Preclinical Development of an anti-5T4 Antibody ... - ACS Publications

Jul 16, 2015 - ABSTRACT: The pharmacokinetics of an antibody (huA1)−drug. (auristatin microtubule disrupting MMAF) conjugate, targeting. 5T4-express...
0 downloads 0 Views 2MB Size
Article pubs.acs.org/bc

Preclinical Development of an anti-5T4 Antibody−Drug Conjugate: Pharmacokinetics in Mice, Rats, and NHP and Tumor/Tissue Distribution in Mice Mauricio Leal,*,† JoAnn Wentland,‡ Xiaogang Han,‡ Yanhua Zhang,‡ Brian Rago,‡ Nicole Duriga,§ Franklin Spriggs,§ Eugene Kadar,‡ Wei Song,‡ James McNally,§ Quazi Shakey,§ Leslie Lorello,‡ Judy Lucas,∥ and Puja Sapra∥ †

Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Pearl River, New York 10965, United States Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut 06340, United States § Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Andover, Massachusetts 01810, United States ∥ Oncology Research Unit, Pfizer Inc., Pearl River, New York 10965, United States ‡

S Supporting Information *

ABSTRACT: The pharmacokinetics of an antibody (huA1)−drug (auristatin microtubule disrupting MMAF) conjugate, targeting 5T4-expressing cells, were characterized during the discovery and development phases in female nu/nu mice and cynomolgus monkeys after a single dose and in S-D rats and cynomolgus monkeys from multidose toxicity studies. Plasma/serum samples were analyzed using an ELISA-based method for antibody and conjugate (ADC) as well as for the released payload using an LCMS/MS method. In addition, the distribution of the Ab, ADC, and released payload (cys-mcMMAF) was determined in a number of tissues (tumor, lung, liver, kidney, and heart) in two tumor mouse models (H1975 and MDA-MB-361-DYT2 models) using similar LBA and LC-MS/MS methods. Tissue distribution studies revealed preferential tumor distribution of cys-mcMMAF and its relative specificity to the 5T4 target containing tissue (tumor). Single dose studies suggests lower CL values at the higher doses in mice, although a linear relationship was seen in cynomolgus monkeys at doses from 0.3 to 10 mg/kg with no evidence of TMDD. Evaluation of DAR (drug−antibody ratio) in cynomolgus monkeys (at 3 mg/kg) indicated that at least half of the payload was still on the ADC 1 to 2 weeks after IV dosing. After multiple doses, the huA1 and conjugate data in rats and monkeys indicate that exposure (AUC) increases with increasing dose in a linear fashion. Systemic exposure (as assessed by Cmax and AUC) of the released payload increased with increasing dose, although exposure was very low and its pharmacokinetics appeared to be formation rate limited. The incidence of ADA was generally low in rats and monkeys. We will discuss cross species comparison, relationships between the Ab, ADC, and released payload exposure after multiple dosing, and insights into the distribution of this ADC with a focus on experimental design as a way to address or bypass apparent obstacles and its integration into predictive models.



INTRODUCTION

Trophoblast glycoprotein (TPBG, also known as 5T4) is a cell surface antigen that internalizes rapidly and thus has the potential to efficiently deliver ADCs into tumor cells.2−4 Expression of 5T4 is associated with advanced disease and/or worse clinical outcome in NSCLC and gastric, colorectal, and ovarian carcinomas and other solid tumors.5−7 Together, these clinical observations suggest that 5T4 is an attractive target for ADC therapeutics.8 A number of investigators have described various aspects of the preclinical development of ADC’s from a

The basic strategy underlying antibody−drug conjugates (ADC) is to combine the exquisite target selectivity of monoclonal antibodies with the potent cytotoxic activity of certain natural products and/or synthetic molecules, with the goal of generating agents that are highly efficacious and also safe. In addition to the two recently approved ADC’s (ADCETRIS and KADCYLA), the ADC platform currently includes a growing repertoire of cytotoxic payloads, linker technologies, and conjugation methods with sufficient investigational new drug applications (INDs) for ADCs to allow the FDA to conduct a preliminary summary analysis of nonclinical development programs for ADCs.1 © XXXX American Chemical Society

Special Issue: Antibody-Drug Conjugates Received: April 17, 2015 Revised: July 3, 2015

A

DOI: 10.1021/acs.bioconjchem.5b00205 Bioconjugate Chem. XXXX, XXX, XXX−XXX

Article

Bioconjugate Chemistry

Figure 1. Total Ab, ADC, and released payload (cys-mcMMAF) concentration profiles in A1mcMMAF dosed animals are similar in tumor and nontumor bearing mice, and the released payload concentrations are very low. Data from the tumor bearing mice from Shah et al. (2014).14

Table 1. Pharmacokinetic Parameters (ADC and Released Payload, cys-mcMMAF) in A1mcMMAF Dosed Nude (nu/nu) Mice (Tumor and Nontumor Bearing) Cys-mcMMAFa

ADC model

dose (mg/kg)

Non-Tumor Tumor Non-Tumor Tumor

1 1 10 10

Cmax (μg/mL) 16.5 18.1 181 155

± ± ± ±

3.4 0.7 25 23

AUC0‑∞ (μg*h/mL) 483 564 7840 5850

± ± ± ±

89 139 2470 2320

CL (mL/h/kg) 2.12 1.86 1.42 1.95

± ± ± ±

0.35 0.43 0.58 0.81

t1/2 (day)

Cmax (ng/mL)

Tmax (hr)

AUC0−336 (ng*h/mL)

t1/2b (day)

± ± ± ±

0.192 0.146 0.951 0.945

8 8 8 8

14.6 28.2 58.5 81.9

2.1b 6.0b 3.0b 2.9b

3.5 3.5 3.5 3.3

0.9 1.1 0.9 1.0

a cys-mcMMAF data from pooled (group) samples. bEstimated half-life (not elimination terminal) assuming that the elimination of cys-mcMMAF is considered to be formation-rate limited rather than a reflection of its own clearance.

pharmacokinetics (PK) perspective.9−12 Here, we report the development of A1mcMMAF (an anti-5T4 humanized A1 antibody conjugated to the microtubule disrupting monomethylauristatin F (MMAF) via a maleimidocaproyl linker) from the pharmacokinetics perspective by comparing exposure across species in discovery PK (tumor and nontumor bearing mice) and definitive (rat and cynomolgus monkeys) PK/TK studies, including the profile of the average drug−antibody ratio (DAR) with time from a monkey PK study. In addition, we report a series of tumor distribution studies conducted in tumor-bearing (H1975 and MDAMB361-DYT2) mouse models to determine the distribution of the Ab, ADC, and the released payload (cys-mcMMAF) in a limited number of tissues and document the degree of tumor specificity by using various (negative Ab and tumor target) controls.

models, respectively) the exposures appeared to increase in an approximately proportional manner (Table 1). Peak concentrations of the released payload (cys-mcMMAF) was observed at 8 h after A1mcMMAF dosing and were very low (compared to that of the ADC), its concentration−time profile reflects its formation in plasma from the ADC across time, and its clearance (as reflected in relatively similar t1/2 values) is considered to be formation-rate limited rather than a reflection of its own clearance had cys-mcMMAF been dosed alone (Figure 1).13 Mouse Tumor Distribution Studies. While the observed plasma Ab, ADC, and released payload concentration profile in mice are typical for an ADC,12 we designed a series of tumor distribution studies to determine if the cytotoxic payload reaches the target tissue (tumor), and if so, is it in a target specific manner.14,15 After a 3 mg/kg dose of A1mcMMAF, peak concentrations of the released payload (cys-mcMMAF) in tumor homogenates were approximately 600 times higher than in plasma in both the MDAMB361-DYT2 and H1975 mouse models (0.16 and 0.20 μg/mL in tumor homogenates and 0.00029 and 0.00032 μg/mL in plasma, respectively) indicating efficient binding of the ADC to its 5T4 target on the tumor and subsequent conversion and release of the cytotoxic payload at the tumor target rather than in the systemic circulation (Figure 2a). When expressed on a molar basis (Figure 2b and c), a trend become apparent. The cys-mcMMAF concentrations (μM) in tumor trends in the same concentration range as the Ab or ADC concentration (μM) from plasma samples (a more typical and easier matrix to collect and evaluate) and something



RESULTS Pharmacokinetics in Tumor and Nontumor Mouse Models. The pharmacokinetics of A1mcMMAF in a high (++ +) expressing 5T4 nude mouse tumor model (MDA-MB-435/ 5T4) were similar to that in nontumor bearing mice given a single IV dose of 1 and 10 mg/kg with terminal elimination half-life (t1/2) values in tumor and nontumor bearing mice (3.5 days, 1 mg/kg; 3.3 and 3.5 days, 10 mg/kg; respectively) as seen in Figure 1. The ADC exhibited the typical low plasma clearance (CL; 1.4 to 2.1 mL/h/kg) and small steady-state volume of distribution (Vss; 127 to 153 mL/kg). Although there was a higher degree of variability at the 10 mg/kg dose (CV% for the AUC was 32% and 42% for the nontumor and tumor B

DOI: 10.1021/acs.bioconjchem.5b00205 Bioconjugate Chem. XXXX, XXX, XXX−XXX

Article

Bioconjugate Chemistry

Figure 2. Released payload (cys-mcMMAF) plasma and tumor concentrations in the H1975 and 361DYT2 mouse models after a single 3 mg/kg IV dose of A1mcMMAF. Released payload concentrations (μM) track well (as shown by the shaded areas) with the plasma Ab and ADC concentrations (μM). Data reconfigured from Shah et al. (2014).14

Figure 3. Comparison of Ab, ADC, and Cys-mcMMAF concentrations (ug/mL) in plasma, tumor, and other tissues after a 3 mg/kg IV dose (A1mcMMAF) to a H1975 mouse model. Plasma and tumor data from Shah et al. (2014).14

suggested that the ADC was effectively concentrating the cytotoxic payload in tissue bearing the target antigen (the tumor) (Figure 3) with concentrations of cys-mcMMAF in liver, lung, heart, and kidney in the low single digit ng/mL range and much lower than in tumor tissue. This was observed in both the H1975 and DTY2 mouse models (data not shown). When evaluating the degree of accumulation of the cytotoxic agent (released payload) in the various tissues, the %ID/g (% of injected dose per gram) tissue was higher in the tumor homogenate (4.1% in the H1975 model and 2.4% in the DYT2 model) with very low %ID/g (>0.3%) in the other tissues with the clearance organs (liver and kidney) having relatively higher values among them (Figure 4). This observation further suggests the specificity of the release of the cytotoxic payload

that could prove helpful if the same relationship holds in the clinic. Although this relationship (or trend) was very pronounced and tight in the DYT2 model (Figure 2b), it was also evident in the H1975 model (Figure 2c). A possible next step would be to determine if this relationship (trend) is also seen in the clinic and also with other ADC’s in mice (and in the clinic). The tumor distribution studies also included measurements of cys-mcMMAF in tissues representing the main clearance organs and highly perfused tissues (i.e., liver, lung, heart, and kidney tissue). In general, the results indicated a greater propensity for the ADC (and as a result, for the released payload to distribute to and be retained) in tumor tissue compared to the systemic circulation or in other tissues and C

DOI: 10.1021/acs.bioconjchem.5b00205 Bioconjugate Chem. XXXX, XXX, XXX−XXX

Article

Bioconjugate Chemistry

focused on evaluating the degree of cys-mcMMAF accumulation at the target (tumor). The first data set that suggested a difference in specificity was in the comparison of A1mcMMAF in H1975 or Ramos models. As depicted in Figure 5a, the concentrations of ADC in plasma (using a Ligand Binding Assay platform) were similar in both models but higher in tumor samples from the H1975 mice (compared to those in the Ramos model) indicating a greater degree of binding of A1mcMMAF to the positive tumor control (H1975) model. While the plasma concentrations of cys-mcMMAF were similar in the four different groups (Figure 5b) (suggesting similar nonspecific disassociation of the payload from the ADC while in circulation), the exposure (AUC, area under the curve) of cys-mcMMAF in tumor samples was higher in the positive ADC/target model group (A1mcMMAF in the H1975 model) than in the other groups (Figure 5c).16 Using the control groups (Ab and target/model) we were able to confirm the desired specificity of released payload delivery in H1975-tumor bearing mice given the anti-5T4 ADC. Pharmacokinetics in Cynomolgus Monkeys and DAR Evaluation. In the cynomolgus monkeys, a target expressing species, a low dose PK study was designed in order to evaluate the potential for target mediated drug disposition (TMDD). Following an IV administration of the A1mcMMAF (3 mg/kg) to cynomolgus monkeys, the Ab and ADC exhibited low mean systemic clearance (0.36 and 0.66 mL/h/kg, respectively), a small steady-state volume of distribution (ranging from 56 to 81 mL/kg) and terminal elimination half-life values of approximately 5.4 and 4.2 days, respectively. It is worthwhile noting that, at the 0.3 mg/kg dose, ADC concentrations (measured by an LBA assay) were detectable only until 168 h; thus, the estimates of CL and terminal half-life need to be considered with caution. As anticipated, the systemic exposure to the released payload was low compared with the

Figure 4. Comparison of %ID (for Cys-mcMMAF) in tumor and various tissues after a 3 mg/kg IV dose (A1mcMMAF) in the H1975 and DYT2 mouse models indicating preferential accumulation in the target (tumor) tissue.

into its intended target (the tumor) rather than nonspecifically into other tissues. The %ID/g values observed in tumor homogenates were higher than the low value (∼0.02%) that had been considered to be the case for tumor penetration for an antibody. Target Specificity in Mouse Models. For this program, our approach to evaluate target specificity (important with respect to confidence in patient selection) was to compare our A1mcMMAF under different controlled situations: using a negative antibody (neg 8.8 mAb, that does not bind to the 5T4 target) conjugated to the mcMMAF linker-payload and a tumor model that does not contain the target (the Ramos model) as variables. The followup comparison tumor distribution studies used A1mcMMAF (as the positive ADC) and neg8.8mcMMAF (as the negative ADC) each at a dose of 3 mg/kg on the H1975 (target postive) and Ramos (target negative) models and

Figure 5. Specificity of A1mcMMAF tumor targeting in the H1975 mouse modelcomparison of exposure using various target controls (+ or −mAb and tumor models). D

DOI: 10.1021/acs.bioconjchem.5b00205 Bioconjugate Chem. XXXX, XXX, XXX−XXX

Article

Bioconjugate Chemistry

which showed loss of DARs 2 and 3 (down to 0% by 168 h after dosing) (Supporting Information Figure 2). The fact that at least half of the ADC is loaded (∼average DAR of 2) during the second week of dosing is comforting if this were to be predictive of the clinical situation. Toxicokinetics of A1mcMMAF. The toxicokinetics of huA1, the conjugate (A1mcMMAF), and the released payload as well as the propensity for antidrug antibodies (ADA) to develop after multiple doses of A1mcMMAF were evaluated in rats and cynomolgus monkeys as part of the regulated toxicity studies. Doses of A1mcMMAF (6, 20, and 60 mg/kg every 2 weeks) to Sprague−Dawley rats resulted in systemic exposure after single (day 1) and multiple dosing (day 29, the third cycle) that increased with increasing dose and were comparable after the first dose as after multiple dosing although exposure was slightly higher for the Ab (given its generally longer half-life values, that was expected) on day 29 (Figure 8). Although it was not possible to properly evaluate linearity (mainly due to the limitations of using a staggered bleeding design in the rat study), the mean exposure at the high dose (60 mg/kg) increased in an apparently less than proportional manner. In general, the presence of antidrug antibodies (ADA) in individual animals (2 weeks after the day 29 dose was given) resulted in lower serum concentrations of Ab and ADC, most notably in the 6 mg/kg dose group, relative to the corresponding ADA-negative animals. However, the lower concentrations did not appear to impact overall exposure following multiple dosing. Overall, the incidence of induction of an ADA response was generally low (0%, 58.3%, 8.3%, and 16.7% at 0, 6, 20, and 60 mg/kg A1mc MMAF, respectively). Although a low incidence of positive ADA titers was noted in the 20 and 60 mg/kg dose groups, where concentrations of A1mcMMAF were much higher than in 6 mg/kg, it should be noted that circulating levels of A1mcMMAF may have interfered with the detection of the anti-A1mcMMAF antibodies. As observed in rats, doses of A1mcMMAF (1, 3, 9, and 15 mg/kg every 2 weeks) to cynomolgus monkeys resulted in systemic exposure (after single and multiple doses) that increased with increasing dose in an approximately dose proportional manner (Figure 8). In addition, the exposure was comparable after a single and multiple (day 29, third cycle) dosing with the same observation (as in rat) for the high dose (15 mg/kg) where there was a minor degree of accumulation of the Ab presumably due to its longer elimination half-life (10.4 days). In this study, an Ab alone group (at 15 mg/kg) was also used and resulted in an approximately 35% higher AUC than for Ab after the A1mcMMAF was given (Figure 8a). After multiple dosing, the ADC to Ab ratio was approximately 45% compared to 58% after the first dose; this can be attributed to the tendency for the antibody to accumulate to a small degree after multiple dosing due to its generally longer half-life (8−10 days) compared to that of the ADC (4.8 to 6.2 days). In general, the presence of antidrug antibodies (ADA) in monkeys was minimal with the incidence of ADA response observed in 1 animal each at the 1 and 3 mg/kg doses (and only at the 1 mg/kg dose were the concentrations affected such that the animal was excluded from the summary data). In general, the toxicokinetic data from these rat and monkey studies proved to be in line when compared to single dose data from mouse and monkey pharmacokinetic studies discussed earlier (Figure 8b) further implying a well behaved ADC in

concentrations of both the Ab and ADC and most likely reflects its release from the ADC (A1mcMMAF) over time. Overall exposure (AUC) was relatively linear from the low dose of 0.3 mg/kg to 3 mg/kg indicating that TMDD was not a concern in this target bearing species. As also observed in the mouse pk and distribution studies, and as anticipated based on historical data with other ADC’s, the exposure to the ADC was lower than that of the Ab. At the 3 mg/kg dose, the ADC accounted for an average of 51% of the Ab exposure, consistent with the mouse exposure data. As part of this study, we also evaluated the drug−antibody ratio (DAR) which is used to determine overall stability of the payload on the ADC and can be used to prospectively predict the same in humans.17 The evaluation of DAR was only possible at the 3 mg/kg dose due to lack of sensitivity at the lower dose. After a single 3 mg/kg dose of A1mcMMAF, there was a loss of drug (payload) from the ADC over time, and this observation was consistent between individual animals (Figure 6). On average, drug loading on the ADC changed from

Figure 6. Individual concentration profiles of Ab, ADC, and CysmcMMAF in cynomolgus monkeys after a single IV dose of 0.3 and 3 mg/kg.

approximately 4 to 2 over the 14-day period postdose (Figure 7). In the light chain, total drug decrease about 10% over time (Supporting Information Figure 1). This indicates that while the amount of drug in the light chain appeared stable over time the instability (or loss of DAR) came from the heavy chain

Figure 7. Evaluation of average DAR in individual cynomolgus monkeys after an IV dose (3 mg/kg) of A1mcMMAF. E

DOI: 10.1021/acs.bioconjchem.5b00205 Bioconjugate Chem. XXXX, XXX, XXX−XXX

Article

Bioconjugate Chemistry

Figure 8. Toxicokinetics of Ab (a) and ADC (b) in rats and cynomolgus monkeys after multiple doses of A1mcMMAF or huA1 alone.

Table 2. Pharmacokinetics Comparison in Mice, Rats, and Cynomolgus Monkeys Given IV Doses of anti-5T4-mcMMAF

a

Rat exposure data obtained from a composite profile (thus AUC and CL values are provided as mean values only). bAUC from an incomplete profile (concentrations were below the LLOQ beyond 168 h).

released payload in the plasma compartment, a look into the tumor compartment paints a different picture; in this case, cysmcMMAF concentrations of 200 ng/mL in tumor homogenate compared to 0.3 ng/mL in plasma samples. This type of focused and limited-scale tumor distribution study also showed that the cys-mcMMAF concentrations in tumor were sustained (a desired quality for pharmacodynamic effects such as efficacy) and limited (differentiated) in its disposition when evaluating other highly vascularized tissues (such as lung and heart) and clearance organs (such as liver and kidney). These observations were corroborated in two different mouse tumor models considered important for its development, a lung model (H1975) and a breast model (MDAMB-361/DYT2).8 The observation or trends that came out of these two models can be considered to be significant and worthwhile corroborating in

preclinical species and providing a degree of confidence toward the clinical predictions.14,18



DISCUSSION The plasma pharmacokinetics of A1mcMMAF in mice were similar in nude mice as well as in a highly 5T4 expressing tumor model. Determining the Ab and ADC in mouse tumor models is the most common approach in evaluating ADC exposure in a relevant (efficacious) model, particularly during the early discovery process. However, concentrations of the active released payload at the target site (the tumor compartment) may be considered to be a better indicator of an ADC that should be brought forward into development. In our limited scale tumor distribution studies we proved that even though a dose of A1mcMMAF results in very low concentrations of the F

DOI: 10.1021/acs.bioconjchem.5b00205 Bioconjugate Chem. XXXX, XXX, XXX−XXX

Article

Bioconjugate Chemistry

relatively similar in rats and monkeys, suggesting that the degree of deconjugation in the systemic circulation is slightly more pronounced in monkeysthis can also be seen in the ADC/Ab ratio which is lower (approximately 50%) in monkeys compared to that in rats (65−72%). In these monkey studies, we observed a relatively low degree of ADA incidence is monkeys (3.8%) is similar to what was reported for T-DM1 (4.2%) also without an impact on the pharmacokinetics.28,29 In rats, although the overall ADA incidence was 27% and resulted in some alterations in PK, they were not considered to be significant. The impact of ADA on PK in rats was also reported.30 The evaluation of DAR, using a combination of high affinity capture and capillary LC-MS, from in vivo studies is increasingly becoming an important variable to follow as DAR distribution during its gradual shift to lower DAR with time in vivo.31 For A1mcMMAF, the average loading of 2.1 (down from an average DAR of 3.6 measured from samples taken 5 min after dosing) observed after a week of dosing and an average DAR of 1.9 after 2 weeks were reassuring with respect to the anticipated clinical dosing regimen of Q3W (once every 3 weeks) by suggesting that although the average DAR loading drops to approximately 59% (of the value observed at 5 min after dosing) within the first week of dosing there should be sufficient conjugated payload in circulation available for delivery to the active site (i.e., the tumor) for at least the first 2 weeks of the 3 week dosing cycle. This value (59%) is also similar to the approximately 58% ratio of ADC to Ab observed after the first dose in the monkey toxicology studies. In this case, the LBA-based ADC to Ab ratio proved to be representative of (or predictive of) the DAR-based evaluation determined by LC-MS/MS methodology.

the clinic and/or using different ADC’s: the high and sustained tumor released payload concentrations were similar, when expressed on a molar basis, to the plasma Ab and ADC concentrations. The other trend is that the μM concentrations of Ab and ADC in tumor tissue are similar to the released payload concentration in the other high “value” organs. The trends were apparent in our two models and would need to be corroborated in the clinical situation for this ADC and for other ADC’s. The use of appropriate controls was also helpful in determining target specificity. In our case, using a 5T4 negative Ab (conjugated to mcMMAF) and a 5T4 negative tumor model (Ramos) allowed us to prove that A1mcMMAF and the cytotoxic entity (cys-mcMMAF) does indeed get to the target site (tumor) compared to the other groups. This degree of specificity, with respect to determination of the analyte, was achieved only with very sensitive analytical techniquei.e., an LC-MS/MS method. The evaluation of released payload (cysmcMMAF) exposure and distribution to the target tissue and other organs is of great interest5 and, in our case, was possible because of the exquisite analytical technology rather than the typical radiolabeled isotope techniques (such as 3H, 14C, 111In, and 125I, which are used for tissue distribution studies).11,19−22 Most distribution studies evaluating ADC’s use single labeling techniques (where either the ADC or Ab are labeled) that provide an avenue to determine overall distribution of the label across many tissues; however, they are typically not able to differentiate between the protein (Ab) and the small molecule payload. The same can be said for imaging studies which used a variety of probes (e.g., IR800 or 89Zr).23,24 A notable exception is the work by Alley et al. using both double labeled methods (3H antibody and a 14C labeled MMAF payload) and more recently Cohen et al. (89Zr on the traztuzumab antibody and 131 I on the tubulysin payload) that allows for distinction of the payload-related distribution from that of the antibody.9,25 Although the dual radiolabeled approach is an important tool, the ability to use LC-MS/MS methodology for payload distribution assessment is flexible and has the potential to be utilized in the discovery and screening side of ADC development as well as any potential clinical applicability.26 An other approach that shows potential for determining tumor and tissue distribution, particularly with respect to possibly more precise estimation of dosing and pharmacodynamics of TBBG-targeted therapies, is the use of target specific knock in mice.27 The pharmacokinetic properties of A1mcMMAF were further evaluated in rats and cynomolgus monkeys, the species used in the regulatory toxicology studies, over a wide range of doses (6 to 60 mg/kg in rats and 0.3 to 15 mg/kg in monkeys). The rat toxicokinetic data were taken from satellite groups using a composite profile (staggered bleeding design, typical of such studies given the need to evaluate toxicology end points without interference from the blood volume requirements needed for the various assays); thus, only the mean PK values are provided (Table 2). The single dose data shown in Table 2 indicate that, in general, the pharmacokinetics of A1mcMMAF (specifically the total antibody and ADC) appear linear over a wider range of doses (as noted by the dose normalized AUC values, AUC/dose). While doses normalized AUC for the total Ab are higher in monkeys (ranging from 2390 to 3850) than in rats (ranging from 1767 to 1985), presumably related to the lower clearance in the target expressing species, monkeys, the dose normalized AUC values for the conjugate (ADC) are



CONCLUSION

The pharmacokinetic profile of A1mcMMAF obtained during the various stages of its preclinical development was described. The early focus was on determining, using a combination of LBA and LC-MS/MS analytical methodology, if the cytotoxic payload (cys-mcMMAF) reached the target tissue with the results indicating that it did (with high and sustained concentrations) in two different tumor models (while concentrations remained low in highly vascularized tissues). The next area of focus was to determine target specificity, which, using antibody and tumor model controls, we were able to demonstrate by the high concentrations of cys-mcMMAF in tumor only from the A1mcMMAF group and from the other three control groups. It is important to note that the analytical method used to determine tumor and tissue cys-mcMMAF concentrations in these studies is based on LC-MS/MS technology. The standard approach to tissue distribution studies is to use labeled radioisotopes 3H or 14C or, more recently, fluorophores.9,11,32 Our approach of using “cold” methods was driven by practical and logistical considerations, but mainly because of desired sensitivity and selectivity/ specificity for the small molecule (cys-mcMMAF), something that the typical labeled Ab (or ADC) method is not able to accomplish. Another benefit of using of an LC-MS/MS approach for determining payload concentrations is the flexibility it allows us during early discovery (when multiple linker-payloads are being evaluated) and the ability to take it forward into regulatory and clinical studies (after the appropriate validation work is conducted). G

DOI: 10.1021/acs.bioconjchem.5b00205 Bioconjugate Chem. XXXX, XXX, XXX−XXX

Article

Bioconjugate Chemistry

tissue samples by LC-MS/MS was described earlier.14 Briefly, plasma samples were prepared with solid phase extraction and tumor (as well as liver, lung, heart, and kidney) tissue samples were weighted and lysis buffer was added at 100 mg: 1 mL ratio to the tissue and homogenized. The supernatants were collected after homogenized samples were centrifuged at 5000 × g for 10 min. Supernatant was aliquoted for the ELISA assay and the remaining sample was further processed with SPE cartridge for LC/MS/MS (5500 Qtrap, C18 column and cys-mcMMAD as the internal standard) analysis of the released payload. The lower limit of quantitation was 0.002 and 0.1 ng/mL for plasma and tissue samples, respectively. Rat and Cynomolgus Monkey Studies. Rat and Cynomolgus Monkey Toxicokinetics. As part of a 6-week rat toxicity study, IV doses of A1mcMMAF (DAR = 4.0) were given at dosages of 6, 20, or 60 mg/kg/cycle to satellite groups of male and female S-D rats (3/sex/dose group/time point) with blood samples taken up to 336 h using a staggered bleeding design. In the cynomolgus monkey toxicity study, IV does of A1mcMMAF were given to male and female animals (2−6 kg) at dosages of 1, 3, 9, and 15 mg/kg/cycle with blood samples taken up to 336 h using a serial bleeding design. In both the rat and cynomolgus monkey studies, samples for ADA evaluation were taken at predose and at 336 h after the first and third doses as well as during the recovery period. In these studies, A1mcMMAF was given once every other week for 4 cycles and samples taken after the first and third cycles (days 1 and 29) for Ab and ADC determination using validated methods. In addition, blood samples were obtained from animals pretreatment and during the dosing phase of the study for analysis of ADA induction. Monkey Pharmacokinetics Study. Male cynomolgus monkeys (3/dose group) were given a single IV administration at 0.3 or 3 mg/kg of A1mcMMAF (DAR = 4.0) and plasma samples were taken over 21 days (the 0.03 mg/kg group) or 35 days (the 3 mg/kg group) and assayed for total Ab and ADC (LBA assays) and cys-mcMMAF (LC-MS/MS assay) using validated methods. In addition, plasma samples from the 3 mg/ kg group were also evaluated for drug:antibody ratio (DAR). Validated LBA Assays Used in the Rat and Monkey Studies. In the total antibody assay, the huA1 is captured by its ligand, 5T4, adsorbed on a microtiter plate. The bound huA1 is detected with a biotinylated goat anti-human kappa light chain antibody followed by binding with avidin D horseradish peroxidase (HRP). The addition of the enzyme substrate, 2.2′-azino-di(3-ethyl-benzthiazoline-6-sulfonate) (ABTS), produces a colored end-product. In this total antibody assay, the assay range was 28.6 to 480 ng/mL in 100% matrix. In the ADC assay, A1mcMMAF is captured by a monoclonal antibody, mouse anti-MMAF, adsorbed on a microtiter plate. The bound A1mcMMAF is detected by its ligand, 5T4 conjugated with biotin, followed by binding with poly horseradish peroxidase (HRP) 80 streptavidin. The addition of the enzyme substrate, 2.2′-azino-di(3-ethyl-benzthiazoline-6sulfonate) (ABTS), produces a colored end-product. In this ADC assay, the assay range was 186 to 5000 ng/mL in 100% matrix. In both the A1 and ADC assays, the optical density (OD) is measured using a microplate spectrophotometer at 405 nm. Sample concentrations are determined by interpolation from a standard curve that is fit using a 4-parameter logistic regression model with a weighting factor of 1 in the Watson Laboratory Information Management System (LIMS).

The development path then took us to rats and cynomolgus monkeys where A1mcMMAF was shown to be a well-behaved ADC with respect to exposure and linearity over a wide range of doses and generally a low incidence of ADA, particularly in the monkey. The evaluation of changes in average DAR with time indicated a sufficient degree of stability over multiple weeks in monkeys supporting the intended Q3W dosing regimen in the clinic.



MATERIALS AND METHODS Mouse PK and Tumor Distribution Studies. Mouse Pharmacokinetic Study Design. The pharmacokinetics of A1mcMMAF was investigated in tumor (MDA-MB-435/5T4, ∼300 mm3, +++ expression at 197 000 copies per cell8) and nontumor bearing female athymic nu/nu mice (5/dose group) after single intravenous (IV) administration at 1 or 10 mg/kg of A1mcMMAF (DAR 4.0). All procedures using mice were approved by the Pfizer Institutional Animal Care and Use Committees according to established guidelines. Whole blood samples (10 μL diluted with 190 μL of HEPES-buffered saline [HBS]) were drawn at time points up to 336 h after dosing (the sampling procedure was modified from Boghaert et al.33). Individual concentrations of the A1 and ADC (using LAB methods) and pooled sample concentrations of cys-mcMMAF (the released payload) were used to calculate plasma pharmacokinetic parameters. Mouse Tumor Distribution Study Design. Female nu/nu mice (n = 21/group) were inoculated with MDA-MB-361/ DYT2 (expression at 40 000 per cell) or H1975 (expression at ∼25 000) cells (tumor grown to ∼400−500 mg). All procedures using mice were approved by the Pfizer Institutional Animal Care and Use Committees according to established guidelines. The tumor bearing animals were given a single IV dose of A1mcMMAF (DAR = 4.0) at 3 mg/kg and sacrificed at 1, 6, 24, 48, 96, 168, and 240 h, at which time samples (plasma, tumor, liver, lung, kidney, and heart) were harvested. The second limited mouse distribution study used the female nu/nu mice H1975 model (a positive target model for 5T4) and the Ramos model (a negative target control). Tumor bearing animals were dosed with a single intravenous dose of A1mcMMAF (the positive antibody containing ADC, DAR = 4.0)) or neg8.8-mcMMAF (a negative antibody control conjugated to mcMMAF) at 3 mg/kg, and (as with the initial distribution study) plasma, tumor, liver, lung, kidney, and heart samples were collected at 6, 24, 48, 96, 168, and 240 h. For both distribution studies, plasma and tumor concentrations of total mAb and ADC were quantified using LBA assays, and LCMS/MS analysis was performed to quantify the released cysmcMMAF concentration in plasma and tumor. ELISA Assays Used in the Mouse Studies. Quantitation of the humanized A1 antibody (huA1) and ADC concentrations in mouse plasma using an enzyme-linked immunosorbant assay (ELISA) was described earlier.8,18 The tumor assay used 100 μL of tumor homogenate processed by a bead beater processor. Briefly, for the A1 assay, the capture protein was 5T4 and the detection antibody was biotinylated goat anti-human kappa chain IgG and HRP-Streptavidin conjugate. Optical density was measured on a spectrophotometer. The ADC was detected by ELISA with 5T4 as the capture antigen and a biotinylated antiMMAF antibody and HRP-Streptavidin conjugate for detection. LC-MS/MS Assay to Quantify cys-mc-MMAF. The extraction and quantitation of cys-mcMMAF from plasma and tumor H

DOI: 10.1021/acs.bioconjchem.5b00205 Bioconjugate Chem. XXXX, XXX, XXX−XXX

Article

Bioconjugate Chemistry Validated ADA Assay Used in the Rat and Monkey Studies. In this method, biotin-labeled and ruthenium-labeled A1mcMMAF are coincubated with study samples, positive controls, or negative controls. Antibodies to A1mcMMAF present in the samples must bind to both the biotin- and ruthenium-labeled versions of A1mcMMAF to be detected in this assay. Complexes are captured via the biotinylated A1mcMMAF to blocked streptavidin-coated Multi-Array plates. Final detection is conducted by using the ruthenium-labeled A1mcMMAF and tripropylamine (TPA) to produce an electrochemiluminescent signal employed within the MSD SECTOR Imager 6000 instrument. The resulting relative light units (RLU) are analyzed using the Watson laboratory information management system (LIMS). Validated LC-MS/MS cys-mcMMAF Assay. Serum samples were analyzed for cys-mcMMAF using a validated liquid chromatography tandem mass spectrometry (LC-MS/MS) method. As cys-mcMMAF isomerizes overtime converting from a linear form to a stable cyclic form, the serum samples were treating with base post solid-phase extraction to simplify the assay. Peak areas of the analyte and the corresponding stable label internal standard of cys-mcMMAF were determined in Analyst (MDS Sciex Analyst, version 1.5.2). Responses were imported into Watson LIMS (version 7.4.1, Thermo, Inc., Philadelphia, PA) and a calibration curve was constructed using peak area ratios of the calibration samples and applying weighted (1/×2) linear least-squares regression analysis. All concentrations were calculated using this calibration curve. The assay was validated from 0.100 to 20.0 ng/mL in monkey serum. LC-MS/MS Method for Immunoprecipitation Isolation of the ADC and DAR Evaluation. The ADC was isolated from serum by immunoprecipitation. Briefly, to facilitate deglycosylation, an equal volume of the ADC sample and 100 mM sodium phosphate buffer (pH 7.4) were incubated with IgG ZERO enzyme for 1 h at 37 °C in a low binding eppendorf tube. For enrichment, the ADC was captured by a biotinlabeled goat-anti-human IgG-Fc (Jackson Laboratories) in phosphate buffered saline overnight at 4 °C and then isolated with MyOne Streptavidin T1 magnetic beads (Invitrogen) for 2 h at room temperature with shaking. The isolated and reconstituted ADC was reduced for subsequent LC-MS analysis with 500 mM tris(2-carboxyethyl)phosphine (TCEP) for 30 min at 37 °C. The drug:antibody ratio (DAR) on the heavy and light chains of the ADC was analyzed by reverse-phase LC-MS. Briefly, the reverse-phase separation of reduced ADC was performed on a Waters BEH300 C4 Column (2.1 × 5 mm) using a 25 min gradient (10%−60% B). The mobile phase A was 0.1% formic acid in water, and the mobile phase B contained 0.1% formic acid in acetonitrile. The flow rate and column temperature were maintained at 250 μL/min and 60 °C, respectively. Mass spectrometric analysis was carried out in positive ion mode. The desolvation gas and source temperatures were set to 375 and 90 °C, respectively. The capillary and cone voltages were set at 3.5 and 30 V, respectively. All other voltages were optimized to provide optimal signal intensity. The instrument was calibrated in the m/z range of 400 to 3500 Da using NaI. The deconvolution of ESI mass spectra of the reduced ADC (A1mcMMAF) was performed by Biopharmalynx 1.2 using a MaxEnt 1 algorithm. For deconvolution, the m/z range of 800 to 2500 Da was used with the following MaxEnt 1 parameters: output mass range

from 19 000 to 26 000 Da for light chain and 48 000 to 54 000 Da for heavy chain of the ADC; minimum intensity ratio left and right, 33%; width at half height for uniform Gaussian model, 0.6 (low m/z) and 0.8 (high m/z); number of iteration, 10. Pharmacokinetic Calculations. All toxicokinetic parameters were determined from individual animal data using noncompartmental analysis in WinNonlin (version 5.2, Pharsight, CA) or pharmacokinetics module within Watson LIMS (version 7.4, Thermo, Inc., Philadelphia, PA). The area under the serum concentration−time curve AUC was estimated using the linear trapezoidal. A concentration of 0 μg/mL was used for all results that were below the level of quantitation (BLQ).



ASSOCIATED CONTENT



AUTHOR INFORMATION

* Supporting Information S

Supplemental figures. The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.bioconjchem.5b00205. Corresponding Author

*E-mail: Mauricio.leal@pfizer.com. Tel: 1 845 602 2667. Notes

The authors declare the following competing financial interest(s): All authors are/or were full time employees of Pfizer Inc. when research was performed and own company’s stock options and/or units.



ACKNOWLEDGMENTS The authors are grateful to members of the Oncology In Vivo Pharmacology Group and vivarium staff in Pearl River, NY, and Groton, CT, for the animals studies; John Marcek and M. Giovanni for leading the toxicology studies; Tracey Clark, Steve Hansel, and Dawn Duffield for input and discussion on the pharmacokinetic studies. The authors acknowledge Seattle Genetics Inc. and Oxford BioMedica for access to technology.



REFERENCES

(1) Saber, H., and Leighton, J. K. (2015) An FDA oncology analysis of antibody-drug conjugates. Regul. Toxicol. Pharmacol. 71, 444−452. (2) Hole, N., and Stern, P. L. (1988) A 72 kD trophoblast glycoprotein defined by a monoclonal antibody. Br. J. Cancer 57, 239− 246. (3) Hole, N., and Stern, P. L. (1990) Isolation and characterization of 5T4, a tumor-associated antige. Int. J. Cancer 45, 179−184. (4) Boghaert, E. R., Sridharan, L., Khandke, K. M., Armellino, D., Ryan, M. G., Myers, K., Harrop, R., Kunz, A., Hamann, P. R., Marquette, K., et al. (2008) The oncofetal protein, 5T4, is a suitable target for antibody-guided anti-cancer chemotherapy with calicheamicin. Int. J. Oncol. 32, 221−34. (5) Starzynska, T., Marsh, P. J., Schofield, P. F., Roberts, S. A., Myers, K. A., and Stern, P. L. (1994) Prognostic significance of 5T4 oncofetal antigen expression in colorectal carcinoma. Br. J. Cancer 69, 899−902. (6) Wrigley, E., McGown, A. T., Rennison, J., Swindell, R., Crowther, D., Starzynska, T., and Stern, P. L. (1995) 5T4 oncofetal antigen expression in ovarian carcinoma. Int. J. Gynecol Cancer 5, 269−74. (7) Southall, P. J., Boxer, G. M., Bagshawe, K. D., Hole, N., Bromley, M., and Stern, P. L. (1990) Immunohistological distribution of 5T4 antigen in normal and malignant tissues. Br. J. Cancer 61, 89−95. (8) Sapra, P., Damelin, M., Dijoseph, J., Marquette, K., Geles, K. G., Golas, J., Dougher, M., Narayanan, B., Giannakou, A., Khandke, K., et al. (2013) Long-term tumor regression induced by an antibody-drug conjugate that targets 5T4, an oncofetal antigen expressed on tumorinitiating cells. Mol. Cancer Ther. 12, 38−47.

I

DOI: 10.1021/acs.bioconjchem.5b00205 Bioconjugate Chem. XXXX, XXX, XXX−XXX

Article

Bioconjugate Chemistry (9) Alley, S. C., Zhang, X., Okeley, N. M., Anderson, M., Law, C., Senter, P. D., and Benjamin, D. R. (2009) The pharmacologic basis for antibody-auristatin conjugate activity. J. Pharmacol. Exp. Ther. 330, 932−938. (10) Erickson, H. K., and Lambert, J. M. (2012) ADME of antibodymaytansinoid conjugates. AAPS J. 14, 799−805. (11) Shen, B., Bumbaca, D., Saad, O., Yue, Q., Pastuskovas, C. V., Khojasteh, S. C., Tibbitts, J., Kaur, S., Wang, B., Chu, Y., et al. (2012) Catabolic Fate and Pharmacokinetic Characterization of Trastuzumab Emtansine (T-DM1): an Emphasis on Preclinical and Clinical Catabolism. Current Drug Metabolism 13, 901−910. (12) Lin, K., and Tibbitts, J. (2012) Pharmacokinetic Considerations for Antibody Drug Conjugages. Pharm. Res. 29, 2354−2366. (13) Leal, M., Wentland, J., Clark, T., Marcek, J., Khandke, K., Zhang, J., DiJoseph, J., Lucas, J., Groeber, E., and Han, X., et al. (2011) Preclinical Pharmacokinetics of an Antibody-Drug conjugate Targeting 5T4, Abstract C117, 2011 AACR-NCI-EORTC International conference on Molecular Targets and Cancer Therapeutics, Nov. 12−16, 2011. (14) Shah, D. K., King, L. E., Han, X., Wentland, J. A., Zhang, Y., Lucas, J., Haddish-Berhane, N., Betts, A., and Leal, M. (2014) A priori prediction of tumor payload concentrations: preclinical case study with an auristatin-based anti-5T4 antibody-drug conjugate. AAPS J. 16, 452−463. (15) Leal, M. (2012). Pharmacokinetics and disposition of an anti5T4 antibody-drug conjugate, vol 44 (S1), pp 13−14, Drug Metabolism Reviews Conference: 18th North American Regional ISSX Meeting, Dallas, TX, United States. (16) Rago, B., Han, X., and Leal, M. (2013) Elevated drug level in antigen-expressing tumor demonstrates specific delivery of payload by immunoconjugate, 2013 AAPS-NBC Poster T2082. (17) Quazi, S., Leal, M., Clark, T. and Dufield D. (2012) Stability of Antibody Drug Conjugates In Vivo by Observing Changes in Average Drug-to-Antibody Ratios Using Affinity Capture LC-MS and Software, HUPO 11th Annual Congress, September 9−13, 2012, Boston, Massachusetts. (18) Haddish-Berhane, N., Shah, D. K., Ma, D., Leal, M., Gerber, H. P., Sapra, P., Barton, H. A., and Betts, A. M. (2013) On translation of antibody drug conjugates efficacy from mouse experimental tumors to the clinic: a PK/PD approach. J. Pharmacokinet. Pharmacodyn. 40, 557−571. (19) Yip, V., Palma, E., Tesar, D. B., Mundo, E. E., Bumbaca, D., Torres, E. K., Reyes, N. A., Shen, B. Q., Fielder, P. J., Prabhu, S., et al. (2014) Quantitative cumulative biodistribution of antibodies in mice Effect of modulating binding affinity to the neonatal Fc receptor. mAbs 6, 689−696. (20) Xie, H., Audette, C., Hoffee, M., Lambert, J. M., and Blattler, W. A. (2003) Pharmacokinetics and Biodistribution of the Antitumor Immunoconjugate, Cantuzumab Mertansine (huC242-DM1), and Its Two Components in Mice. J. Pharmacol. Exp. Ther. 308, 1073−1082. (21) Boswell, A. A., Bumbaca, D., Fielder, 1P.J., and Khawli, L. A. (2012) Compartmental Tissue Distribution of Antibody Therapeutics: Experimental Approaches and Interpretations. AAPS J. 14, 612−618. (22) Boswell, C. A., Tesar, D. B., Mukhyala, K., Theil, F. P., Fielder, P. J., and Khawli, L. A. (2010) Effects of charge on antibody tissue distribution and pharmacokinetics. Bioconjugate Chem. 21, 2153−2163. (23) Conner, K. P, Rock, B. M., Kwon, G. K., Balthasar, J. P., Abuqayyas, L., Wienkers, l.C., and Rock, D. A. (2014) Evaluation of near infrared fluorescent labeling of monoclonal antibvodies as a tool for tissue distribution. Drug Metab. Dispos. 42, 1906−1913. (24) Williams, S.-P. (2012) Tissue distribution studies of protein therapeutics using molecular probes: molecular imaging. AAPS J. 14, 389−399. (25) Cohen, R., Vugts, D. J., Visser, G. W. M., Stigter-van Walsum, M., Bolijn, M., Spiga, M., Lazzari, P., Shankar, S., Sani, M., Zanda, M., et al. (2014) Development of novel ADCs: Conjugation of tubulysin analogues to Trastuzumab monitored by dual radiolabeling. Cancer Res. 74, 5700−5710.

(26) Srinivasan, M., Thevanayagam, L., Chakraborty, I., Bell, A., Deshpande, S., Pan, C., Cardarelli, P. M., Gangwar, S., Chong, C., and Cortez, O., et al. (2010) Biodistribution of antibody drug conjugates by LC-MS. Cancer Res. 70, (suppl 8) Abstract 2446, Proceedings of the 101st Annual Meeting of the American Association for Cancer Research, Washington, DC, 10.1158/1538-7445.AM10-2446 (27) Hu, G., Leal, M., Lin, Q., Affolter, T., Sapra, P., Bates, B., and Damelin, D. (2015) Phenotype of TPBG Gene Replacement in the Mouse and Impact on the Pharmacokinetics of an Antibody−Drug Conjugate. Mol. Pharmaceutics 12, 1730−1737. (28) Hock, M. B., Thudium, K. E., Carrasco-Triguero, M., and Schwabe, N. F. (2015) Immunogenicity of Antibody Drug Conjugates: Bioanalytical Methods and Monitoring Strategy for a Novel Therapeutic Modality. AAPS J. 17, 35−43. (29) Carrasco-Triguero, M., Yi, J. H., Dere, R., Qiu, Z. J., Lei, C., Li, Y., Mahood, C., Wang, B., Leipold, D., Poon, K. A., and Kaur, S. (2013) Immunogenicity assays for antibody-drug conjugates: case study with ado-trastuzumab emtansine. Bioanalysis 5, 1007−23. (30) Hoofring, S. A., Lopez, R., Hock, M. B., Kaliyaperumal, A., Patel, S. K., Swanson, S. J., Chirmule, N., and Starcevic, M. (2013) Immunogenicity testing strategy and bioanalytical assays for antibodydrug conjugates. Bioanalysis 5, 1041−55. (31) Xu, D., et al. (2011) Characterization of intact antibody−drug conjugates from plasma/serum in vivo by affinity capture capillary liquid chromatography-mass spectrometry. Anal. Biochem. 412, 56−66. (32) Boswell, C. A., Mundo, E. E., Zhang, C., Bumbaca, D., Valle, N. R., Kozak, K. R., et al. (2011) Impact of drug conjugation on pharmacokinetics and tissue distribution of anti-STEAP1 antibodydrug conjugates in rats. Bioconjugate Chem. 22, 1994−2004. (33) Boghaert, E. R., Khandke, K. M., Sridharan, L., Dougher, M., DiJoseph, J. F., Kunz, A., Hamann, P. R., Moran, J., Chaudhary, I., and Damle, N. K. (2008) Determination of pharmacokinetic values of calicheamicin-antibody conjugates in mice by plasmon resonance analysis of small (5 ul) blood samples. Cancer Chemother. Pharmacol. 61, 1027−1035.

J

DOI: 10.1021/acs.bioconjchem.5b00205 Bioconjugate Chem. XXXX, XXX, XXX−XXX