Elsje Pienaar 1,2, Jansy Sarathy 3, Brendan Prideaux 3, Jillian Dietzold 4, Véronique Dartois 3, Denise E. Kirschner 2 and Jennifer J.

Similar documents
Louis Joslyn Dept of Bioinformatics With Dr. Denise Kirschner and Dr. Jennifer Linderman

A dynamic integrated drug, Mtb and host archetype for testing novel treatment interventions John I Fors

1 + denotes a higher variable value for the deletion or depletion than the control scenario; - denotes a lower value.

A METAPOPULATION MODEL OF GRANULOMA FORMATION IN THE LUNG DURING INFECTION WITH MYCOBACTERIUM TUBERCULOSIS. Suman Ganguli.

Multi-scale model of Mycobacterium tuberculosis infection maps metabolite and gene

Synergy between Individual TNF-Dependent Functions Determines Granuloma Performance for Controlling Mycobacterium tuberculosis

The general Concepts of Pharmacokinetics

The Effects of HIV 1 Infection on Latent Tuberculosis

What He Said: Rifampin versus Rifapentine

Gap identification as a critical step to enable integrated clinical trial simulation platforms

No Triage! Every submission reviewed by practicing scientists

PHA Final Exam Fall 2006

Pharmacodynamics: the methods

Systems Pharmacology Approach Toward the Design of Inhaled Formulations of Rifampicin and Isoniazid for Treatment of Tuberculosis

Penetration of rifampin and rifapentine into diseased lung in the rabbit cavity pulmonary disease model of TB

PHARMACOKINETIC & PHARMACODYNAMIC OF ANTIBIOTICS

PHA First Exam Fall 2003

Clinical Pharmacology. Pharmacodynamics the next step. Nick Holford Dept Pharmacology & Clinical Pharmacology University of Auckland, New Zealand

Basic Concepts of PK/PD -pharmacodynamic indices- Johan W. Mouton MD PhD FIDSA Professor pharmacokinetics and pharmacodymamics

MONTE CARLO SIMULATION & PK-PD TARGET ATTAINMENT ANALYSIS:

Overview. Therapeutic Window. Does drug concentration matter? Treatment of Tuberculosis Therapeutic Drug Monitoring

ZIN EN ONZIN VAN ANTIBIOTICASPIEGELS BIJ NEONATEN

Treatment of Tuberculosis Therapeutic Drug Monitoring

ICU Volume 11 - Issue 3 - Autumn Series

Cell Responses in Lymph Nodes

Modeling Intercellular Interactions in Early Mycobacterium Infection

Lessons from recent studies. João Gonçalves Pereira UCIP DALI

PHA Final Exam. Fall On my honor, I have neither given nor received unauthorized aid in doing this assignment.

Improving Translation in TB Drug Development Through Quantitative Modeling. CPTR Workshop 2016, Washington DC

A Mathematical Model For Interaction Macrophages, T Lymphocytes and Cytokines at Infection of Mycobacterium tuberculosis with Age Influence

C OBJECTIVES. Basic Pharmacokinetics LESSON. After completing Lesson 2, you should be able to:

Muhammad Fawad Rasool Feras Khalil Stephanie Läer

Lippincott Questions Pharmacology

Nontraditional PharmD Program PRDO 7700 Pharmacokinetics Review Self-Assessment

WHY... 8/21/2013 LEARNING OUTCOMES PHARMACOKINETICS I. A Absorption. D Distribution DEFINITION ADME AND THERAPEUIC ACTION

Basic Concepts of TDM

Supplementary Information. Tissue-wide immunity against Leishmania. through collective production of nitric oxide

Pharmacodynamics of sulfamethoxazole in in vitro and in vivo models of tuberculosis

Pharmacodynamic indices in targeting therapy of critical infections

Journal of Theoretical Biology

Supplementary Material

ACTIVATION OF T LYMPHOCYTES AND CELL MEDIATED IMMUNITY

Pharmacokinetics One- compartment Open Model Lec:2

Population pharmacokinetics and bacterial dynamics of sutezolid in patients with active tuberculosis

Assessing the utility of an anti-malarial pharmacokinetic-pharmacodynamic model for aiding drug clinical development

Identification of influential proteins in the classical retinoic acid signaling pathway

Simulation of Membrane and Cell Culture Permeability and Transport. Michael B. Bolger and Viera Lukacova Simulations Plus, Inc.

Differences in Reactivation of Tuberculosis Induced from Anti-TNF Treatments Are Based on Bioavailability in Granulomatous Tissue

General Principles of Pharmacology and Toxicology

ACTIVATION AND EFFECTOR FUNCTIONS OF CELL-MEDIATED IMMUNITY AND NK CELLS. Choompone Sakonwasun, MD (Hons), FRCPT

Pharmacokinetics and pharmacodynamics of anti-tuberculosis drugs in patients with tuberculosis

PHA5128 Dose Optimization II Case Study I Spring 2013

It the process by which a drug reversibly leaves blood and enter interstitium (extracellular fluid) and/ or cells of tissues.

BASIC PHARMACOKINETICS

Pharmacokinetic Modeling & Simulation in Discovery and non-clinical Development

The Influence of Physicochemical Properties on ADME

Immunology lecture: 14. Cytokines: Main source: Fibroblast, but actually it can be produced by other types of cells

Cathepsin S Inhibitors for the Treatment of Inflammatory Bowel Disease. Therapeutic Area Partnerships Presentation.

Comparison of Omadacycline and Tigecycline Pharmacodynamics in the Plasma, Epithelial Lining Fluid, and Alveolar Macrophages in Healthy Subjects

Chromatography on Immobilized Artificial Membrane

Define the terms biopharmaceutics and bioavailability.

DMPK. APRIL 27 TH 2017 Jan Neelissen Scientific Adviser Science & Technology

Volume of Distribution. Objectives. Volume of Distribution

The Multi-scale Immune Response to Pathogens: M. tuberculosis as an Example

PHA Second Exam. Fall On my honor, I have neither given nor received unauthorized aid in doing this assignment.

Scott Abrams, Ph.D. Professor of Oncology, x4375 Kuby Immunology SEVENTH EDITION

SUPPLEMENTAL MATERIAL

Jaline Gerardin *, Philip Eckhoff and Edward A Wenger

A NOVEL PHARMACODYNAMIC MODEL FOR TREATMENT OF TUBERCULOSIS USING DAYS TO POSITIVITY IN AUTOMATED LIQUID MYCOBACTERIAL CULTURE

A population pharmacokinetic model for rifampicin auto induction

Pharmacokinetic-Pharmacodynamic (PKPD) Modeling Characterizing Resistance for Predictions of Bacterial Kill in vivo

Pharmacodynamic principles and the time course of immediate drug effects

Topics. Humoral Immune Response Part II Accessory cells Fc Receptors Opsonization and killing mechanisms of phagocytes NK, mast, eosynophils

PHA Spring First Exam. 8 Aminoglycosides (5 points)

Absolute bioavailability and pharmacokinetic studies in early clinical development using microdose and microtracer approaches.

Time course of immune response

Outline 8/2/2013. PK/PD PK/PD first-line drug กก PK/PD กก

The ADME properties of most drugs strongly depends on the ability of the drug to pass through membranes via simple diffusion.

Dr. Nele Plock. Dr. N. Plock, March 11, 2008, American Conference on Pharmacometrics

Cell-mediated Immunity

Chapter 11 CYTOKINES

One of the major causes of death by infectious disease

Human Immunodeficiency Virus type 1 (HIV-1) gp120 / Glycoprotein 120 ELISA Pair Set

UNIVERSITY OF CAMBRIDGE INTERNATIONAL EXAMINATIONS International General Certificate of Secondary Education

Drug Dosing in Renal Insufficiency. Coralie Therese D. Dimacali, MD College of Medicine University of the Philippines Manila

The clinical trial information provided in this public disclosure synopsis is supplied for informational purposes only.

ANTIBIOTIC DOSE AND DOSE INTERVALS IN RRT and ECMO

Rational Dose Prediction. Pharmacology. φαρμακον. What does this mean? pharmakon. Medicine Poison Magic Spell

Phagocytosis: An Evolutionarily Conserved Mechanism to Remove Apoptotic Bodies and Microbial Pathogens

PHA Final Exam. Fall On my honor, I have neither given nor received unauthorized aid in doing this assignment.

CLINICAL PHARMACOKINETICS INDEPENDENT LEARNING MODULE

One-Compartment Open Model: Intravenous Bolus Administration:

Development of Canagliflozin: Mechanistic Absorption Modeling During Late-Stage Formulation and Process Optimization

IMMUNE EFFECTOR MECHANISMS. Cell-Mediated Reactions

1. If the MTC is 100 ng/ml and the MEC is 0.12 ng/ml, which of the following dosing regimen(s) are in the therapeutic window?

Introduction to Pharmacokinetics (PK) Anson K. Abraham, Ph.D. Associate Principal Scientist, PPDM- QP2 Merck & Co. Inc., West Point, PA 5- June- 2017

COMPARTMENTAL ANALYSIS OF DRUG DISTRIBUTION Juan J.L. Lertora, M.D., Ph.D. Director Clinical Pharmacology Program September 23, 2010

Biomarkers for Tuberculosis. Robert S. Wallis, MD, FIDSA Senior Director, Pfizer

The CLSI Approach to Setting Breakpoints

The human thymus is the central lymphoid organ that provides

Transcription:

Supplement to Comparing efficacies of moxifloxacin, levofloxacin and gatifloxacin in tuberculosis granulomas using a multi-scale systems pharmacology approach Elsje Pienaar 1,2, Jansy Sarathy 3, Brendan Prideaux 3, Jillian Dietzold 4, Véronique Dartois 3, Denise E. Kirschner 2 and Jennifer J. Linderman 1 Pharmacodynamic parameter estimation Figure S1: Overlay of in vitro and fitted in silico dose response curves for Mtb in liquid culture after 5 days (A) and in mouse macrophages after 3 days (B). Red data points show in vitro data, and black lines show model fit. X-axis shows antibiotic concentration added in the culture medium.

Fluoroquinolone dynamics in uninvolved rabbit lung samples There are experimental challenges to isolating truly uninvolved lung tissue in Mtb-infected rabbits. Most lung tissue samples showed some inflammation even in the absence of granulomas. Therefore LCMS measurements in uninvolved lung were not used for calibration of tissue PK parameters. Nonetheless, our in silico predicted concentrations of uninfected simulations are in agreement with measurements in uninvolved (i.e. non-granulomatous) rabbit lung samples (Figure S2). Differences between simulation predictions and rabbit data are most notable for MXF and LVX, and involve earlier peak concentrations predicted in the simulations compared to rabbit data. GFX MXF LVX Concentration 8 6 4 Rabbit (ug/g tissue) Simulation (ug/ml) C 5,BN C 5,BI Concentration 8 6 4 C 5,BN C 5,BI Concentration 8 6 4 C 5,BI C 5,BN C 5,BE 1 3 Time (Hours) C 5,BE 1 3 Time (Hours) C 5,BE 1 3 Time (Hours) Figure S2: Comparison of average concentrations from simulated uninvolved lung (solid lines) and LCMS measurements in rabbit granulomas (data points). Lines and data points show means and standard deviations for 1 simulations, and between 1 and 67 rabbit samples. Horizontal dashed lines show C 5 values for intracellular (C 5,BI ), extracellular replicating (C 5,BE ) and extracellular non-replicating bacteria (C 5,BN ).

Spatial fluoroquinolone distribution in granulomas with rabbit and human PK 2 hr 6 hr 24 hr 5 5 5 Rabbit PK 4 3 1 4 3 1 4 3 1 GFX 5 1 15 5 5 1 15 5 5 1 15 5 Human PK 4 3 1 5 1 15 4 3 1 5 1 15 4 3 1 5 1 15 6 6 6 Rabbit PK 4 4 4 MXF 5 1 15 6 5 1 15 6 5 1 15 6 Human PK 4 5 1 15 4 5 1 15 4 5 1 15 4 4 4 LVX Rabbit PK 3 1 5 1 15 4 3 1 5 1 15 4 3 1 5 1 15 4 Human PK 3 1 5 1 15 3 1 5 1 15 5 1 15 Figure S3: FQ concentrations from simulated granulomas plotted as a function of distance from the edge of the granuloma at 2, 6 and 24 hrs post dose. Solid lines show mean and dashed lines show standard deviation for 1 simulated granulomas. 3 1

Bacterial and immune responses following treatment interruption after 7 days Following treatment interruption after 1 days of treatment (at day 39), bacterial load rebounds more quickly and more steeply for GFX and LVX compared to MXF (Figure 1, main text). However, if treatment is interrupted after 7 days (at day 45) the outcomes are more similar between FQs (Figure S4 A-D). This indicates that bacterial and immunological differences between MXF and the other two FQs that are influential during the first 1 days of treatment, have largely been removed at this later time point (Figure S4 E-F) A Total Mtb B Intracellular Mtb Average Bacteria per granuloma (N=21) 1 1 1 1 GFX MXF LVX FQ 1 1 3 4 45 5 55 Time (Days) Average Bacteria per granuloma (N=21) 1 1 1 1 FQ 1 1 3 4 45 5 55 Time (Days) C Average Bacteria per granuloma (N=21) 1 1 1 1 Extracellular Mtb FQ 1 1 3 4 45 5 55 Time (Days) D Average Bacteria per granuloma (N=21) 1 1 1 1 Non-replicating extracellular Mtb FQ 1 1 3 4 45 5 55 Time (Days) E Infection 5 4 3 2 1. Treatment start (Day 38) F Infection 5 4 3 2 1. GFX MXF LVX Treatment interrupted Treatment start (Day 38).5.5 Treatment complete (Day 56)...5 1. 1.5 Immune response Treatment interruption (Day 45)...5 1. 1.5 Immune response Figure S4: (A-D) Simulations showing intracellular and extracellular bacteria increasing slightly following GFX and LVX interruption, where these populations are eliminated by MXF before day 45. The non-replicating bacterial population shows little change following interruption of any of the FQs. Lines show means of 21 in silico granulomas, with infection starting at day, daily FQ treatment starting at day 38 (arrows). Treatment is interrupted after 7 days (vertical dotted lines), and the simulation is continued to day 56 without antibiotics. (E-F) Progression of treatment as a function of the collective immune response metric (x-axes) and the collective infection metric (y-axes), during complete treatment (E) and interrupted treatment (F). The start of treatment is located at the intersection of the dotted lines. Filled circles indicate the treatment phase, and open circles indicated progression following treatment interruption.

Host immune parameters varied to capture granuloma variability for tissue PK calibration Table S1: Host immune parameter ranges used to generate collections of test granulomas to calibrate tissue PK parameters 1,2 Host Immune Parameters Unit* Range (min max) Time to heal caseation Days 8 12 TNF threshold for causing apoptosis Molecules 9 14 Rate of TNF induced apoptosis s -1 1.3x1-6 2x1-6 Minimum chemokine concentration allowing chemotaxis Molecules.4.6 Maximum chemokine concentration allowing chemotaxis Molecules 38 57 Initial macrophage density Fraction of grid comp..3.5 Time steps before a resting macrophage can move Timesteps 2 4 Time steps before an activated macrophage can move Timesteps 15 24 Time steps before an infected macrophage can move Timesteps 135 TNF threshold for activating NFkB Molecules 6 9 Rate of TNF induced NFkB activation s -1 8x1-6 1.5x1-5 Probability of resting macrophage killing bacteria.1.15 Adjustment for killing probability of resting macrophages with.15.25 NFkB activated Number of extracellular bacteria in the Moore neighborhood Bacteria 3 that can activate NFkB Threshold for intracellular bacteria causing chronically infected macrophages Bacteria 1 15 Threshold for intracellular bacteria causing macrophage to burst Bacteria 18 3 Number of bacteria activated macrophage can phagocytose Bacteria 4 6 Probability of an activated macrophage healing a caseated compartment in its Moore neighborhood Number of host cell deaths causing caseation.4.7 4 (Caseous granulomas) 15 (Cellular granulomas) Probability of a T-cell moving to the same compartment as a.35.55 macrophage IFN γ producing T-cell probability of inducing Fas/FasL.3.4 mediated apoptosis IFN γ producing T-cell probability of producing TNF.4.5 IFN γ producing T-cell probability of producing IFN.3.45 Cytotoxic T-cell probability of killing a macrophage.7.1 Cytotoxic T-cell probability of, when it kills a macrophage, also killing all of its intracellular bacteria.6.9 Cytotoxic T-cell probability of producing TNF.4.6 Regulatory T-cell probability of deactivating activated.6.1 macrophage Time before maximum recruitment rates are reached Timesteps* 79 118 Macrophage maximal recruitment probability.25.4 Macrophage chemokine recruitment threshold Molecules.7 1 Macrophage TNF recruitment threshold Molecules.9.15 Macrophage half sat for TNF recruitment Molecules 1.3 2 Macrophage half sat for chemokine recruitment Molecules 1.8 2.6 IFN γ producing T-cell maximal recruitment probability.12.18 IFN γ producing T-cell chemokine recruitment threshold Molecules..6.9 IFN γ producing T-cell TNF recruitment threshold Molecules 1 1.6 IFN γ producing T-cell half sat for TNF recruitment Molecules 1 1.6

IFN γ producing T-cell half sat for chemokine recruitment Molecules 1.5 2.5 Cytotoxic T-cell maximal recruitment probability.1.15 Cytotoxic T-cell chemokine recruitment threshold Molecules 3.6 5.4 Cytotoxic T-cell TNF recruitment threshold Molecules 1 1.5 Cytotoxic T-cell half sat for TNF recruitment Molecules 1 1.5 Cytotoxic T-cell half sat for chemokine recruitment Molecules 7 1 Regulatory T-cell maximal recruitment probability.2.4 Regulatory T-cell chemokine recruitment threshold Molecules 1.5 2.5 Regulatory T-cell TNF recruitment threshold Molecules 1.3 2 Regulatory T-cell half sat for TNF recruitment Molecules 1.8 2.7 Regulatory T-cell half sat for chemokine recruitment Molecules 1.2 1.8 *Conversion factor: 1 min/timestep.

Tissue PK parameter fitting We estimate tissue PK parameters by calibrating GranSim to the in vivo data summarized in Table 2 and Table M2 in the main text. We use Latin Hypercube sampling to sample the parameter space (See Methods). The ranges sampled for each parameter are based on a collection of in vitro and literature data (Table S2). FQ diffusivity ranges in tissue are estimated based on molecular weight, logp and the number of hydrogen donor and acceptor sites based on diffusion studies in tumors 3. Vascular permeability estimates based on molecular radius alone predict vascular permeability of ~5x1-5 cm/s for all three FQs 4. However, we use vascular permeability ranges one log lower than this estimate (5x1-6 cm/s), since we noted that the predicted diffusivity dropped by ~1-fold if one includes the physicochemical properties listed above in addition to the molecular size alone. Furthermore, in vitro permeability studies showed that MXF has consistently higher permeability than GFX and LVX by 2- to 1-fold 5. We estimate initial MXF permeability ranges 2-fold higher than GFX and LVX. Cellular uptake ratio estimates are based on in vitro cell uptake assays in THP-1 cells described below. Permeability coefficient estimates are based on plasma protein binding measurements 5. Caseum unbound fractions are estimated from in vitro rapid equilibrium dialysis assays described below. Table S2: Parameter ranges used for Tissue PK Parameter fitting. The ranges explored during calibration were chosen based on best estimates from experiments and literature for all three FQs. Where no references are given, ranges are based on in vitro data obtained in this work. Final parameter estimates resulting from calibration are given in Table M2 in the main text. Parameter Units MXF GFX LVX Effective diffusivity (D) Cellular accumulation ratio (2) (a) Vascular permeability (p) Permeability coefficient (PC) Caseum unbound fraction (f u ) Caseum binding rate constant (k fc ) Epithelium binding association constant (K a ) Epithelium binding rate constant (k fe ) Cellular exit rate constant (k out ) Experimental/ Literature estimate cm 2 /s 2.6x1-7 Range used in calibration 2.6x1-8 Experimenta l/literature estimate Range used in calibration 7x1-8 Experiment al/literatur e estimate Range used in calibration 4x1-8 4x1-6 2.6x1-6 7x1-6 3 3 3 7x1-7 4x1-7 - 4.35.4 4 2.78.2 2.9.2 cm/s 1x1-5 -.5 5 4 1x1-6 5x1-6 1x1-4 4.5 5.8 5 5x1-7 5x1-6 5x1-5 4.8 8.7 5 5x1-7 5x1-5.7 7 -.13.5.3.16.5.3.18.1.4 cu -1 s -1.2.2 -.1.2 s -1.4.99.2.2.1.2.4.99.2.2.1.2.4.99 s -1.1.5.1.5.1.5

Sensitivity analysis For sensitivity analysis using Partial rank correlations coefficients (PRCC, see Methods), parameters were sampled simultaneously and uniformly in the ranges given in Table S3 using Latin Hypercube sampling (LHS). Significant correlations are summarized in Table S4. Table S3: Parameters and ranges used in PRCC calculation for sensitivity analysis. Parameter Units Min Max Plasma PK parameters (2,3) Absorption rate constant (k a ) h -1 1 1 Intercompartmental clearance rate constant (Q) L/h/kg 1 1 Plasma volume of distribution (V p ) L/kg.1 1 Peripheral volume of distribution (V pe ) L/kg.1 1 Plasma clearance rate constant (CL) L/h/kg.1 1 Lung tissue PK parameters (4) Effective diffusivity (D) cm 2 /s 1x1-7 1x1-6 Cellular accumulation ratio (2) (a) - 1 1 Vascular permeability (p) cm/s 1x1-6 1x1-5 Permeability coefficient (PC) - 1 1 Caseum unbound fraction (f u ) -.1.9 Caseum binding rate constant (k fc ) cu -1 s -1.5.5 Epithelium binding association constant (K a ) -.1.3 Epithelium binding rate constant (k fe ) s -1.2.9 Cellular exit rate constant (k out ) s -1.2.2 PD parameters (5) Max activity extracellular (E max,be ) s -1.1.1 Max activity intracellular (E max,bi ) s -1.1.1 C5 for extracellular replicating Mtb (C 5,BE ) mg/l.1.1 C5 for extracellular non-replicating Mtb (C 5,BN ) (6) mg/l 5 5 C5 for intracellular Mtb (C 5,BI ) mg/l 5 5 Hill constant for intracellular Mtb (H BI ) - 1 5 Hill constant for extracellular replicating Mtb (H BE ) - 1 5 Hill constant for extracellular non-replicating Mtb (H BN ) - 1 5

Table S4: Significant correlations between model parameters and model outputs. Only significantly correlated parameters are shown. Relationship: + : positive correlation; - : negative correlation. Significance: *: p <.1; **: p <.1; ***: p <.1. Significant(Parameters ka Vp CL a p PC Emax,BI Emax,BE C5,BN C5,BI HBE fu Resting(macrophages +++ 555 555 555 + +++ Infected(macrophages +++ 555 555 555 +++ Chronically(infected(macrophages +++ 555 555 555 +++ Activated(macrophages +++ 555 555 555 +++ IFNg(producing(T(cells +++ 555 555 555 +++ Activated(IFNg(producing(T(cells +++ 555 555 555 +++ Cytotoxic(T(cells +++ 555 555 555 +++ Activated(cytotoxic(T(cells +++ 555 555 555 +++ Regulatory(T(cells +++ 555 555 555 +++ Activated(regulatory(T(cells +++ 555 555 555 +++ Intracellular(Mtb +++ 555 555 555 +++ Extracellular(Mtb +++ 555 555 555 555 +++ +++ +++ Replicating(extracellular(Mtb 5 +++ 555 +++ 555 555 555 +++ + Non@replicating(extracellular(Mtb +++ 555 555 555 555 +++ +++ +++ Total(MTb +++ 555 555 555 5 +++ +++ +++ AUC(in(Blood((@24hr) 555 +++ 555 AUC(in(Granuloma((@24hr) 555 +++ +++ 5 555 TNF +++ 555 555 555 +++ IL1 +++ 555 555 555 +++ Lesion(Size +++ 555 555 555 +++ Caseation(level +++ 555 555 555 +++ Model(outputs

References 1 Pienaar, E. et al. A computational tool integrating host immunity with antibiotic dynamics to study tuberculosis treatment. J Theor Biol 367, 166-179, doi:1.116/j.jtbi.14.11.21 (15). 2 Pienaar, E., Dartois, V., Linderman, J. J. & Kirschner, D. In silico evaluation and exploration of antibiotic tuberculosis treatment regimens. BMC systems biology 9, 79, doi:1.1186/s12918-15-221-8 (15). 3 Pruijn, F. B., Patel, K., Hay, M. P., Wilson, W. R. & Hicks, K. O. Prediction of Tumour Tissue Diffusion Coefficients of Hypoxia-Activated Prodrugs from Physicochemical Parameters. Australian Journal of Chemistry 61, 687-693 (8). 4 Schmidt, M. M. & Wittrup, K. D. A modeling analysis of the effects of molecular size and binding affinity on tumor targeting. Molecular cancer therapeutics 8, 2861-2871, doi:1.1158/1535-7163.mct-9-195 (9). 5 Lakshminarayana, S. B. et al. Comprehensive physicochemical, pharmacokinetic and activity profiling of anti-tb agents. The Journal of antimicrobial chemotherapy 7, 857-867, doi:1.193/jac/dku457 (15).