1
Abstract Due to its exposure to high concentrations of xenobiotics, the kidney proximal tubule (PT) is the primary site of nephrotoxicity which results in late-stage attrition during drug development. We have developed a 3D bioprinted, fully human in vitro model of the proximal tubulointerstitial interface to enable more accurate prediction of tissue- level clinical outcomes 1 . We challenged this model with a diverse set of known renal toxicants and assessed their impact using biochemical and histologic endpoints. Treatment of 3D PT tissues with the chemotherapeutic agent, Cisplatin, induced dose- and time-dependent loss of tissue viability and epithelial function over the 14 day time-course evaluated. Temporal evaluation of the soluble biomarkers N-acetyl-β-D-glucosamindade (NAG) and lactate dehydrogenase (LDH) demonstrated maximum NAG release earlier than maximum LDH release. Histologic assessment of the tissues showed primarily epithelial cell-specific effects at the 2.5 μM and 5 μM doses, while additional toxicity in the interstitium was observed at the 10 uM dose. Co-treatment of the tissues with 5uM Cisplatin and the OCT2 inhibitor Cimetidine (1mM) attenuated the Cisplatin-induced toxicity. To assess another class of nephrotoxicants, 3D PT tissues were treated with the aminoglycoside antibiotics, Amphotericin B and Gentamicin. Amphotericin B induced a dose-dependent decrease in epithelial function without a corresponding decrease in overall tissue metabolic activity, while Gentamicin treatment resulted in decreases in both epithelial function and tissue metabolic activity. Examination of the soluble biomarkers NAG and LDH showed a rapid response of the tissues to even low concentrations of Amphotericin B early in the treatment period. Consistent with the biochemical findings, histologic assessment of the Amphotericin B-treated tissues demonstrated a flattening of the epithelial cells with no observable impact on the other cells within the tissue. Collectively, these results suggest that bioprinted kidney tissues are well-suited to assess multiple mechanisms of nephrotoxicity including fibrosis in vitro, following biologically-relevant dosing regimens, and that biochemical, transcriptional and histologic endpoints provide a comprehensive means of examining the progression of tissue injury on a mechanistic basis. Methods Using Organovo’s proprietary bioprinting platform, we have developed a human in vitro 3D tissue model of the proximal tubule to enable more accurate prediction of clinical outcomes. The tissue is composed of a renal interstitium of primary human renal fibroblasts and endothelial cells, providing a basement membrane and support for a polarized monolayer of primary human PT epithelial cells. Test articles were dosed daily. Results Figure 2. Biochemical evaluation of Cisplatin-induced toxicity and rescue with inhibition of OCT2. 3D PT kidney tissues were dosed daily for 14 days and assessed for biochemical endpoints of toxicity. Cimetidine was co-administered as indicated to inhibit OCT2-mediated uptake of Cisplatin. A and B, Release of LDH or NAG as biomarkers of toxicity. C, Concentration-dependent reduction of overall metabolic activity as measured by alamarBlue®. D, Reduction in PT epithelial cell function as measured by GGT activity. Figure 3. Histological evaluation of Cisplatin-induced toxicity and rescue with inhibition of OCT2. 3D PT kidney tissues were dosed daily for 14 days and assessed for histological phenotype. Cimetidine co-administered as indicated to inhibit OCT2-mediated uptake of Cisplatin and protect PT epithelial cells from toxicity. Panels display H&E stains for: A, Vehicle. B, 1mM Cimetidine. C, 5μM Cisplatin. D, 5μM Cisplatin + 1mM Cimetidine. Insets show magnified view of PT epithelial cell morphology. Figure 1. Design and Characterization of ExVive™ Human Kidney Tissue. A, H&E cross-section of native human kidney proximal tubules. B, Schematic of laminar model of the proximal tubule, comprising an interstitial layer of renal fibroblasts and endothelial cells supporting a polarized epithelial monolayer in standard Transwell format. C, Bioprinted tissues are highly reproducible from well to well. D, H&E. E, Masson’s Trichrome stain for collagen (blue). F, CD31 (red) staining for endothelial cell networks. G, Cytokeratin 18 (red) staining for epithelial cells. H, E-cadherin staining (green) for junctions between epithelial cells, Collagen IV staining (red) for basement membrane. I, Basolateral localization of Na + K + ATPase. DAPI (blue) staining for nuclei. J, Gamma-glutamyl transferase (GGT) activity as a measure of renal proximal tubule epithelial cell-specific function over time. Summary Conventional preclinical renal assays such as in vitro cell culture and animal models are insufficient in modeling the complexity of drug-induced organ toxicity due to limited functionality or species- specific variation. ExVive™ Human Kidney Tissue is a 3D multicellular model of the renal tubulointerstitial interface, comprised of a polarized monolayer of primary human proximal tubule epithelial cells supported by an interstitium of human renal fibroblasts and endothelial cells. Drug-induced, dose-dependent renal injury following exposure to classical nephrotoxicants including the chemotherapeutic agent, Cisplatin, and aminoglycoside antibiotics, Amphotericin B and Gentamicin, can be detected at the biochemical and histological levels. TGFb exposure demonstrates extended capability of ExVive™ Human Kidney Tissue to mount measurable responses at the biochemical, transcriptional, and histological levels consistent with renal injury and fibrosis, a disease phenotype not achievable in traditional systems of epithelial cell monolayer culture with limited longevity. Together, these results support the utility of this novel 3D tissue model of the proximal tubule for assessment of human renal toxicity over extended time in culture. Figure 7. TGFb-induced fibrosis. 3D PT kidney tissues were dosed daily for 7 days with Vehicle, 0.37 ng/ml, 1.1ng/ml, 3.3ng/ml, or 10ng/ml TGFb. A, Increasing doses of TGFb do not impact overall tissue metabolic activity as measured by alamarBlue®. B, Reduction in PT epithelial cell function at higher doses as measured by GGT activity. C, Increased tissue thickening and ECM deposition as assessed by Gomori’s Trichrome stain. D, Dose-dependent induction of fibrotic markers, collagen I (COL1A1), connective tissue growth factor (CTGF), fibroblast-activating protein (FAP), or platelet-derived growth factor receptor β (PDGFRB) as assessed by quantitative RT-PCR. *p<0.0001. E, Quantification of collagen in Sirius red-stained tissue sections. *p<0.05. Utilization of the ExVive Human Kidney Tissue Model of the Proximal Tubule to Assess Nephrotoxicity Across a Diverse Set of Chemical Classes J. William Higgins, Timothy R. Smith, Shelby M. King, Elizabeth Paffenroth, Casey Fairbairn, Alice E. Chen, and Deborah G. Nguyen. | Organovo, 6275 Nancy Ridge Drive, San Diego, CA 92121 CHANGING THE SHAPE OF RESEARCH AND MEDICINE www.organovo.com Figure 5. Histological evaluation of Amphotericin B-induced epithelial toxicity. 3D PT kidney tissues were dosed daily for 7 days and assessed for histological phenotype. Panels display H&E stains for: A, Vehicle. B, 1μM Amphotericin B. C, 10μM Amphotericin B. D, 30μM Amphotericin B. Insets show magnified view of PT epithelial cell morphology. Figure 4: Biochemical evaluation of Amphotericin B-induced toxicity. 3D PT kidney tissues were dosed daily for 7 days and assessed for biochemical endpoints of toxicity . A and B, Release of LDH or NAG as biomarkers of toxicity. C, Concentration-dependent reduction of overall metabolic activity as measured by alamarBlue®. D, Reduction in PT epithelial cell function as measured by GGT activity. Figure 6. Biochemical evaluation of Gentamicin-induced toxicity 2 . 3D PT kidney tissues were dosed daily for 14 days and assessed for biochemical endpoints of toxicity. A, Release of LDH as a biomarker of toxicity. TX, treatment day. B, Concentration-dependent reduction of overall metabolic activity as measured by alamarBlue®. C, Concentration-dependent reduction in PT epithelial cell function as measured by GGT activity. Forward Looking Statement This presentation contains statements about future events and expectations known as “forward-looking statements” within the meaning of Section 27A of the Securities Act of 1933, as amended (the “Securities Act”), and Section 21E of the Securities Exchange Act of 1934, as amended (the “Exchange Act”). The Company has based these forward-looking statements on its current expectations and the information currently available to it, but any forward-looking statements are subject to a number of risks and uncertainties. The factors that could cause the Company’s actual future results to differ materially from its current expectations, or from the results implied by any forward-looking statements, include, but are not limited to, risks and uncertainties relating to the Company's ability to develop, market and sell products and services based on its technology; the expected benefits and efficacy of the Company's products, services and technology; the Company’s ability to successfully complete studies and provide the technical information required to support market acceptance of its products, services and technology, on a timely basis or at all; the Company’s ability to generate revenue and control its operating losses; the validity of the Company’s intellectual property rights and the ability to protect those rights; the Company's ability to implement and achieve its business, research, product development, regulatory approval, marketing and distribution plans and strategies; the Company’s ability to secure additional contracted collaborative relationships; and the Company’s ability to meet its fiscal-year 2017 outlook and/or its long-range outlook. These and other factors are identified and described in more detail in the Company’s filings with the Securities and Exchange Commission (“the SEC”), including those factors listed under the caption "Risk Factors" in the Company’s Form 10-K for the year ended March 31, 2016, filed with the SEC on June 9, 2016, as well as other filings Organovo makes with the SEC from time to time. Readers are cautioned not to place undue reliance on forward-looking statements, which speak only as of the date of this presentation. Except as required by applicable law, we do not intend to update any of the forward-looking statements to conform these statements to reflect actual results, later events or circumstances or to reflect the occurrence of unanticipated events. A. B. One-Way ANOVA Dunnet’s Multiple Comparisons * P<0.05 *** P<0.001 C. Vehicle 0.37 ng/ml 1.1 ng/ml 3.3 ng/ml 10 ng/ml D. References 1. King, S.M., et al. (2017). 3D Proximal Tubule Tissues Recapitulate Key Aspects of Renal Physiology to Enable Nephrotoxicity Testing. Frontiers in Physiology. In Press. 2. Gentamicin studies were done in partnership with Dr. Kristine de Dios and Dr. Andrew Seacat at La Jolla Pharmaceuticals. A. B. C. D. A. B. C. D. Abstract No. 1804 Poster No. P344 E. A. B. C. D. One-Way ANOVA Dunnet’s Multiple Comparisons *** P<0.001 **** P<0.0001 TX = treatment day A. B. C. D. TX = treatment day One-Way ANOVA Dunnet’s Multiple Comparisons *** P<0.001 **** P<0.0001 Vehicle 0.1mM Gentamicin 1mM Gentamicin 10mM Gentamicin 0 25 50 75 100 125 Metabolic Activity 14 Days of Gentamicin Treatment Percent of Control (%) **** **** Vehicle 0.1mM Gentamicin 1mM Gentamicin 10mM Gentamicin 0 25 50 75 100 125 GGT Activity 14 Days of Gentamicin Treatment Percent of Control (%) **** **** **** TX1 TX3 TX5 TX7 TX14 0 2 4 6 8 LDH Release 14 Days of Gentamicin Treatment Treatment Day Fold LDH Induction (Relative to Vehicle) A. B. C. Vehicle 0.1mM Gentamicin 1mM Gentamicin 10mM Gentamicin I.

F R Utilization of the ExVive Human Kidney Tissue Model of ...Abstract Due to its exposure to high concentrations of xenobiotics, the kidney proximal tubule (PT) is the primary site

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: F R Utilization of the ExVive Human Kidney Tissue Model of ...Abstract Due to its exposure to high concentrations of xenobiotics, the kidney proximal tubule (PT) is the primary site

Abstract Due to its exposure to high concentrations of xenobiotics, the kidney proximal tubule (PT) is the primary site ofnephrotoxicity which results in late-stage attrition during drug development. We have developed a 3D bioprinted,fully human in vitro model of the proximal tubulointerstitial interface to enable more accurate prediction of tissue-level clinical outcomes 1.

We challenged this model with a diverse set of known renal toxicants and assessed their impact using biochemicaland histologic endpoints. Treatment of 3D PT tissues with the chemotherapeutic agent, Cisplatin, induced dose-and time-dependent loss of tissue viability and epithelial function over the 14 day time-course evaluated. Temporalevaluation of the soluble biomarkers N-acetyl-β-D-glucosamindade (NAG) and lactate dehydrogenase (LDH)demonstrated maximum NAG release earlier than maximum LDH release. Histologic assessment of the tissuesshowed primarily epithelial cell-specific effects at the 2.5 µM and 5 µM doses, while additional toxicity in theinterstitium was observed at the 10 uM dose. Co-treatment of the tissues with 5uM Cisplatin and the OCT2inhibitor Cimetidine (1mM) attenuated the Cisplatin-induced toxicity.

To assess another class of nephrotoxicants, 3D PT tissues were treated with the aminoglycoside antibiotics,Amphotericin B and Gentamicin. Amphotericin B induced a dose-dependent decrease in epithelial function withouta corresponding decrease in overall tissue metabolic activity, while Gentamicin treatment resulted in decreases inboth epithelial function and tissue metabolic activity. Examination of the soluble biomarkers NAG and LDH showeda rapid response of the tissues to even low concentrations of Amphotericin B early in the treatment period.Consistent with the biochemical findings, histologic assessment of the Amphotericin B-treated tissuesdemonstrated a flattening of the epithelial cells with no observable impact on the other cells within the tissue.

Collectively, these results suggest that bioprinted kidney tissues are well-suited to assess multiple mechanisms ofnephrotoxicity including fibrosis in vitro, following biologically-relevant dosing regimens, and that biochemical,transcriptional and histologic endpoints provide a comprehensive means of examining the progression of tissue

injury on a mechanistic basis.

Methods Using Organovo’s proprietary bioprinting platform, we have developed a human in vitro 3D tissue model of theproximal tubule to enable more accurate prediction of clinical outcomes. The tissue is composed of a renalinterstitium of primary human renal fibroblasts and endothelial cells, providing a basement membrane and supportfor a polarized monolayer of primary human PT epithelial cells. Test articles were dosed daily.

Results

Figure 2. Biochemical evaluation of Cisplatin-induced toxicity and rescue with inhibition of OCT2. 3D PTkidney tissues were dosed daily for 14 days and assessed for biochemical endpoints of toxicity. Cimetidinewas co-administered as indicated to inhibit OCT2-mediated uptake of Cisplatin. A and B, Release of LDH orNAG as biomarkers of toxicity. C, Concentration-dependent reduction of overall metabolic activity asmeasured by alamarBlue®. D, Reduction in PT epithelial cell function as measured by GGT activity.

Figure 3. Histological evaluation of Cisplatin-induced toxicity and rescue with inhibition of OCT2. 3D PT kidneytissues were dosed daily for 14 days and assessed for histological phenotype. Cimetidine co-administered asindicated to inhibit OCT2-mediated uptake of Cisplatin and protect PT epithelial cells from toxicity. Panelsdisplay H&E stains for: A, Vehicle. B, 1mM Cimetidine. C, 5µM Cisplatin. D, 5µM Cisplatin + 1mM Cimetidine.Insets show magnified view of PT epithelial cell morphology.

Figure 1. Design and Characterization of ExVive™ Human Kidney Tissue. A, H&E cross-section of native human kidney proximaltubules. B, Schematic of laminar model of the proximal tubule, comprising an interstitial layer of renal fibroblasts and endothelial cellssupporting a polarized epithelial monolayer in standard Transwell format. C, Bioprinted tissues are highly reproducible from well towell. D, H&E. E, Masson’s Trichrome stain for collagen (blue). F, CD31 (red) staining for endothelial cell networks. G, Cytokeratin 18(red) staining for epithelial cells. H, E-cadherin staining (green) for junctions between epithelial cells, Collagen IV staining (red) forbasement membrane. I, Basolateral localization of Na+K+ATPase. DAPI (blue) staining for nuclei. J, Gamma-glutamyl transferase (GGT)activity as a measure of renal proximal tubule epithelial cell-specific function over time.

Summary• Conventional preclinical renal assays such as in vitro cell culture and animal models are insufficient

in modeling the complexity of drug-induced organ toxicity due to limited functionality or species-specific variation.

• ExVive™ Human Kidney Tissue is a 3D multicellular model of the renal tubulointerstitial interface,comprised of a polarized monolayer of primary human proximal tubule epithelial cells supported byan interstitium of human renal fibroblasts and endothelial cells.

• Drug-induced, dose-dependent renal injury following exposure to classical nephrotoxicantsincluding the chemotherapeutic agent, Cisplatin, and aminoglycoside antibiotics, Amphotericin Band Gentamicin, can be detected at the biochemical and histological levels.

• TGFb exposure demonstrates extended capability of ExVive™ Human Kidney Tissue to mountmeasurable responses at the biochemical, transcriptional, and histological levels consistent withrenal injury and fibrosis, a disease phenotype not achievable in traditional systems of epithelial cellmonolayer culture with limited longevity.

• Together, these results support the utility of this novel 3D tissue model of the proximal tubule forassessment of human renal toxicity over extended time in culture.

Figure 7. TGFb-induced fibrosis. 3D PT kidney tissues were dosed daily for 7 days with Vehicle, 0.37 ng/ml, 1.1ng/ml, 3.3ng/ml,or 10ng/ml TGFb. A, Increasing doses of TGFb do not impact overall tissue metabolic activity as measured by alamarBlue®. B,Reduction in PT epithelial cell function at higher doses as measured by GGT activity. C, Increased tissue thickening and ECMdeposition as assessed by Gomori’s Trichrome stain. D, Dose-dependent induction of fibrotic markers, collagen I (COL1A1),connective tissue growth factor (CTGF), fibroblast-activating protein (FAP), or platelet-derived growth factor receptor β(PDGFRB) as assessed by quantitative RT-PCR. *p<0.0001. E, Quantification of collagen in Sirius red-stained tissue sections.*p<0.05.

Utilization of the ExVive Human Kidney Tissue Model of the Proximal Tubule to Assess Nephrotoxicity Across a Diverse Set of Chemical ClassesJ. William Higgins, Timothy R. Smith, Shelby M. King, Elizabeth Paffenroth, Casey Fairbairn, Alice E. Chen, and Deborah G. Nguyen. | Organovo, 6275 Nancy Ridge Drive, San Diego, CA 92121

CHANGING THE SHAPE OF RESEARCH AND MEDICINE www.organovo.com

Figure 5. Histological evaluation of Amphotericin B-induced epithelial toxicity. 3D PT kidney tissues weredosed daily for 7 days and assessed for histological phenotype. Panels display H&E stains for: A, Vehicle. B, 1µMAmphotericin B. C, 10µM Amphotericin B. D, 30µM Amphotericin B. Insets show magnified view of PT epithelialcell morphology.

Figure 4: Biochemical evaluation of Amphotericin B-induced toxicity. 3D PT kidney tissues were dosed

daily for 7 days and assessed for biochemical endpoints of toxicity. A and B, Release of LDH or NAG as

biomarkers of toxicity. C, Concentration-dependent reduction of overall metabolic activity as measured by

alamarBlue®. D, Reduction in PT epithelial cell function as measured by GGT activity.

Figure 6. Biochemical evaluation of Gentamicin-induced toxicity2. 3D PT kidney tissues were dosed daily for 14days and assessed for biochemical endpoints of toxicity. A, Release of LDH as a biomarker of toxicity. TX, treatmentday. B, Concentration-dependent reduction of overall metabolic activity as measured by alamarBlue®. C,Concentration-dependent reduction in PT epithelial cell function as measured by GGT activity.

Forward Looking Statement

This presentation contains statements about future events and expectations known as “forward-looking statements” within the meaning of Section 27A of the Securities Act of 1933, as amended (the “Securities Act”), and Section 21E of the Securities Exchange Act of 1934, as amended (the “Exchange Act”). The Company has based these forward-looking statements on its current expectations andthe information currently available to it, but any forward-looking statements are subject to a number of risks and uncertainties. The factors that could cause the Company’s actual future results to differ materially from its current expectations, or from the results implied by any forward-looking statements, include, but are not limited to, risks and uncertainties relating to the Company's ability todevelop, market and sell products and services based on its technology; the expected benefits and efficacy of the Company's products, services and technology; the Company’s ability to successfully complete studies and provide the technical information required to support market acceptance of its products, services and technology, on a timely basis or at all; the Company’s ability to generaterevenue and control its operating losses; the validity of the Company’s intellectual property rights and the ability to protect those rights; the Company's ability to implement and achieve its business, research, product development, regulatory approval, marketing and distribution plans and strategies; the Company’s ability to secure additional contracted collaborative relationships; and theCompany’s ability to meet its fiscal-year 2017 outlook and/or its long-range outlook. These and other factors are identified and described in more detail in the Company’s filings with the Securities and Exchange Commission (“the SEC”), including those factors listed under the caption "Risk Factors" in the Company’s Form 10-K for the year ended March 31, 2016, filed with the SEC on June 9, 2016,as well as other filings Organovo makes with the SEC from time to time.

Readers are cautioned not to place undue reliance on forward-looking statements, which speak only as of the date of this presentation. Except as required by applicable law, we do not intend to update any of the forward-looking statements to conform these statements to reflect actual results, later events or circumstances or to reflect the occurrence of unanticipated events.

A. B.

One-Way ANOVADunnet’s Multiple Comparisons* P<0.05*** P<0.001

C.

Vehicle 0.37 ng/ml 1.1 ng/ml

3.3 ng/ml 10 ng/ml

D.

References1. King, S.M., et al. (2017). 3D Proximal Tubule Tissues Recapitulate Key Aspects of Renal Physiology to Enable Nephrotoxicity Testing. Frontiers

in Physiology. In Press.2. Gentamicin studies were done in partnership with Dr. Kristine de Dios and Dr. Andrew Seacat at La Jolla Pharmaceuticals.

A. B.

C. D.

A. B.

C. D.

Abstract No. 1804Poster No. P344

E.

A. B.

C. D.

One-Way ANOVADunnet’s Multiple Comparisons*** P<0.001**** P<0.0001

TX = treatment day

A. B.

C. D.

TX = treatment day

One-Way ANOVADunnet’s Multiple Comparisons*** P<0.001**** P<0.0001

Ve h

icle

0 .1m

M G

e nta

mic

in

1 mM

Ge n

tam

icin

1 0 mM

Ge n

tam

icin

0

2 5

5 0

7 5

1 0 0

1 2 5

M e ta b o lic A c t iv it y

1 4 D a y s o f G e n ta m ic in T re a tm e n t

Pe

rce

nt

of

Co

ntr

ol

(%)

****

****

Ve h

icle

0 .1m

M G

e nta

mic

in

1 mM

Ge n

tam

icin

1 0 mM

Ge n

tam

icin

0

2 5

5 0

7 5

1 0 0

1 2 5

G G T A c t iv ity

1 4 D a y s o f G e n ta m ic in T r e a tm e n t

Pe

rce

nt

of

Co

ntr

ol

(%)

****

****

****

T X1

T X3

T X5

T X7

T X1 4

0

2

4

6

8

L D H R e le a s e

1 4 D a y s o f G e n ta m ic in T re a tm e n t

T r e a t m e n t D a y

Fo

ld L

DH

In

du

cti

on

(Re

lati

ve

to

Ve

hic

le)

V e h ic le

0 .1 m M G e n t a m ic in

1 m M G e n t a m ic in

1 0 m M G e n t a m ic in

A. B. C.

T X1

T X3

T X5

T X7

T X1 4

0

2

4

6

8

L D H R e le a s e

1 4 D a y s o f G e n ta m ic in T re a tm e n t

T r e a t m e n t D a y

Fo

ld L

DH

In

du

cti

on

(Re

lati

ve

to

Ve

hic

le)

V e h ic le

0 .1 m M G e n t a m ic in

1 m M G e n t a m ic in

1 0 m M G e n t a m ic in

I.