaoG, Bianchini G, SimeoniM, PoggesiI, et al. A minimal model of tumor growth inhibition. IEEE TransBiomed Eng. 2008;55(12):2683–90. doi:10.1109/TBME.2008.913420.

15. Magni P, Terranova N, Del Bene F, Germani M, De Nicolao G.A minimal model of tumor growth inhibition in combinationregimens under the hypothesis of no interaction between drugs.IEEE Trans Biomed Eng. 2012;59(8):2161–70. doi:10.1109/TBME.2012.2197680.

16. Goteti K, Garner CE, Utley L, Dai J, Ashwell S, Moustakas DT,et al. Preclinical pharmacokinetic/pharmacodynamic models topredict synergistic effects of co-administered anti-cancer agents.Cancer Chemother Pharmacol. 2010;66(2):245–54. doi:10.1007/s00280-009-1153-z.

17. Rocchetti M, Del Bene F, Germani M, Fiorentini F, Poggesi I,Pesenti E, et al. Testing additivity of anticancer agents in pre-clinical studies: a PK/PD modelling approach. Eur J Cancer.2009;45(18):3336–46. doi:10.1016/j.ejca.2009.09.025.

18. Rocchetti M, Germani M, Del Bene F, Poggesi I, Magni P,Pesenti E, et al. Predictive pharmacokinetic-pharmacodynamicmodeling of tumor growth after administration of an anti-angiogenic agent, bevacizumab, as single-agent and combinationtherapy in tumor xenografts. Cancer Chemother Pharmacol.2013;71(5):1147–57. doi:10.1007/s00280-013-2107-z.

19. Terranova N, Germani M, Del Bene F, Magni P. A predictivepharmacokinetic-pharmacodynamic model of tumor growthkinetics in xenograft mice after administration of anticanceragents given in combination. Cancer Chemother Pharmacol.2013;72(2):471–82. doi:10.1007/s00280-013-2208-8.

20. Koch G, Waltz A, Lahu G, Schropp J. Modeling of tumor growthand anticancer effects of combination therapy. J PharmacokinetPharmacodyn. 2009;36(2):179–97. doi:10.1007/s10928-009-9117-9.

21. Chou TC. Drug combination studies and their synergy quantifi-cation using the Chou-Talalay method. Cancer Res.2010;70(2):440–6. doi:10.1158/0008-5472.CAN-09-1947.

22. Foucquier J, Guedj M. Analysis of drug combinations: currentmethodological landscape. Pharmacol Res Perspect. 2015;3(3):e00149.doi:10.1002/prp2.149.

23. Tallarida RJ. Drug synergism: its detection and applications. JPharmacol Exp Ther. 2001;298(3):865–72.

24. Zhao L, Wientjes MG, Au JL. Evaluation of combinationchemotherapy: integration of nonlinear regression, curve shift,isobologram, and combination index analyses. Clin Cancer Res.2004;10(23):7994–8004.

25. Loewe S. Die Mischiarnei. Klin Wochenschr. 1927;6:1077–85.26. Loewe S. Die quantitativen probleme der pharmaqkologie.

Ergebn Physiol. 1928;27:47–187.27. Loewe S. The problem of synergism and antagonism of

combined drugs. Arzneimittelforschung. 1953;3(6):285–90.28. Tallarida RJ. An overview of drug combination analysis with

isobolograms. J Pharmacol Exp Ther. 2006;319(1):1–7.29. Grabovsky Y, Tallarida RJ. Isobolographic analysis for combi-

nations of a full and partial agonist: curved isoboles. J PharmacolExp Ther. 2004;310(3):981–6.

30. Prewett M, Rockwell P, Rose C, Goldstein NI. Anti-tumor andcell cycle responses in kb cells treated with a chimeric anti-EGFR monoclonal antibody in combination with cisplatin. Int JOncol. 1996;9(2):217–24.

31. Fan Z, Baselga J, Masui H, Mendelsohn J. Antitumor effect ofanti-epidermal growth factor receptor monoclonal antibodiesplus- cis-diamminedichloroplatinum on well established A431cell xenografts. Cancer Res. 1993;53(19):4637–42.

32. Jumbe NL, Xin Y, Leipold DD, Crocker L, Dugger D, Mai E,et al. Modeling the efficacy of trastuzumab-DM1, an antibodydrug conjugate, in mice. J Pharmacokinet Pharmacodyn.2010;37(3):221–42. doi:10.1007/s10928-010-9156-2.

33. Cardilin T, Sostelly A, Jirstrand M, Amendt C, El Bawab S,Gabrielsson J. Modelling and analysis of tumor growth inhibitionfor combination therapy using tumor static concentration curves.PAGE, Abstr 3568: 2015. www.page-meeting.org/?abstract=3568.

34. Gabrielsson J, Gibbons FD, Peletier LA. Mixture dynamics:combination therapy in oncology. Eur J Pharm Sci. 2016;88:132–46. doi:10.1016/j.ejps.2016.02.020.

35. Amendt C, Staub E, Friese-HamimM, Störkel S, Stroh C. Associationof EGFR expression level and cetuximab activity in patient-derivedxenograft models of human non-small cell lung cancer. Clin CancerRes. 2014;20(17):4478–87. doi:10.1158/1078-0432.CCR-13-3385.

36. Lou FR, Yang Z, Dong H, Camuso A, McGlinchey K, Fager K,et al. Prediction of active drug plasma concentrations achieved incancer patients by pharmacodynamic biomarkers identified fromthe geo human colon carcinoma xenograft model. Clin CancerRes. 2005;11(15):5558–65.

37. Johnsson A, Olsson C, Nygren O, Nilsson M, Seiving B,Cavallin-Ståhl E. Pharmacokinetics and tissue distribution ofcisplatin in nude mice: platinum levels and cisplatin DNA-adducts. Cancer Chemother Pharmacol. 1995;37(1–2):23–31.

38. Almquist J, Leander J, Jirstrand M. Using sensitivity equations forcomputing gradients of the FOCE and FOCEI approximations tothe population likelihood. J Pharmacokinet Pharmacodyn.2015;42(3):191–209. doi:10.1007/s10928-015-9409-1.

39. Davies AM, Ho C, Lara Jr PN, Mack P, Gumerlock PH,Gandara DR. Pharmacodynamic separation of epidermalgrowth factor receptor tyrosine kinase inhibitors and chemo-therapy in non-small cell lung cancer. Clin Lung Cancer.2006;7(6):385–8.

467Tumor Static Concentration Curves