Table 1.
In Vitro to In Vivo translational model.
| In Silico | In Vitro Assay | Examples of In Vitro to In Vivo Translation | ||||
|---|---|---|---|---|---|---|
| Technologies Used in Examples | Parameter | In Vitro to In Vivo Translation Result | Application in Drug Development | |||
| PBPK | Absorption | Caco-2 [81], MDCK [82], gut MPS [83] | MDCK-MDR1 and Caco-2 [84] | Obtaining half maximal inhibitory concentration (IC50) for P-gp and integrating it to models | Demonstrating non-interaction between Axitinib and P-gp substrate | Prediction of drug-drug interaction and exemption of related clinical trials |
| Distribution | MDCK [85], hiPSC- brain endothelial cells [86], co-culture [87] | MDCK Ⅱ [88] | Using apparent permeability coefficient (Papp) to obtain in vitro efflux transporter-mediated clearance and scaling it to the whole-brain in vivo efflux transporter-mediated clearance | Exploring the penetration of AZD1775 across BBB | Prediction of drug distribution and target concentration | |
| Metabolism | Recombinant enzymes [89], microsomes [90], primary hepatocytes [91], HepG2 [92], HepaRG [93], hESC or hiPSC-hepatocytes [94], liver MPS [95] | Primary hepatocytes [96] | Inputting the intrinsic clearance (CLint) to Simcyp software to establish PBPK model | Predicting the difference of AUC in patients with different liver damage after a single oral administration of sirolimus | Prediction of drug metabolism and inter-population extrapolations | |
| Excretion | MDCK, CHO, HEK-293, HeLa [97], primary cultured renal tubule cells [98], renal MPS [99] | renal MPS [99] | Scaling renal clearance (CLR) based on surface area | Predicting human renal excretion for cisplatin and nicotine | Prediction of excretion | |
| PBPK | Integrate ADME | MPS [99] | MPS [99] | Scaling intestinal permeability (Papp) based on absorptive surface, liver clearance (CLint, in vivo) based on the number of hepatocytes, renal clearance (CLR) based on surface area | Reproducing the clinical PK profiles for both nicotine and cisplatin at different doses and different routes of administration | Simulation of clinical PK profiles |
| PK/PD | Disease-related cell [100], 2D [80,101], 3D [102], MPS [103], organoids [104] | Six human epithelial cancer cell lines [100] | Directly combining maximal killing rate (Kmax), drug concentrations yielding 50% of Kmax (KC50) and hill index (γ) into in vivo model | Demonstrating that low doses and high dosing frequency for paclitaxel is prior to maximum tolerated doses | Dose and schedule selection | |
| L540cy cells, Karpas cells [99] | Integrating association and dissociation rate constants (Kon and Koff) to describe the interaction between ADC and target | Predicting therapy in clinical trials employing different dosing regimens | Clinical response prediction | |||
| primary liver cells, red blood cells and brain homogenates [101] | Based on the total enzyme content, scaling metabolic capacity (Vmax) and clearance (CLint); Correcting bimolecular inhibition constant (Ki) considering different states of targets in vitro and in vivo | Evaluating the biotoxicity of carbaryl and other carbamates with an anticholinesterase mode of action | Toxicity prediction | |||
| MPS [105] | Based on the number of nephrons in human kidney, scaling maximal injury rate (Emax) and drug concentrations yielding 50% of Emax (EC50) into in vivo model | Assessing renal proximal tubule injury caused by three nephrotoxic drugs | Toxicity prediction | |||
| QSP (QST) | Disease-related cell [106], 2D [106,107], 3D [107], MPS [108], organoids [109] | Primary hepatocytes [106] | Applying directly the IC50 values for the bile acid transporters to DILIsym, fitting the mitochondrial toxicity parameters (Vmax, Km) in MITOsym, and converting them to DILIsym | Explaining the liver toxicity mechanism of PF-04895162 and expound the differences of species | Characterization of target mechanism | |
| JIMT-1 cells in 2D or 3D and dynamic cell Culture [107] | Integrating drug inhibition or stimulation coefficient (S1p, S2p, Kp etc.) to describe signal pathway molecules perturbation | Optimizing the sequence and inter-dose interval of the three agents (paclitaxel, dasatinib, and everolimus) in the combination | Design of drug administration protocol and evaluation of drug combination | |||
| effector T cells (Teffs), EL4 and E.G7-OVA thymoma cells [110,111,112] | Integrating rate constants defining the half-life of engagement or dissociation between cancer cells and effector T cells (CancerTEng, CancerTInt) directly into the QSP model; scaling number of CD28 receptors expressed on each T cell during priming (CD28_receptors-per-Tcell) by the number of T cell in vivo | Predicting the checkpoint inhibitors’ therapies administered as mono-, combo- and sequential therapies | Clinical response prediction | |||
Abbreviations: Caco-2: human colon adenocarcinoma cells; MDCK: Madin–Darby canine kidney epithelial cells; MPS: microphysiological systems; hiPSC: human-induced pluripotent stem cell; PBPK: physiologically based pharmacokinetic model; PK/PD: pharmacokinetic/pharmacodynamic model; QSP: quantitative systems pharmacology model; QST: quantitative systems toxicology model; hESG: human embryonic stem cell lines; CHO: Chinese hamster ovary cells; HEK-293: human embryonic kidney cells.