Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Jul 15.
Published in final edited form as: Clin Cancer Res. 2017 Mar 31;23(14):3489–3498. doi: 10.1158/1078-0432.CCR-16-3083

Clinically relevant concentrations of anticancer drugs: A guide for nonclinical studies

Dane R Liston 1, Myrtle Davis 1
PMCID: PMC5511563  NIHMSID: NIHMS863879  PMID: 28364015

Abstract

Approved and marketed drugs are frequently studied in nonclinical models to evaluate the potential application to additional disease indications or to gain insight about molecular mechanisms of action. A survey of the literature reveals that nonclinical experimental designs (in vitro or in vivo) often include evaluation of drug concentrations or doses that are much higher than what can be achieved in patients (i.e., above the maximally tolerated dose or much higher than the clinically relevant exposures). The results obtained with these high concentrations may be particularly helpful in elucidating off-target effects and toxicities, but it is critical to have a dose-response curve that includes the minimally effective or clinically effective concentration for comparison. We have reviewed the clinical literature and drug product labels for all small molecules and biological agents approved by the U.S. Food and Drug Administration (FDA) for use in oncology in order to identify and compile the available pharmacokinetic parameters. The data summarized here can serve as a guide for selection of in vitro concentrations and in vivo plasma exposures for evaluation of drug effects in nonclinical studies. Inclusion of drug concentrations or exposures that are relevant to those observed in clinical practice can improve translation of nonclinical mechanism of action findings into potentially relevant clinical effects.

Keywords: oncology, pharmacokinetics

Introduction

Nonclinical studies are important foundations for modern drug discovery. Beyond initial discovery, nonclinical investigations with approved drugs are frequently conducted to explore possibilities for expanded use and additional disease indications. In this situation, nonclinical experiments can take advantage of existing pharmacokinetic and toxicity findings, along with related exposure data, to design studies to test drugs at concentrations in vitro or in vivo that are relevant to observed clinical exposures. In so doing, concentrations known to be achievable and efficacious in patients can be included in the design of novel nonclinical studies. In a recent commentary, Smith and Houghton (1) cited several examples of reported activities of anti-cancer agents that were derived from in vitro studies that used concentrations far greater than those that could be realistically achieved in a clinical setting. In some cases, these drug concentrations were several orders of magnitude greater than concentrations needed to inhibit the desired targets of the drug. The use of such high concentrations increases the possibility that the effects observed are due to off-target activities that are not relevant when the drug is provided at therapeutic concentrations in clinical practice, and efforts to translate conclusions drawn from these studies may be unsuccessful. Therefore, awareness of the relationship between concentrations tested nonclinically and what is achievable in a clinical context can greatly assist the interpretation and translation of such studies.

As an aid to guide dose and concentration selection, we provide herein a comprehensive compilation of human plasma exposures for drugs approved by the U.S. FDA for use in oncology. We sought to identify the maximum plasma concentration at the highest single dose recommended in the Drug Product Label to be used as a guide to derive a range of drug concentrations to include in nonclinical studies. We have focused on therapies that have direct effects on tumor growth or cancer cell viability. Adjunct or strictly palliative therapies such as analgesics and anti-emetics were excluded, although these are widely used for supportive care during cancer treatment. We also excluded diagnostics, imaging agents and radiological therapies. Several drugs that are not approved specifically for use in cancer but are increasingly being reported in experimental settings (e.g., metformin, celecoxib) have been included where possible.

Methods

A comprehensive list of agents approved for use as anticancer therapies in the U.S. was assembled from several sources. The National Cancer Institute (NCI) maintains a list of approved drugs with drug information summaries (2) ; this list includes most individual agents plus many commonly used drug combinations in oncology. A list of single agents derived from this source was cross-checked against lists of oncology therapies compiled by MediLexicon (3) and Centerwatch (4), two databases which allow searching of FDA-approved drugs by therapeutic area (oncology). The resulting combined list was triaged to remove strictly palliative agents, such as analgesics and anti-emetics. Combination drug therapies were removed from the list, since each component within the combinations was included as the individual drug. Biological agents were parsed into a separate list. Within biological agents, vaccines were not included. All compounds on the final list were verified for approval status at fda.gov, and drug product labels were downloaded from FDA (5) or DailyMed (6), a service provided by the National Library of Medicine.

Human pharmacokinetic (PK) data were identified by examination of the drug product label, the original literature or conference abstracts, with priority given to the clinical pharmacology section within the drug product label. The intent was to determine the exposure defined by maximum plasma concentration (Cmax) and the integrated area under the plasma concentration-time curve (AUC) associated with the highest recommended dose of the drug. If the information in the label was not sufficiently explicit or detailed to derive exposures associated with discrete dosing levels, the original publications describing the PK data were identified using Thompson-Reuters Integrity® (7) and PubMed. These sources were reviewed and a single reference was selected for each compound based on 1) the use of a dose equivalent to the highest dose recommended in the label and 2) the availability of the key parameters of Cmax and AUC, typically calculated from time zero to infinity. Whenever possible, studies reporting Cmax and AUC following a single administration at the highest dose recommended in the product label were chosen for review. When these data were not found, the study reporting a dose as close as possible to the highest recommended dose was selected.

Results

Our survey identified 145 unique small molecule drugs approved to treat cancer, of which 10 are prodrugs. Table 1 provides a summary of the human PK parameters for all 135 unique small molecule drugs and 5 alternative formulations approved for use in oncology indications in the U.S, excluding prodrugs. Table 2 includes all 10 drugs delivered as prodrugs, where the chief pharmacological activity is provided by an active metabolite. The 40 unique biological agents that have been approved for oncology are summarized in Table 3. We noted 16 additional drugs that are not currently approved for cancer indications but have been reported in clinical trials for various cancers; these have been included in Table 4.

Table 1.

Key human pharmacokinetic parameters for small-molecule drugs approved for oncology indications

Generic Name Brand Name Dose Dose Unit Route infusion Cmax (uM) Cmax (ng/ml) AUC (ng•hr/ml) Tmax (hr) T1/2 (hr) Protein binding
Abarelix Plenaxis 100 mg IM - 0.031 43.4 12000 72 316.8 96–99%
Afatinib Gilotrif 40 mg PO - 0.052 25 324 4.0 26.9 95%
Alectinib Alecensa 600 mg PO - 1.38 665 7430 4.0 33 >99%
Allopurinol Zyloprim 300 mg PO - 14.3 1940 4814 1.4 1.4 negligible
Altretamine Hexalen 200 mg PO - 3.76 790 - 0.5–3 4.7– 10.2 94%
Amifostine Ethyol 200 mg/m2 IV 7.5 min 105 22472 4238 - 0.26 negligible
Aminolevulinic Acid Levulan Kerastick 100 mg IV* 1 min 129 16900 13700 - 0.83 -
Anastrozole Arimidex 1 mg PO - 0.035 10 536 - 41.3 40%
Arsenic Trioxide Trisenox 0.1 mg/kg IV 2h 0.910 180 see notes - - 75%
Axitinib Inlyta 5 mg PO - 0.163 63 466 1.8 3.1 >99%
Azacitidine Vidaza 75 mg/m2 SC - 3.07 750 960 0.50 0.68 -
Azacitidine Vidaza 75 mg/m2 IV 10–40 min 11.3 2750 1044 - 0.36 -
Belinostat Beleodaq 1000 mg/m2 IV 30 min 134 42657 29005 - 1 94%
Bendamustine Treanda 120 mg/m2 IV 60 min 16.3 5840 13635 - 0.7 94–96%
Bexarotene Targretin 300 mg/m2 PO - 3.39 1180 5980 2.5 3.4 >99%
Bicalutamide Casodex 50 mg PO - 1.78 768 230838 31 139 96%
Bleomycin Blenoxane 15 mg/m2 IV bolus 706 1000000 4.99E+06 - 4 1%
Bortezomib Velcade 1.3 mg/m2 IV bolus 0.312 120 196 0.08 48.7 83%
Bosutinib Bosulif 500 mg PO - 0.377 200 3650 4–6 22.5 96%
Busulfan Busulflex 0.8 mg/kg IV 2h 4.96 1222 4790 - - 32%
Cabazitaxel Jevtana 25 mg/m2 IV 1 h 0.270 226 991 1.0 95 89–92%
Cabozantinib Cometriq 140 mg PO - 4.61 2310 41600 2–5 55 99.7%
Capecitabine Xeloda 1250 mg/m2 PO - 21.1 7570 8450 0.82 0.43 approx 35%
Carboplatin Paraplatin 400 mg/m2 IV 30 min 135 50000 83333 0.50 3 0%
Carfilzomib Kyorolis 27 mq/m2 IV 5 min 5.88 4232 379 - < 1 97%
Carmustine BiCNU 600 mg/m2 IV 2h 19.4 4150 - - - 80%
Ceritinib Zykadia 750 mg PO - 1.21 674 14,000 5.0 41 97%
Chlorambucil Leukeran 0.2 mg/kg PO - 1.62 492 883 0.83 1.3 99%
Cisplatin Platinol 80 mg/m2 IV 1 h 14.4 4321 42921 - 0.44 n/a*
Cladribine Leustatin 0.09 mg/kg/day IV 24 h 0.020 5.7 - - - 20%
Cladribine Leustatin 0.12 mg/kg IV 2h 0.168 48 - - 5.4 20%
Clofarabine Clolar 40 mg/m2 IV 2h 0.744 226 931 - 4.9 47%
Cobimetinib Cotellic 60 mg PO - 0.514 273 4340 2.4 44 95%
Crizotlnib Xalkori 250 mg PO - 0.913 411 3880 4.0 34.9 91%
Cyclophosphamide Cytoxan 600 mg/m2 IV bolus 128 33408 226082 - 3–12 20%
Cytarabine Cytosar-U 3000 mg/m2 IV 3h 54.4 13219 38928 - 3.82 13%
Dabrafenib Tafinlar 150 mg PO - 4.86 2527 10751 2.0 4.8 99.7%
Dacarbazine DTIC-Dome 200 mg/m2 IV 30min 34.4 6270 4860 - 5 <5%
Dactinomycin Cosmegen 0.70–150 mg/m2 IV bolus 0.020 25 44.5 0.25 14–43 5%
Dasatinib Sprycel 100 mg PO - 0.264 129 478 2.0 6.2 96%
Daunorubicin Daunoxome 50 mg/m2 IV 1 h 0.310 175 575 - 11 97%
Decitabine Dacogen 15 mg/m2 IV 3h 0.323 74 163 2.5 0.62 <1%
Degarelix Firmagon 240 mg SC - 0.016 26 25296 48 - 90%
Dexrazoxane Zinecard 500 mg/m2 IV 15 min 136 36500 - - 2.5 <2%
Docetaxel Taxotere 100 mg/m2 IV 1 h 5.47 4420 5900 - 41 97%
Doxorubicin Adriamycin 60 mg/m2 IV 5 min 6.73 3660 1850 - 14.2 75%
Doxorubicin (liposomal) Doxil 20 mg/m2 IV 30 min 15.34 8340 590000 - 55 70%
Enzalutamide Xtandi 160 mg PO - 35.7 16600 - 1.0 5.8 98%
Epirubicin Ellence 120 mg/m2 IV 10 min 16.6 9000 3400 - 33.7 77%
Eribulin Mesylate Halaven 1.4 mg/m2 IV −5 min 0.508 371 757 0.17 40.4 49–65%
Erlotinib Tarceva 150 mg PO - 3.15 1238 18.6 5.5 24.4 93%
Etoposide VePesid 100 mg/m2 IV 60 min 33.4 19660 29800 - 3.62 97%
Everolimus Afinitor 10 mg PO - 0.064 61 514 1.0 - 74%
Exemestane Aromasin 25 mg PO - 0.027 7.9 52 1.2–2.9 24 90%
Fluorouracil (5-FU) Adrucil 400 mg/m2 IV push 426 55400 11590 - - 10%
Flutamide Eulexin 250 mg PO - 0.409 113 - 1.3 7.8 94–96%
Fulvestrant Faslodex 500 mg IM - 0.041 25 11400 - 960 99%
Gefitinib Iressa 250 mg PO - 0.356 159 5115 3.0 50.5 90%
Gemcitabine Gemzar 1250 mg/m2 IV iO min 89.3 23500 12500 - 0.23 negligible
Goserelin acetate Zoladex 10.8 mg SC - 0.007 8.9 - 1.8 - 27%
Histrelin acetate Supprelin 0.057 mg/day SC - 0.00083 1.1 2318 12 - 70%
Hydroxyurea Droxia 2000 mg PO - 795 60441 299181 1.2 3.32 -
Ibrutinib Imbruvica 560 mg PO - 0.277 122 1263 2.0 9.2 98%
Idarubicin Idamycin 15 mg/m2 IV 5 min 0.123 61 173 - 21.8 97%
Idelalisib Zydelig 150 mg PO - 5.18 2152 9599 1.8 5.75 84%
Ifosfamide Ifex 3000 mg/m2 IV 3h 431 112534 2E+06 - 4.1 negligible
Imatinib Gleevec 600 mg PO - 7.50 3700 48800 - 17 95%
Imiquimod Aldara 75 mg topical - 0.0056 1.4 0.0291 - - 90–95%
Ingenol Picato 0.5 mg topical - BLOQ BLOQ - - - >99%
Ixabepilone Ixempra 40 mg/m2 IV 3h 0.497 252 2143 - 35 67–77%
Ixazomib Ninlaro 4 mg PO - 0.118 61 1160 1.0 228 99%
Lanreotide Somatuline Depot 120 mg SC - 0.007 7.7 - - - -
Lapatinib Tykerb 1250 mg PO - 4.18 2430 36200 4.0 14.2 >99%
Lenalidomide Revlimid 25 mg PO - 1.74 451 3820 1.5 5.3 30%
Lenvatinib Lenvima 24 mg PO - 0.761 325 3010 2.0 28 98–99%
Letrozole Femara 2.5 mg PO - 0.406 116 2246 1.5 - 60%
Leuprolide Acetate Eligard 30 mg SC - 0.124 150 - 3.3 - 43–49%
Mechlorethamine (Chlormethine) Mustargen 0.4 mg/kg IV bolus BLOQ BLOQ - - - -
Megestrol acetate Megace 800 mg PO 1.96 753 10476 5.0 - -
Melphalan Alkeran 20 mg/m2 IV 15–20 min 9.17 2800 - - 1.25 60–90%
Mercaptopurine Purinethol 75 mg/m2 PO - 0.590 90 274 - 1.3 19%
Mercaptopurine Purixan 50 mg PO 0.625 95 136 - 2 19%
Methotrexate Abitrexate 30 mg PO - 1.31 594 2466 1.2 2.9 50%
Methoxsalen Uvadex; 8-MOP 40 mg PO - 0.624 135 440 2.0 2 90%
Mitomycin C Mitozytrex 15 mg/m2 IV 30min 2.18 729 691 - 0.81 24%
Mitotane Lysodren 6000 mg/day PO - 50.9 16300 - - 432–3816 6%
Mitoxantrone Novantrone 12 mg/m2 IV 30 min 0.715 318 298 - 17 78%
nab-Paclitaxel Abraxane 260 mg/m2 IV 30 min 21.9 18740 20324 - 27 89–98%
Nilotinib Tasigna 400 mg PO - 0.840 445 11900 4.0 13 98%
Nilutamide Nilandron 150 mg PO - 2.84 900 39000 2.8 56 80–84%
Octreotide Sandostatin 0.1 mg SC - 0.0038 4.0 12.4 0.64 2.25 65%
Olaparib Lynparza 400 mg PO - 13.1 5700 58000 1.3 11.9 82%
Omacetaxine Synribo 1.25 mg/m2 SC - 0.046 25 136 0.55 7 50%
Osimertinib Tagrisso 80 mg PO qd 0.126 63 3132 6.0 64 99%
Oxaliplatin Eloxatin 110 mg/m2 IV 2h 4.96 1970 4990 - 1.86 >90%
Paclitaxel Taxol 175 mg/m2 IV 3h 4.27 3650 15007 - 20.2 89–98%
Palbociclib Ibrance 125 mg PO - 0.101 45 1427 6.0 22.2 85%
Pamidronate Aredia 90 mg IV 4h 11.1 2610 17120 4.0 - -
Panobinostat Farydak 20 mg PO - 0.082 29 280 1.0 15.5 90%
Pazopanib Votrient 800 mg PO - 133 58100 1037000 2–4 30.9 >99%
Pemetrexed Alimta 500 mg/m2 IV 10 min 306 131000 188000 - 4.4 81%
Pentostatin Nipent 4 mg/m2 IV 15 min 1.82 489 1232 - 5.3 4%
Plerixafor Mozobil 0.24 mg/kg SC - 1.84 926 4741 0.50 5.1 58%
Pomalidomide Pomalyst 4 mg PO - 0.274 75 400 2–3 9.5 12–44%
Ponatinib Iclusig 45 mg PO - 0.137 73 1253 - 24 >99%
Porfimer Photofrin 2 mg/kg IV 3–5 min 33.9 40000 2400000 - 415 90%
Pralatrexate Folotyn 40 mg/m2 IV 5 min 10.3 4900 4900 - 1.8 67%
Prednisone Deltasone 50 mg PO - 0.145 52 - - - extensive
Procarbazine Matulane 300 mg PO - 3.13 692 217 0.21 0.154 -
Raloxifene Evista 60 mg PO - 0.0011 0.5 272 (nb•hr/ml)/(mg/kg) - 27.7 95%
Regorafenib Stivarga 160 mg PO - 8.08 3900 58300 4.0 28 99.5%
Romidepsin Istodax 14 mg/m2 IV 4h 0.697 377 1549 - 3 92–94%
Rucaparib Rubraca 600 mg PO - 6.000 1940 16900 1.9 17 70%
Ruxolitinib Jakafi 25 mg PO - 1.09 335 979 0.63 2.3 97%
Sonidegib Odomzo 200 mg PO - 2.12 1030 22000 2–4 672 >97%
Sorafenib Nexavar 400 mg PO - 20.1 9350 107000 2.5 23.8 99.5%
Streptozocin Zanosar 1500 mg/m2 IV push 1438 381400 72150 - 0.22 -
Sunitinib Malate Sutent 50 mg PO - 0.181 72 1296 8.5 41–86 95%
Tamoxifen Citrate Nolvadex 20 mg PO - 0.108 40 - 5.0 120–168 >99%
Tegafur Utefos 50 mg/m2 PO - 19.0 3803 26480 1.0 7.88 -
Temsirolimus Torisel 25 mg IV 0–60 min 0.568 585 1627 - 17 87%
Teniposide Vumon 300–750 mg/m2 IV 72 h 23.1 15200 - - - >99%
Thalidomide Thalomid 200 mg PO - 8.91 2300 23300 5.8 4.12 55% (+)-(R); 66%
Thiolguanine Tabloid 40 mg/m2 PO - 0.313 52 0.0979 1.5 - -
Topotecan Hycamtin 2.3 mg/m2 PO - 0.015 6.3 22.7 3.49 35%
Toremifene Fareston 120 mg PO - 4.56 1850 115100 2.9 56.5 >99.5%
Trabectedin Yondelis 1.5 mg/m2 IV 24 hr 0.0024 1.8 56.8 175 97%
Trametinib Mekinist 2 mg PO - 0.021 13 69.7 2.0 3.9–4.8 97%
Tretinoin Vesanoid 45 mg/m2 PO - 1.15 347 682 2.2 - >95%
Triptorelin Trelstar Depot 22.5 mg IM - 0.034 44 - 1–3 - 0%
Valrubicin Valstar 800 mg Intra Ciste - 0.011 8 56.4 2–6 - >99%
Vandetanib Caprelsa 300 mg PO - 2.16 1025 20460 5 456 90%
Vemurafenib Zelboraf 960 mg PO - 127 62000 601000 3.0 57 >99%
Venetoclax Venclexta 400 mg PO - 4.48 21000 32800 5–8 26 >99%
Vinblastine Velban 1** mg/m2 IV bolus 0.035 28 45 0.12 26.20 98–99%
Vincristine Vincasar PFS 1 mg/m2 IV bolus 0.007 5.4 - - - 75%
Vincristine (liposomal) Marqibo 2.25 mg/m2 IV 60 min 1.479 1220 14566 - 7.66 75%
Vinorelbine Navelbine 25 mg/m2 IV 20 min 0.811 632 585 - 21.40 89%
Vismodegib Erivedge 150 mg PO - 33.9 14282 2283446 - 96 >99%
Vorinostat Zolinza 400 mg PO - 1.20 317 1110 1.5 2 71%
Zoledronic Acid Zometa 4 mg IV 15 min 0.971 264 420 - 146 33–40%

Abbreviations: Cmax: maximum plasma concentration; AUC: Area Under the time-plasma Concentration curve; : Tmax: Time post-dose of maximum plasma concentration; : T1/2: Plasma half-life

Table 2.

Key human pharmacokinetic parameters for small-molecule pro-drugs and their *active metabolites approved for oncology indications

Generic Name Brand Name Dose Dose Unit Route infusion Cmax (uM) Cmax (ng/ml) AUC (ng•hr/ml) Tmax (hr) T1/2 (hr) Protein binding
Abiraterone Acetate Zytiga 1000 mg PO - BLOQ BLOQ - - - -
*Abiraterone (Zytiga) as above PO - 0.647 226 1173 - 12 >99%
Estramustine phosphate Emcyt 2000 mg/m2 IV 60min 797 414700 889884 - 2.5 -
* estramustine (Emcyt) as above IV - 10.4 4420 37605 - 99 -
* estramustrone (Emcyt) as above IV - 15.6 6620 211351 - 129 -
Floxuridine FUDR 30 mg/kg IV 8h 1.05 259 1365 - 0.22 -
* 5-FU (FUDR) as above - 0.88 115 - - - -
Fludarabine Phosphate Fludara 25 mg/m2 IV 30min BLOQ BLOQ - - - 19–29%
* 2-F-araA (Fludara) as above IV - 3.00 808 3285 - 11.3 -
Irinotecan Camptosar 340 mg/m2 IV 90min 5.78 3392 20604 - 11.7 30–68%
* SN-38 (Camptosar) as above IV - 0.143 56 474 - 21 95%
Irinotecan (liposomal) Onivyde 70 mg/m2 IV 90min 63.4 37200 1364000 - 25.8 <0.44%
* SN-38 (liposomal) (Onivyde) as above IV - 0.014 5.4 620 - 67.8 95%
Lomustine (CCNU) Gleostine 130 mg/m2 PO - BLOQ BLOQ - - - 50%
* cis+trans 4-OH-CCNU (Gleostine) as above PO - 3.39 847 3400 2–4 1.8 -
Nelarabine Arranon 1500 mg/m2 IV 2h 16.8 5000 4400 - 0.3 <25%
* ara-G (Arranon) as above IV - 111 31400 162000 - 3.2 <25%
Temozolomide Temodar 150 mg/m2 IV 90min 37.6 7300 24600 1.0 1.8 15%
*MTIC (Temodar) as above IV - 1.64 276 891 - - -
Testosterone enanthate Delatestryl 200 mg IM - n.d. n.d. - - - -
* Testosterone (Delatestryl) as above IM - 0.051 15 - 55 - 98%
Thiotepa Thioplex 80 mg IV push 9.66 1828 4127 - 2.3 10%
*TEPA (Thioplex) as above IV - 2.04 353 7452 - 15.7 -

Abbreviations: Cmax: maximum plasma concentration; AUC: Area Under the time-plasma Concentration curve; : Tmax: Time post-dose of maximum plasma concentration; : T1/2: Plasma half-life

Table 3.

Key human pharmacokinetic parameters for biological drugs approved for oncology indications

Generic Name Brand Name Dose Dose Unit Route infusion Cmax (uM) AUC (ug•hr/ml) Tmax T1/2 (Days)
Ado-trastuzumab-emtansine Kadcyla 3.6 mg/kg IV 90-min 0.572 19.8 end-of-infusio 4.2
  free--DM1 (Kadcyla) - - - - 0.006 - - -
Aldesleukin Proleukin- 1.1 mg SC - 0.00159 0.0157 2.5-h 0.071
Alemtuzumab Campath- 30 mg IV 2-h 0.07356 - - 6
Atezolizumab- Tecentriq 1200 mg IV 60-min 2.79000 - - 27
Asparaginase-(E.-coli) Elspar 5000 U/m2 IV 30-min 0.51684 307 - 0.771
Asparaginase-(Erwinia-chrysanthemi) Erwinaze 30,000 IU/m2 IV 3-h 20-IU/ml - - 0.267
Bevacizumab Avastin- 10 mg/kg IV 30-min 1.90604 - - 21
Blinatumomab Blincyto 28 ug IV 24-h 0.000011 - - 0.0875
Brentuximab-Vedotin Adcetris 1.8 mg/kg IV 30-min 0.20915 3.19 0.089-d 4.43
  free-MMAE (Adcetris) - - - - 0.00696 0.0015 2.09-d
Cetuximab Erbitux- 400 mg/m2 IV 2-h 1.40622 19000 3-h 3.13
Daratumumab Darzalex 16 mg/kg IV 6.5-h 6.18243 - - 18
Denileukin-Diftitox Ontak 19 ug/kg IV 60-min 1309-U/ml 33482-U•min/ml - 0.056
Denosumab Prolia 60 mg SC - 0.04592 13.2 10-d 25.4
Denosumab Xgeva- 180 mg SC - 0.21156 55 10-d 29.1
Dinutuximab Unituxin 17.5 mg/m2 IV 10-20-h 0.07667 - - 10
Elotuzumab- Empliciti 10 mg/kg IV 15-60-min 2.27819 40701 4.1-h 0.197
Filgrastim Neupogen 5 ug/kg IV 15-30-min 0.01004 0.638 - 0.463
Gemtuzumab-Ozogamicin Mylotarg- 9 mg/m2 IV 2-h 0.01869 123 - 3.02
 free-calichaemicin (Mylotarg) - - - - 0.00365 0.22 - 4.21
Interferon-alfa-2b Intron-A 20×106 IU IV 20-min 0.00031 0.00950 - -
Ipilimumab Yervoy- 3 mg/kg IV 90-min 0.54730 - - -
Necitumumab Portrazza 800 mg IV 60min 3.51519 67821 - 5.21
Nivolumab Opdivo 3 mg/kg IV 60 min 0.41034 15813 3.1 h 17
Obinutuzumab Gazyva 1000 mg IV see label 3.75770 366 - 28.4
Ofatumumab Arzerra 2000 mg IV see label 10.14374 674463 - 15.8
Olaratumab Lartruvo 15 mg/kg IV 60 min 3.09700 42400 2.53 h 7.21
Oprelvekin Neumega 50 ug/kg SC - 0.00100 0.242 2.7 h 0.3375
Palifermin Kepivance 90 ug/kg IV bolus 0.12228 0.232 - 0.196
Panitumumab Vectibix 6 mg/kg IV 60 min 1.44898 54.4 7.5 d -
Pegaspargase Oncaspar 2500 IU/m2 IV 1–2 hr 0.35740 - - 7
Pegfilgrastim Neulasta 100 ug/kg SC - 0.00587 29.3 48 h 0.883
Peginterferon alfa-2b PEG-lntron 6 ug/kg SC - 0.00014 0.43 - 2.125
Pembrolizumab Keytruda 2 mg/kg IV 30 min 0.44090 58.7 0.17d 26
Pertuzumab Perjeta 420 mg IV 30–60 min 1.01351 2762 - 19.1
Ramucirumab Cyramza 8 mg/kg IV 60 min 1.16327 18300 - 7.54
Rasburicase Elitek 0.2 mg/kg IV 30 min 0.11375 45.2 - 0.879
Rituximab Rituxan 375 mg/m2 IV see label 3.22397 - - -
Romiplostim Nplate 1 ug/kg IV bolus 0.00022 0.0267 - 0.1
Siltuximab Sylvant 11 mg/kg IV 60 min 2.28966 - - 20.6
Thyrotropin alfa Thyrogen 0.9 mg IM - 0.00122 - 10 h 1.04
Trastuzumab Herceptin 6 mg/kg IV 90 min 1.48422 - - 12
Ziv-Aflibercept Zaltrap 4 mg/kg IV 1 h 1.00516 12.2 - 5.5

Abbreviations: Cmax: maximum plasma concentration; AUC: Area Under the time-plasma Concentration curve; : Tmax: Time post-dose of maximum plasma concentration; : T1/2: Plasma half-life

Table 4.

Key human pharmacokinetic parameters for selected small-molecule drugs NOT approved for oncology indications

Generic Name Brand Name Dose Dose Unit Route infusion Cmax (uM) Cmax (ng/ml) AUC (ng•hr/ml) Tmax (hr) T1/2 (hr) Protein binding
Alfuzosin UroXatral 10 mg PO - 0.035 14 194 8.0 10 82–90%
Aminoglutethimide Cytadren 500 mg PO - 25.4 5900 - 1.5 12.5 21–25%
Celecoxib Celebrex 400 mg PO - 4.60 1752 13049 - 8.8 97%
Chloroquine Aralen 750 mg PO - 0.725 232 8385 3.7 - 55%
Dutasteride Avodart 0.5 mg PO - 0.076 40 - 2–3 840 99%
Finasteride Proscar 5 mg PO - 0.124 46 389 1.8 6 90%
Histrelin acetate Supprelin 0.0567 mg/day SC 0.00083 1.1 2318 12 - 70%
Hydroxychloroquine Plaquenil 200 mg PO - 0.35000 117.4 12015 3.8 564 45%
Ibandronate Boniva 6 mg IV 30 min 1.02 327 942 0.63 12 86%
Medroxyprogesterone acetate Provera 20 mg PO - 0.0026 1.0 6.95 2.7 12.1 90%
Metformin Glucophage 1500 mg PO - 24.0 3100 18400 1.5 5.98 negligible
Quinacrine Acrichine 100 mg PO - 0.300 120 - - - 80–90%
Sildenafil Viagra 100 mg PO - 0.794 377 1295 1.0 2.76 96%
Sirolimus (Rapamycin) Rapamune 2 mg PO - 0.016 15 230 3.5 62 92%
Tacrolimus Prograf 5 mg PO - 0.037 30 243 1.6 34.8 99%
Zalcitabine (Dideoxycytidine) Hivid 1.5 mg PO - 0.119 25 72 0.80 - <4%

Abbreviations: Cmax: maximum plasma concentration; AUC: Area Under the time-plasma Concentration curve; : Tmax: Time post-dose of maximum plasma concentration; : T1/2: Plasma half-life

Each table is sorted alphabetically by the unique generic name. Also provided is one proprietary (brand) name; some drugs are sold under multiple brands, particularly outside of the U.S. market. The dose and route of administration from which the PK data are derived is shown, which is typically the highest dose recommended in the label. For drugs administered IV, the duration of injection is included. The maximum plasma concentration (Cmax) is usually reported as ng/ml, from which we calculated micromolar concentration units. Also collected are the time of maximum plasma concentration (Tmax) and the plasma half-life (T1/2). The area under the plasma concentration-time curve (AUC) is shown, following conversion to a consistent unit of ng•hr/ml. The raw values and units for Cmax and AUC as reported in the cited reference are included in the Supplementary Tables. The fraction bound to plasma protein is also included, as this is an important parameter to consider when translating from in vivo settings to in vitro systems with varied protein composition.

A few agents included in Table 1 have been discontinued, and some older agents are no longer listed in the FDA “Orange Book” (8). Although these agents are no longer marketed in the U.S., they may be used experimentally, particularly in drug combination studies, so the PK data for these drugs have been included.

Several agents are intentionally administered as a prodrug that is converted in vivo into the active drug; these are summarized in Table 2. In these cases, the metabolite carries the predominant pharmacological activity, so the levels of these metabolites following recommended doses of the parent have been reported. For abiraterone acetate, fludarabine phosphate and lomustine, the levels of the parent prodrug were below the limit of quantitation in vivo, so only the levels of the active metabolite are shown. Three drugs are precursors of the cytotoxic pyrimidine, 5-fluorouracil: floxuridine, capecitabine and tegafur. Each of these is converted to 5-FU in the liver and other tissues. Because plasma 5-FU levels following floxuridine may be as high as the parent, it was considered a prodrug and included in Table 2. Two compounds (dacarbazine and temozolomide) are precursors of the same active species, N-demethyldacarbazine (MTIC). Dacarbazine is converted intracellularly to MTIC through cytochrome P450 oxidation (9), with little accumulation of the active metabolite in plasma, so only exposure of the parent is shown in Table 1. However, temozolomide is rapidly converted non-enzymatically to MTIC at physiological pH, resulting in readily detectable plasma exposure of the active metabolite, so MTIC levels following temozolomide administration are included in Table 2. In one instance, mechlorethamine, the parent molecule undergoes such rapid chemical transformation that plasma levels of the parent drug were difficult to measure reliably, and a Cmax could not be determined.

Table 3 presents a summary of the human PK parameters reported for all 40 biological therapeutics approved for cancer indications in the U.S, excluding vaccines and radiologicals. The table is sorted alphabetically by unique generic name, followed by proprietary brand name. As in Table 1, the dose and route of administration are given, followed by the Cmax provided as micromoles/liter and the integrated area under the plasma concentration-time curve (AUC). The time of maximum plasma concentration (Tmax) and the plasma half-life (T1/2) are also shown, where reported, as well as the duration of injection for drugs administered IV.

The 16 drugs included in Table 4 are approved for other indications, but are not specifically approved for use in cancer, although finasteride, dutasteride and alfuzosin have been approved to treat benign prostatic hyperplasia. All of these agents are currently undergoing clinical trials for cancer indications, thus the PK data will be useful to researchers and have been included.

An expanded Table is available for download in the supplemental materials which combines all small molecules in Tables 1, 2 and 4 merged into a single, sortable spreadsheet (Supplemental Table 1). A separate file containing the biological agents from Table 3 is also provided (Supplemental Table 2). These are Excel files that include full references for the primary sources of the PK values and the official Drug Product Labels. Annotations of the molecular target (if known), the Cmax and AUC in raw units as reported in the reference, the plasma clearance (Cl) and volume of distribution (Vd) are included as available. The year of initial approval in the U.S. is shown, along with the indication approved for use in the U.S.

The Cmax values reported here represent the peak exposures observed at the highest clinically recommended doses delivered as a single administration (except where noted). In the clinic, most agents are typically given by repeated administration which may lead to accumulation, so some agents may achieve higher exposures at steady-state. With IV administration, the Cmax is typically reported at the end of the infusion, but is sometimes reported as C0, a calculated concentration extrapolated from the plasma concentration-time curve to time zero. The Cmax following IV administration is highly dependent on the duration of the injection, so the recommended injection duration is provided in the tables for reference. The Cmax and AUC values are presented as total plasma exposure (bound and unbound) of the parent molecule only, without consideration of the presence of active metabolites. It is important to note that the Cmax and AUC values presented here are average values, and interindividual variability can be quite large due to genetic polymorphisms in clearance and other factors (e.g., Mercaptopurine).

A few alternative formulations of equivalent active ingredients have been included in this compilation. For example, the nanoparticle formulation of paclitaxel (nab-paclitaxel; Abraxane®) has recently been approved, and allows shorter infusion times and higher doses to be delivered compared to paclitaxel (Taxol®). A suspension formulation of mercaptopurine (Purixan®), which offers greater dosing flexibility than the original tablet, was approved in 2014. Liposomal formulations of doxorubicin (Doxil®), irinotecan (Onivyde®) and vincristine sulfate (Marqibo®) have been approved. These formulations have distinct PK properties compared to the original dosage forms, so have been included in Table 1.

Three biological agents are available both as the native compound and as polyethylene glycol (PEG) conjugates: asparaginase (pegaspargase; Oncaspar®), filgrastim (pegfilgrastim; Neulasta®) and interferon alpha-2b (peginterferon; Sylantron®, PEGintron®). Pegylation retains the biological activity, but alters the molecular form of the drug; it is not processed in vivo to release the parent biological agent, so these are not prodrugs. Hence, these were regarded as independent species distinct from the parent compound, and have been included separately in Table 3. Three biological agents are antibody-drug conjugates (ADC), with cytotoxic agents covalently bound to an antibody (ado-tratuzumab emtansine, bretuximab vedotin, gemtuzumab ozogamicin). In these cases, the plasma levels of the free cytotoxic agents are also included in Table 3.

The plasma Cmax and AUC were considered to be the key pharmacokinetic parameters to enable translation of clinical drug exposure to a nonclinical study application. The Cmax data were found for all but 2 of the 145 unique small molecule drugs or their active metabolites listed in Tables 1 and 2; the AUC was not found for 21 of these. Two agents (Ingenol and Mechlorethamine) were reported as Below the Limit of Quantitation (BLOQ). Additional pharmacokinetic parameters (including half-life, Tmax, clearance, volume of distribution) were included when available, but not all of these parameters were reported for every agent. For the biological agents, Cmax was found for all 40 unique drugs listed in Table 3 and AUC was found for 27 of these.

Plasma protein binding of small molecule drugs varies widely across agents (and occasionally between species) and can have significant impact on plasma free drug concentrations. This can be an important factor when designing nonclinical studies to examine drug mechanisms, particularly for in vitro studies, so plasma protein binding data have been included for all but 14 of the small molecule drugs in Tables 1, 2 and 4. Protein binding of biological agents was not considered.

Discussion

Translational medicine can be aided greatly by the establishment of pharmacokinetic-pharmacodynamic (PK/PD) relationships in nonclinical models (1013). A fundamental aspect of translational studies is the determination of the concentrations of drug that are likely to be observed in clinical use. Attempts to translate doses or plasma exposures from nonclinical models to humans most often utilize allometric scaling (14,15), in vivo PK (16,17) or PK/PD and physiologically-based pharmacokinetic (PBPK) models to bridge doses from animal studies to humans (13,18,19). Very few studies have incorporated a “reverse translation” of clinical exposure data to aid design of studies in nonclinical oncology models. When testing approved oncology drugs in nonclinical studies to explore expanded cancer indications, awareness of clinically achievable exposure can facilitate study design. Similarly, when attempting to repurpose approved agents from their original indications to use in oncology (e.g., metformin, celecoxib, sirolimus), it is valuable to have an appreciation of clinically relevant exposures to assist translation to nonclinical models. For in vivo studies, these human exposures can be used to help determine the appropriate doses to use in model species, either by allometric scaling or empirical measurement. Allometric scaling is frequently based on body surface area, although this practice has limitations (20). However, direct comparison of plasma exposure associated with a measured clinical activity in humans (a clinical pharmacodynamic response) to an exposure observed in nonclinical models which used doses and routes of administration that differ from clinical use can assist the interpretation of pharmacodynamic results in the animal model. In this regard, Spilker et al. (21) have recently proposed a rigorous strategy by which human PK parameters can be utilized in conjunction with mouse PK studies to determine the doses and routes of administration that can most closely mimic the clinically relevant exposures in the animal model.

The plasma Cmax is highly dependent on route of administration, formulation and physical properties of the drug. It provides an indication of the highest concentration that the subject is exposed to during therapy, and the Cmax may be considered as an upper limit for drug concentration during in vitro studies or the highest plasma exposure for in vivo studies to minimize off-target effects. During in vitro studies, it is often possible to increase the drug concentration to levels far in excess of what could be achieved in vivo. However, testing targeted agents at concentrations ten or one hundred times greater than the IC50 or Ki for the molecular target increases the possibility of introducing off-target activities unrelated to the clinical benefit (1), or from on-target activity (enzyme inhibition, receptor occupancy) that is not realistically achievable in the clinic. Either of these situations can lead to a misinterpretation of responses in nonclinical studies. Furthermore, the Cmax is maintained only transiently for most compounds, and sustained levels well below Cmax may be sufficient to achieve therapeutic efficacy. In cases where a pharmacodynamic response is tightly linked to exposure, it may be important to maintain a minimum plasma concentration to sustain inhibition of the target (e.g., receptor occupancy, enzyme inhibition) above a certain threshold. In other cases, particularly when the drug interacts with the target irreversibly (e.g., several alkylating agents; afatinib), the duration of the pharmacodynamic effect (target binding) is uncoupled from the plasma PK and can be much longer than the plasma half-life of the drug (22), so the Cmax may be more directly related to efficacy than AUC.

Differences in free drug levels due to protein binding can be critical for translation of clinical exposures to nonclinical models, particularly during in vitro studies. For small-molecule drugs, plasma drug analysis is typically performed following organic extraction of samples, and the reported values represent total plasma concentration (free + protein bound) rather than free (unbound) drug. Since the free (unbound) drug is generally the species that interacts with the molecular target, reduction in free drug levels due to protein binding can dramatically alter the concentration of drug available for interaction with the target during in vitro studies (i.e., cell culture or biochemical assays). Discrepancies between free drug concentrations in vitro in cell culture media containing 5–10% bovine serum and in vivo (e.g., human plasma) will be dependent on the degree of protein binding for each drug and the binding capacity of the added protein/serum.

Species differences in plasma protein binding may warrant consideration when designing translational studies, as some drugs show clinically significant species differences in plasma protein binding. For example, at clinically relevant concentrations, vismodegib is primarily bound to α-1-acid glycoprotein (AAG), with a much lower affinity for albumin (23). Quantitative assays of protein binding revealed an approximately 100-fold difference in binding Kd between rat and human AAG, which resulted in significant PK differences between species. In general, any species differences in binding affinity to the target should also be factored into the calculation of the concentrations deemed to be equivalent to human clinical exposure.

Plasma AUC is another key parameter to consider in planning nonclinical studies, particularly when comparing exposures between species or routes of administration. The AUC is the integration of plasma drug exposure over time, and as such takes into account bioavailability, different absorption rates (e.g., IV vs oral) and elimination rates. This provides a more complete picture of drug exposure than Cmax, which represents exposure at only the Tmax. While AUC can be useful to compare exposure following different routes of administration or formulations, it is particularly useful to translate the exposure achieved in humans to that seen in animal models. Modifying the dose or route in animals to mimic more closely the AUC observed in the clinic, using a protocol such as suggested by Spilker et al. (21), may provide a more relevant drug exposure in the model and help to avoid high exposures which would not be tolerated in humans or that may lead to off-target activities of the drug.

In cases where active metabolites contribute significantly to efficacy, the concentration of those metabolites in plasma may need to be monitored to capture the exposure responsible for the full pharmacological activity of the administered product. One example of this is tamoxifen. Through the action of two cytochrome P450 oxidases (CYP2D6 and CYP3A4/5), three metabolites are produced with affinities for the estrogen receptor that are similar to or more potent than the parent molecule (2427). In patients, these metabolites may be responsible for much of the pharmacodynamic action of the drug (24,25). It has been shown that mice can produce these metabolites (28), but levels and metabolite profile vary with dose. However, these metabolites may not be produced in all in vitro systems under test (e.g., cell culture, biochemical assays), so activity due to parent alone may not reflect the full potential in vivo activity of the drug. In addition, polymorphisms in cytochrome P450 enzymes responsible for activation (or degradation) can influence the plasma concentrations of parent and metabolites. Hence, when testing tamoxifen in nonclinical models, levels of parent and these active metabolites should be considered. As can be seen from this example, the interpretation of results is complex in nonclinical models where active metabolites of the parent have the potential to contribute significantly to the pharmacological action.

Several alkylating agents undergo activation to the reactive species in vivo and this activation should be considered when designing nonclinical studies. Activation of these agents is described in the Drug Product Labels. The platinum-containing drugs (carboplatin, cisplatin and oxaliplatin) are subject to aquation in water, which will occur in most in vitro and in vivo systems to generate the active agents. Temozolomide undergoes rapid non-enzymatic hydrolysis to MTIC, which is present at about 3% of parent levels in plasma. Similar to temozolomide, busulfan undergoes non-enzymatic hydrolysis in aqueous media to become activated, releasing methanesulfonate groups. Three alkylating agents are activated by cytochrome P450 enzymes (altretamine, cyclophosphamide, ifosfamide). For the agents requiring enzymatic activation, evaluation in nonclinical models (in vitro and in vivo) should ensure that the appropriate P450 enzymes are present within the system to allow full activity to be manifest. Three drugs (aminolevulinic acid, methoxysalen and porfimir) require photoactivation, which yields free radicals or derivatives that form covalent bonds with nucleic acids and proteins, in order to generate cytoxicity.

The most common routes of administration for oncology drugs in the clinic are oral and IV. In animal studies, particularly in rodents, the IP route is often preferred. Following IP injection, many small molecule drugs are absorbed by capillaries within the visceral peritoneum, which collect into the mesenteric and omental veins and drain into the hepatic portal vein. Drugs absorbed through this route will be subject to first-pass hepatic metabolism, similar to orally administered drugs (29). For certain high molecular weight drugs, such as biologics, and some lipophilic small molecules, absorption into the lymphatic drainage predominates, thereby avoiding first-pass hepatic clearance (30). Some biological agents are subject to target-mediated clearance, in addition to the the hepatic and renal clearance mechanisms more typical with small molecule drugs. The formulation of drugs (vehicle and excipients) can have a significant effect on the rate and extent of absorption following either IP or oral delivery. In animal studies, oral drugs are often delivered as suspensions. While suspensions are generally tolerated for oral administration, drugs delivered IP should be fully solubilized. All of these factors can influence Cmax and AUC, so should be considered during the design and interpretation of nonclinical studies.

Our goal in this compilation is to provide a convenient data resource of PK estimates for clinically relevant plasma exposures (Cmax and AUC) for all single agents marketed for oncology indications in the U.S. We chose the highest dose recommended in the label delivered as a single administration (except where noted), the intent being to provide a benchmark for achievable and relevant human exposures. The therapeutic effects of many of the agents are likely due to repeated administration of these doses using a variety of schedules which may lead to accumulation, so some agents may achieve higher exposures at steady-state. Consideration of these clinically achievable exposures in conjunction with animal studies to characterize the PK in the model species, such as described by Spilker et al. (21) and including dose-response curves, will ultimately improve the translation of pharmacological activity in the model back to the clinic. We expect the greatest utility of this report will be a source from which an upper boundary on the clinically achievable plasma concentrations of anti-cancer agents can be readily applied to in vitro studies. Admittedly, there are limitations and caveats associated with any attempt to reduce something as complex as efficacy and human pharmacokinetics down to a single approach, and successful application of these clinical exposure values will also require a thorough understanding of the underlying biology of the target and disease processes.

Supplementary Material

1
2

Acknowledgments

Financial Support: supported by the Developmental Therapeutics Program in the Division of Cancer Treatment and Diagnosis of the National Cancer Institute.

Footnotes

The authors declare no potential conflicts of interest.

The content of this publication does not necessarily reflect the views or policies of the Department of Health and Human Services, nor does mention of trade names, commercial products, or organizations imply endorsement by the U.S. Government.

References

  • 1.Smith MA, Houghton P. A proposal regarding reporting of in vitro testing results. Clin Cancer Res. 2013;19(11):2828–33. doi: 10.1158/1078-0432.CCR-13-0043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.National Cancer Institute. [Accessed 2017 02/17/2017];A to Z List of Cancer Drugs. 2017 Feb 17; < https://www.cancer.gov/about-cancer/treatment/drugs>.
  • 3.MediLexicon. [Accessed 2017 02/17/2017];FDA Approved Drugs » Cancer & Oncology. 2017 Feb 17; < http://www.medilexicon.com/drugs-list/cancer.php>.
  • 4.Centerwatch. [Accessed 2017 02/17/2017];FDA Approved Drugs for Oncology. 2017 Feb 17; < http://www.centerwatch.com/drug-information/fda-approved-drugs/therapeutic-area/12/oncology>.
  • 5.U.S. Food and Drug Administration. [Accessed 2017 02/17/2017];Drugs@FDA: FDA Approved Drug Products. 2017 Feb 17; < http://www.accessdata.fda.gov/scripts/cder/daf/index.cfm>.
  • 6.National Library of Medicine DailyMed. [Accessed 2017 02/17/2017];DailyMed. 2017 Feb 17; < https://dailymed.nlm.nih.gov/dailymed/index.cfm>.
  • 7.Thompson-Reuters PS. [Accessed 2017 02/17/2017];Integrity®. 2017 Feb 17; < https://integrity.thomson-pharma.com/integrity/xmlxsl/pk_self_reg.show_initial_page>.
  • 8.U.S. Food and Drug Administration. [Accessed 2017 02/17/2017];Approved Drug Products with Therapeutic Equivalence Evaluations. 2017 Feb 17; < http://www.accessdata.fda.gov/scripts/cder/ob/>.
  • 9.Reid JM, Kuffel MJ, Miller JK, Rios R, Ames MM. Metabolic Activation of Dacarbazine by Human Cytochromes P450: The Role of CYP1A1, CYP1A2, and CYP2E1. Clinical Cancer Research. 1999;5:2192–7. [PubMed] [Google Scholar]
  • 10.Peterson JK, Houghton PJ. Integrating pharmacology and in vivo cancer models in preclinical and clinical drug development. European Journal of Cancer. 2004;40(6):837–44. doi: 10.1016/j.ejca.2004.01.003. [DOI] [PubMed] [Google Scholar]
  • 11.Kumar R, Knick VB, Rudolph SK, Johnson JH, Crosby RM, Crouthamel MC, et al. Pharmacokinetic-pharmacodynamic correlation from mouse to human with pazopanib, a multikinase angiogenesis inhibitor with potent antitumor and antiangiogenic activity. Mol Cancer Ther. 2007;6(7):2012–21. doi: 10.1158/1535-7163.MCT-07-0193. [DOI] [PubMed] [Google Scholar]
  • 12.Haznedar JO, Patyna S, Bello CL, Peng GW, Speed W, Yu X, et al. Single- and multiple-dose disposition kinetics of sunitinib malate, a multitargeted receptor tyrosine kinase inhibitor: comparative plasma kinetics in non-clinical species. Cancer Chemother Pharmacol. 2009;64(4):691–706. doi: 10.1007/s00280-008-0917-1. [DOI] [PubMed] [Google Scholar]
  • 13.Yamazaki S, Skaptason J, Romero D, Vekich S, Jones HM, Tan W, et al. Prediction of oral pharmacokinetics of cMet kinase inhibitors in humans: physiologically based pharmacokinetic model versus traditional one-compartment model. Drug Metab Dispos. 2011;39(3):383–93. doi: 10.1124/dmd.110.035857. [DOI] [PubMed] [Google Scholar]
  • 14.Reagan-Shaw S, Nihal M, Ahmad N. Dose translation from animal to human studies revisited. FASEB J. 2008;22(3):659–61. doi: 10.1096/fj.07-9574LSF. [DOI] [PubMed] [Google Scholar]
  • 15.Tang H, Mayersohn M. A novel model for prediction of human drug clearance by allometric scaling. Drug Metab Dispos. 2005;33(9):1297–303. doi: 10.1124/dmd.105.004143. [DOI] [PubMed] [Google Scholar]
  • 16.Obach RS. Predicting clearance in humans from in vitro data. Curr Top Med Chem. 2011;11(4):334–9. doi: 10.2174/156802611794480873. BSP/CTMC/E-Pub/-00027-11-5 [pii] [DOI] [PubMed] [Google Scholar]
  • 17.Obach RS, Baxter JG, Liston TE, Silber BM, Jones BC, MacIntyre F, et al. The prediction of human pharmacokinetic parameters from preclinical and in vitro metabolism data. J Pharmacol Exp Ther. 1997;283(1):46–58. [PubMed] [Google Scholar]
  • 18.Hoshino-Yoshino A, Kato M, Nakano K, Ishigai M, Kudo T, Ito K. Bridging from preclinical to clinical studies for tyrosine kinase inhibitors based on pharmacokinetics/pharmacodynamics and toxicokinetics/toxicodynamics. Drug Metab Pharmacokinet. 2011;26(6):612–20. doi: 10.2133/dmpk.DMPK-11-RG-043. [DOI] [PubMed] [Google Scholar]
  • 19.Hosea NA, Collard WT, Cole S, Maurer TS, Fang RX, Jones H, et al. Prediction of human pharmacokinetics from preclinical information: comparative accuracy of quantitative prediction approaches. J Clin Pharmacol. 2009;49(5):513–33. doi: 10.1177/0091270009333209. [DOI] [PubMed] [Google Scholar]
  • 20.Mathijssen RH, de Jong FA, Loos WJ, van der Bol JM, Verweij J, Sparreboom A. Flat-fixed dosing versus body surface area based dosing of anticancer drugs in adults: does it make a difference? Oncologist. 2007;12(8):913–23. doi: 10.1634/theoncologist.12-8-913. [DOI] [PubMed] [Google Scholar]
  • 21.Spilker ME, Chen X, Visswanathan R, Vage C, Yamazaki S, Li G, et al. Found in Translation: Maximizing the Clinical Relevance of Nonclinical Oncology Studies. Clin Cancer Res. 2017;23(4):1080–90. doi: 10.1158/1078-0432.CCR-16-1164. [DOI] [PubMed] [Google Scholar]
  • 22.Singh J, Petter RC, Baillie TA, Whitty A. The resurgence of covalent drugs. Nat Rev Drug Discov. 2011;10(4):307–17. doi: 10.1038/nrd3410. [DOI] [PubMed] [Google Scholar]
  • 23.Giannetti AM, Wong H, Dijkgraaf GJ, Dueber EC, Ortwine DF, Bravo BJ, et al. Identification, characterization, and implications of species-dependent plasma protein binding for the oral Hedgehog pathway inhibitor vismodegib (GDC-0449) J Med Chem. 2011;54(8):2592–601. doi: 10.1021/jm1008924. [DOI] [PubMed] [Google Scholar]
  • 24.Jordan VC, Collins MM, Rowsby L, Prestwich G. A monohydrxylated metabolite of tamoxifen with potent antiestrogenic activity. J Endocr. 1977;75:305–16. doi: 10.1677/joe.0.0750305. [DOI] [PubMed] [Google Scholar]
  • 25.Jordan VC, Bain Richard R, Brown Raymond R, Gosden Barbara, Santos MA. Determination and Pharmacology of a New Hydroxylated Metabolite of Tamoxifen Observed in Patient Sera during Therapy for Advanced Breast Cancer. Cancer Res. 1983;43:1446–50. [PubMed] [Google Scholar]
  • 26.Jin Y, Desta Z, Stearns V, Ward B, Ho H, Lee KH, et al. CYP2D6 genotype, antidepressant use, and tamoxifen metabolism during adjuvant breast cancer treatment. J Natl Cancer Inst. 2005;97(1):30–9. doi: 10.1093/jnci/dji005. [DOI] [PubMed] [Google Scholar]
  • 27.Dickschen K, Willmann S, Thelen K, Lippert J, Hempel G, Eissing T. Physiologically Based Pharmacokinetic Modeling of Tamoxifen and its Metabolites in Women of Different CYP2D6 Phenotypes Provides New Insight into the Tamoxifen Mass Balance. Front Pharmacol. 2012;3:92. doi: 10.3389/fphar.2012.00092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Robinson SP, Langan-Fahey SM, Jordan VC. Implications of Tamoxifen Metabolism in the Athymic Mouse for the Study of Antitumor Effects upon Human Breast Cancer Xenografts. Eu J Cancer Clin Oncol. 1989;25(12):1769–76. doi: 10.1016/0277-5379(89)90347-7. [DOI] [PubMed] [Google Scholar]
  • 29.Lukas G, Brindle SD, Greengard P. The route of absorption of intraperitoneally administered compounds. J Pharmacol Exp Ther. 1971;178(3):562–6. [PubMed] [Google Scholar]
  • 30.Lu Z, Wang J, Wientjes MG, Au JL-S. Intraperitoneal Therapy for Peritoneal Cancer. Future Oncol. 2010;6(10):1625–41. doi: 10.2217/fon.10.100. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1
2

RESOURCES