Modeling NAFLD Using 3D Bioprinted Human Liver Tissue · Except as required by applicable law,...

1
Background Nonalcoholic fatty liver disease (NAFLD) is a chronic condition that originates as lipid accumulation within hepatocytes (steatosis) and progresses into nonalcoholic steatohepatitis (NASH), characterized by lipid accumulation, inflammation, oxidative stress, and fibrosis. NAFLD is now recognized as the most common cause of chronic liver disease, with a prevalence of 25% worldwide, and is projected to become the leading indication for liver transplant by 2025. Despite decades of research, the mechanisms of NAFLD progression, therapeutic approaches and non-invasive diagnostics are still resoundingly absent. The study of steatosis and NASH has traditionally utilized rodent models, which are time consuming to generate and do not fully recapitulate the complex phenotypes associated with the human disease. Furthermore, current 2D cell culture models lack relevant liver cell types and have limited utility due to rapid loss of cell viability and function. Thus, there is a significant need for a more predictive human multicellular 3D in vitro model to study the progression of steatosis into NASH. Methods ExVive™ Human Liver Tissue, a human in vitro 3D bioprinted liver model comprising primary human hepatocytes, hepatic stellate cells, and endothelial cells exhibits a complex multicellular architecture similar to that of native liver and retains metabolic competence and liver-specific functions for at least 4 weeks in culture. To mimic the proposed pathogenesis of NASH via a “Two-Hit Hypothesis”, standard ExVive™ Human Liver Tissue were supplemented with Kupffer cells to achieve immune competency then exposed to steatogenic cues via a nutrient overload approach of simple sugars and fatty acids, followed by inflammatory stimulation using prototypical inducers. Modeling NAFLD Using 3D Bioprinted Human Liver Tissue Dwayne E. Carter a , Deborah G. Nguyen a , David Brenner b , Sharon C. Presnell a , and Alice E. Chen a a Organovo, 6275 Nancy Ridge Drive, San Diego, CA 92121 b University of California San Diego, School of Medicine, 9500 Gilman Drive, La Jolla, CA 92093 Safe Harbor Statement Any statements contained in this presentation that do not describe historical facts constitute forward-looking statements as that term is defined in the Private Securities Litigation Reform Act of 1995. Any forward-looking statements contained herein are based on current expectations, but are subject to a number of risks and uncertainties. The factors that could cause the Company's actual future results to differ materially from current expectations 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 business, research, product development, regulatory approval, marketing and distribution plans and strategies, including its use of third party distributors; the Company's ability to successfully complete the contracts and recognize the revenue represented by the contracts included in its previously reported total contract bookings and secure additional contracted collaborative relationships; the final results of the Company's preclinical studies may be different from the Company's studies or interim preclinical data results and may not support further clinical development of its therapeutic tissues; the Company may not successfully complete the required preclinical and clinical trials required to obtain regulatory approval for its therapeutic tissues on a timely basis or at all; 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 SEC, including its Annual Report on Form 10-K filed with the SEC on June 9, 2016 and its Quarterly Report on Form 10-Q filed with the SEC on February 9, 2017. You should not place undue reliance on these forward-looking statements, which speak only as of the date that they were made. These cautionary statements should be considered with any written or oral forward-looking statements that the Company may issue in the future. Except as required by applicable law, including the securities laws of the United States, the Company does 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. Poster No. 1963 Steatosis Induction in 3D Bioprinted Liver Tissues CHANGING THE SHAPE OF RESEARCH AND MEDICINE Technology Overview Figure 1: 3D human tissue development using the NovoGen Bioprinter® Platform. Cells reside in heterogeneous and architecturally structured 3D environments in vivo. Using the proprietary NovoGen Bioprinter® Platform, Organovo builds 3D tissues through automated, spatially-controlled cellular deposition to better recapitulate native tissue structure and function. Figure 2: [A] Schematic of ExVive™ Human Liver Tissue, comprised of primary human hepatocytes (HCs), hepatic stellate cells (HSCs), and endothelial cells (ECs) bioprinted in a compartmentalized geometry onto the membranes of 24-well transwell culture inserts. [B] Representative immunofluorescence image of 3D human liver tissue showing distinct zones of non-parenchymal cells (NPC) in green and parenchymal (HC) cells in red. [Norona, et al. (2016) Tox Sci. 154(2):354-367]. [C] ExVive™ Liver tissue exhibits sustained hepatocyte function as indicated by albumin levels versus standard 2D hepatocyte culture, as well as sustained CYP3A4 activity [D] (2D = matched hepatocytes, grown on collagen 1 coated plates). A. B. One-way ANOVA * p<0.05 ** p<0.001 *** p<0.001 **** p<0.0001 Day 3 Day 7 Day 14 Day 28 0 1 2 3 4 5 6 Fold Change Albumin vs Day 3 2D Bioprinted **** **** **** *** **** C. D. One-way ANOVA * p<0.05 ** p<0.001 *** p<0.001 **** p<0.0001 Figure 3: Characterization of bioprinted liver tissue with incorporation of Kupffer cells (KC). [A] Kupffer cells in bioprinted liver express prototypical markers such as CD68 and CD168, and a staining pattern similar to native liver. [B] ExVive™ Liver Tissue with Kupffer cells exhibited greater cytokine induction after lipopolysaccharide (LPS) treatment. Media samples from tissue treated with LPS (100 μg/mL for 24h) were analyzed via electrochemiluminesce. A. B. CD68 DAPI CD163 DAPI 3D Bioprinted Tissue Native Tissue ExVive Liver Tissue ExVive Liver Tissue + KCs IL-1β TNF-α ExVive Liver Tissue ExVive Liver Tissue + KCs Figure 4: Chronic exposure (21 days) of 3D bioprinted liver tissues to palmitic acid (PA) induces lipid droplet formation. H&E staining of PA treated tissues shows both macro- (arrows) and micro- (arrowheads) vesicular phenotypes. Perilipin (PLIN5) staining of lipid vesicles further confirms the steatotic phenotype. Control (glucose) + PA [low] + PA [high] Native Liver H&E H&E PLIN Perilipin Oil Red O Control +PA [low] +PA [high] Native tissues from Samsara Sciences, Inc. Figure 5: Chronic exposure of 3D bioprinted liver tissues to palmitic acid (PA) induces increased lipid accumulation (steatosis). Oil Red O (ORO) staining of PA treated tissues shows a dose dependent increase in steatosis. ORO Control PA [low] PA [high] Lipid Accumulation [Fold x] One-way ANOVA * p<0.05 ** p<0.001 **** p<0.0001 Control Initial Conditions (21 Days) Optimized Conditions (7 Days) H&E + PA (High) + PA (High) +PA only +FFA [A] +FFA [B] +FFA [C] +FFA [D] Control H&E H&E Figure 6: [A] Earlier onset of steatosis under optimized conditions. [B] Optimized regimen of FFA’s increases degree of steatotic phenotype. H&E H&E A. B. Native Liver NASH-Induced 3D Bioprinted Liver Tissues ExVive + KC - LPS - PA - + + + + Native NASH aSMA DESMIN DAPI F F F B. A. F F ExVive + KC - IL1b - PA - + + + + Native NASH TCM F F C. Figure 9: Chronic dosing of Immune competent 3D bioprinted liver tissues with inflammatory inducer (LPS) and palmitic acid (PA) on a high sugar background led to [A] Increased activation of hepatic stellate cells (α-SMA staining), and subsequent increased fibrosis (F; regions of increased collage staining in blue) and steatosis (arrows), similar to native tissue samples and [B] Significantly increased inflammatory cytokine levels. Chronic dosing with inflammatory cytokine IL1β along with PA on a high sugar background led to [C] Increased fibrosis (F), similar to native tissue samples. F TCM Control (glucose only) Trichrome + Sugars + Sugars + FFA [C] Figure 7: Titration of sugars and fatty acids alone can induce fibrosis (red arrows). Addition of sugars and FFA’s appear to induce microvesicular (green arrowhead) and macrovesicular steatosis (green arrows). TCM Normal Steatosis NASH H&E H&E Native tissues from Samsara Sciences, Inc. NASH Induction in 3D Bioprinted Liver Tissues H&E Figure 8: Immune competent (+ Kupffer cells) 3D bioprinted liver tissues treated with high dose FFA regimens displayed significantly increased triglyceride (TG) accumulation vs. controls. The fold changes seen in bioprinted tissues correlate with fold changes seen in native NASH tissues. Trichrome Trichrome Steatosis Induction in 3D Bioprinted Liver Tissues Cont’d Native Liver Summary and Conclusion Key features of NASH such as steatosis, increased inflammatory cytokine release, hepatic stellate cell activation, and subsequent fibrogenesis, which are to date, largely lacking in other Nafld in vitro models are attainable in the fully human ExVive™ Liver Tissue via a nutrient overload approach, analogous to a western style diet and inflammatory stimuli. The longevity of the ExVive™ Liver Tissue allows for the testing of several induction strategies such as various dosing and durations of insults (nutrients, inflammatory inducers, xenobiotics), and also has the potential to overlay a range of modulatory approaches to profile prophylactic and treatment oriented drug strategies. Together, these features suggest that the ExVive Human Liver Tissue hold promise for the study of complex, chronic conditions such as NASH, which will enable a better understanding of disease processes, discovery of novel therapeutics, potential biomarkers, and the safety assessment of drugs in a disease-relevant background. Future Directions Refinement of induction protocol to achieve optimal steatosis and controlled stimulation of chronic inflammation representative of native disease onset. Correlation of in vitro model to clinical phenotype. Modulation of disease progression in vitro: testing reference compounds and client proprietary drug candidates. Funding Research reported in this publication was supported by the National Institute Of Diabetes And Digestive And Kidney Diseases of the National Institutes of Health under Award Number R44DK115242. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. References Norona, et al. (2016) Toxicological Sciences. 2016, Vol.154, No. 2. Nguyen, et al. (2016) PLoS One. 2016 Jul 7;11(7):e0158674. One-way ANOVA * p<0.05 ** p<0.001 Native tissues from Samsara Sciences, Inc. Native tissues from Samsara Sciences, Inc.

Transcript of Modeling NAFLD Using 3D Bioprinted Human Liver Tissue · Except as required by applicable law,...

Page 1: Modeling NAFLD Using 3D Bioprinted Human Liver Tissue · Except as required by applicable law, including the securities laws of the United States, the Company does not intend to update

BackgroundNonalcoholic fatty liver disease (NAFLD) is a chronic condition that originates as lipid accumulation within hepatocytes

(steatosis) and progresses into nonalcoholic steatohepatitis (NASH), characterized by lipid accumulation, inflammation,

oxidative stress, and fibrosis. NAFLD is now recognized as the most common cause of chronic liver disease, with a prevalence

of 25% worldwide, and is projected to become the leading indication for liver transplant by 2025. Despite decades of research,

the mechanisms of NAFLD progression, therapeutic approaches and non-invasive diagnostics are still resoundingly absent. The

study of steatosis and NASH has traditionally utilized rodent models, which are time consuming to generate and do not fully

recapitulate the complex phenotypes associated with the human disease. Furthermore, current 2D cell culture models lack

relevant liver cell types and have limited utility due to rapid loss of cell viability and function. Thus, there is a significant need

for a more predictive human multicellular 3D in vitro model to study the progression of steatosis into NASH.

MethodsExVive™ Human Liver Tissue, a human in vitro 3D bioprinted liver model comprising primary human hepatocytes, hepatic

stellate cells, and endothelial cells exhibits a complex multicellular architecture similar to that of native liver and retains

metabolic competence and liver-specific functions for at least 4 weeks in culture. To mimic the proposed pathogenesis of

NASH via a “Two-Hit Hypothesis”, standard ExVive™ Human Liver Tissue were supplemented with Kupffer cells to achieve

immune competency then exposed to steatogenic cues via a nutrient overload approach of simple sugars and fatty acids,

followed by inflammatory stimulation using prototypical inducers.

Modeling NAFLD Using 3D Bioprinted Human Liver TissueDwayne E. Cartera, Deborah G. Nguyena, David Brennerb, Sharon C. Presnella, and Alice E. Chena

aOrganovo, 6275 Nancy Ridge Drive, San Diego, CA 92121bUniversity of California San Diego, School of Medicine, 9500 Gilman Drive, La Jolla, CA 92093

Safe Harbor StatementAny statements contained in this presentation that do not describe historical facts constitute forward-looking statements as that term is defined in the Private Securities Litigation Reform Act of 1995. Any forward-looking statements contained herein are based on current expectations, but are subject to a number of risks and uncertainties. The factors that could cause the Company's actual future results to differ materially from current expectations include, but are not limited to, risks anduncertainties 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 business, research, product development, regulatory approval, marketingand distribution plans and strategies, including its use of third party distributors; the Company's ability to successfully complete the contracts and recognize the revenue represented by the contracts included in its previously reported total contract bookings and secure additional contracted collaborative relationships; the final results of the Company's preclinical studies may be different from the Company's studies or interim preclinical data results and may not support further clinical development ofits therapeutic tissues; the Company may not successfully complete the required preclinical and clinical trials required to obtain regulatory approval for its therapeutic tissues on a timely basis or at all; 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 SEC, including its Annual Report on Form 10-K filed with the SEC on June 9, 2016 and its Quarterly Reporton Form 10-Q filed with the SEC on February 9, 2017. You should not place undue reliance on these forward-looking statements, which speak only as of the date that they were made. These cautionary statements should be considered with any written or oral forward-looking statements that the Company may issue in the future. Except as required by applicable law, including the securities laws of the United States, the Company does not intend to update any of the forward-looking statements toconform these statements to reflect actual results, later events or circumstances or to reflect the occurrence of unanticipated events.

Poster No. 1963

Steatosis Induction in 3D Bioprinted Liver Tissues

CHANGING THE SHAPE OF RESEARCH AND MEDICINE

Technology Overview

Figure 1: 3D human tissue development using the NovoGen Bioprinter® Platform. Cells reside in heterogeneous and architecturallystructured 3D environments in vivo. Using the proprietary NovoGen Bioprinter® Platform, Organovo builds 3D tissues through automated,spatially-controlled cellular deposition to better recapitulate native tissue structure and function.

Figure 2: [A] Schematic of ExVive™ Human Liver Tissue,comprised of primary human hepatocytes (HCs), hepaticstellate cells (HSCs), and endothelial cells (ECs) bioprinted in acompartmentalized geometry onto the membranes of 24-welltranswell culture inserts. [B] Representativeimmunofluorescence image of 3D human liver tissue showingdistinct zones of non-parenchymal cells (NPC) in green andparenchymal (HC) cells in red. [Norona, et al. (2016) Tox Sci.154(2):354-367]. [C] ExVive™ Liver tissue exhibits sustainedhepatocyte function as indicated by albumin levels versusstandard 2D hepatocyte culture, as well as sustained CYP3A4activity [D] (2D = matched hepatocytes, grown on collagen 1coated plates).

A. B.

One-way ANOVA* p<0.05** p<0.001*** p<0.001**** p<0.0001

Day 3

Day 7

Day 1

4

Day 2

8

0

1

2

3

4

5

6

Fo

ld C

ha

ng

e A

lbu

min

vs

Da

y 3

2 D

B io p r in te d****

****

****

*******

C. D.

One-way ANOVA* p<0.05** p<0.001*** p<0.001**** p<0.0001

Figure 3: Characterization of bioprinted liver tissue with incorporation of Kupffer cells (KC). [A] Kupffer cells in bioprinted liver expressprototypical markers such as CD68 and CD168, and a staining pattern similar to native liver. [B] ExVive™ Liver Tissue with Kupffer cellsexhibited greater cytokine induction after lipopolysaccharide (LPS) treatment. Media samples from tissue treated with LPS (100 μg/mL for24h) were analyzed via electrochemiluminesce.

A. B.

CD68DAPI

CD163DAPI

3D Bioprinted Tissue Native Tissue

ExVive Liver Tissue ExVive Liver Tissue + KCs

IL-1β TNF-α

ExVive Liver Tissue ExVive Liver Tissue + KCs

Figure 4: Chronic exposure (21 days) of 3D bioprinted liver tissues to palmitic acid (PA) induces lipid droplet formation.H&E staining of PA treated tissues shows both macro- (arrows) and micro- (arrowheads) vesicular phenotypes. Perilipin(PLIN5) staining of lipid vesicles further confirms the steatotic phenotype.

Control (glucose) + PA [low] + PA [high] Native Liver

H&

E

H&E

PLIN

Pe

rilip

in

Oil

Re

d O

Control +PA [low] +PA [high]

Na

tive

tis

sues

fro

m S

am

sara

Sci

ence

s, In

c.

Figure 5: Chronic exposure of 3D bioprinted liver tissues to palmitic acid (PA) induces increased lipid accumulation(steatosis). Oil Red O (ORO) staining of PA treated tissues shows a dose dependent increase in steatosis.

ORO

Control PA [low] PA [high]

Lip

id A

ccu

mu

lati

on

[Fo

ld x

]

One-way ANOVA* p<0.05** p<0.001**** p<0.0001

Control

Initial Conditions (21 Days)

Optimized Conditions (7 Days)

H&

E

+ PA (High) + PA (High)

+PA only +FFA [A]

+FFA [B] +FFA [C] +FFA [D]

Control

H&

EH

&E

Figure 6: [A] Earlier onset of steatosis under optimized conditions. [B] Optimized regimen of FFA’s increases degree ofsteatotic phenotype.

H&E

H&E

A.

B.

Native Liver

NASH-Induced 3D Bioprinted Liver Tissues

ExVive™ +KC -LPS -PA -

++++

Native NASH

aSMADESMINDAPI

FF

F

B.

A.

FF

ExVive™ +KC -IL1b -PA -

++++

Native NASH

TCM

F FC.

Figure 9: Chronic dosing of Immune competent 3D bioprinted liver tissues with inflammatory inducer (LPS) and palmitic acid (PA)on a high sugar background led to [A] Increased activation of hepatic stellate cells (α-SMA staining), and subsequent increasedfibrosis (F; regions of increased collage staining in blue) and steatosis (arrows), similar to native tissue samples and [B]Significantly increased inflammatory cytokine levels. Chronic dosing with inflammatory cytokine IL1β along with PA on a highsugar background led to [C] Increased fibrosis (F), similar to native tissue samples.

F

TCM

Control (glucose only)

Tric

hro

me

+ Sugars + Sugars + FFA [C]

Figure 7: Titration of sugars and fatty acids alone can induce fibrosis (red arrows). Addition of sugars and FFA’s appear toinduce microvesicular (green arrowhead) and macrovesicular steatosis (green arrows).

TCM

Normal Steatosis NASH

H&E

H&E

Na

tive

tis

sues

fro

m S

am

sara

Sci

ence

s, In

c.

NASH Induction in 3D Bioprinted Liver Tissues

H&

E

Figure 8: Immune competent (+ Kupffer cells) 3D bioprinted liver tissues treated with high dose FFA regimens displayedsignificantly increased triglyceride (TG) accumulation vs. controls. The fold changes seen in bioprinted tissues correlatewith fold changes seen in native NASH tissues.

Tric

hro

me

Tric

hro

me

Steatosis Induction in 3D Bioprinted Liver Tissues Cont’d

Native Liver

Summary and ConclusionKey features of NASH such as steatosis, increased inflammatory cytokine release, hepatic stellate cell activation, and subsequent

fibrogenesis, which are to date, largely lacking in other Nafld in vitro models are attainable in the fully human ExVive™ Liver Tissue

via a nutrient overload approach, analogous to a western style diet and inflammatory stimuli. The longevity of the ExVive™ Liver

Tissue allows for the testing of several induction strategies such as various dosing and durations of insults (nutrients, inflammatory

inducers, xenobiotics), and also has the potential to overlay a range of modulatory approaches to profile prophylactic and

treatment oriented drug strategies. Together, these features suggest that the ExVive™ Human Liver Tissue hold promise for the

study of complex, chronic conditions such as NASH, which will enable a better understanding of disease processes, discovery of

novel therapeutics, potential biomarkers, and the safety assessment of drugs in a disease-relevant background.

Future Directions• Refinement of induction protocol to achieve optimal steatosis and controlled stimulation of chronic inflammation

representative of native disease onset.

• Correlation of in vitro model to clinical phenotype.

• Modulation of disease progression in vitro: testing reference compounds and client proprietary drug candidates.

FundingResearch reported in this publication was supported by the National Institute Of Diabetes And Digestive And Kidney Diseases of the National Institutes of Health under Award Number R44DK115242. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

References• Norona, et al. (2016) Toxicological Sciences. 2016, Vol.154, No. 2.• Nguyen, et al. (2016) PLoS One. 2016 Jul 7;11(7):e0158674.

One-way ANOVA* p<0.05** p<0.001

Na

tive

tis

sues

fro

m S

am

sara

Sci

ence

s, In

c.N

ati

ve t

issu

es f

rom

Sa

msa

ra S

cien

ces,

Inc.