Feeds:
Posts
Comments

Posts Tagged ‘preclinical testing’

Ninth Annual
New Approaches for Predicting Drug Toxicity
Discovering New Models and Integrating Innovative Strategies
June 15-16, 2016  |  Boston, MA
WorldPreclinicalCongress.com/Drug-Safety-Conference

Final Agenda Now Available

Adverse drug events such as cardiotoxicity, hepatotoxicity and other organ toxicities, keep surfacing in the clinic and idiosyncratic drug toxicity continues to haunt the drug development process. So what are scientists and clinicians doing to make sure that compounds fail early and cheaply? New screening technologies such as, in vitro assays and in vivo models continue to be developed, but are the right tools being used at the right time to predict and detect adverse events? Cambridge Healthtech Institute’s ninth annual conference on Models to Approaches, looks at the scientific and technological progress being made to better predict drug related toxicities at the preclinical stage, and avoid unexpected and costly findings in the clinic. What assays and models are being used, how reliable and predictable is the data, and how is this information impacting decisions before compounds are tested in patients? Hear experiences shared by experts and join the interactive sessions and panel discussions on issues related to drug toxicity.

Register Now!  [Register by March 4th and save up to $400]

Agenda-at-a-Glance


Day 1

DRUG TRANSPORTERS AND THEIR ROLE IN DRUG TOXICITY

Transporter-Mediated Drug Interactions with Endobiotics, Toxins and Nutrients
Adrian Ray, Ph.D., Senior Director, Department of Drug Metabolism, Gilead Sciences, Inc.

Combination of Top-Down and Bottom-Up Strategy to Elucidate Mechanistic Roles of Transporters in Organ Toxicity
Yurong Lai, Ph.D., Senior Principal Scientist, Pharmaceutical Candidate Optimization, Bristol-Myers Squibb

In vitro Human Intestinal Tissue Model to Assess and Predict Drug-Induced-GI Damage
MatTek Corporation
Seyoum Ayehunie, Ph.D., Vice President, Immunological Systems, MatTek Corporation

Assessing Off-Target Drug Activities by Transcription Factor Profiling in FACTORIAL™ AssaysAttagene
Sergei Makarov, Ph.D., CEO, Attagene

UNDERSTANDING TRANSLATIONAL CHALLENGES AND INTERPRETING SAFETY GUIDELINES

The Importance of Reverse Translation for Preclinical Off-Target Mitigation
Laszlo Urban, M.D., Ph.D., Global Head, Preclinical Secondary Pharmacology, Novartis Institutes for BioMedical Research, Inc.

Moving beyond the S6(R1): A Snapshot of Toxicity & Safety Pharmacology Tools to Evaluate Biotherapeutics
Susan M.G. Goody, Ph.D., Senior Principal Scientist, Global Safety Pharmacology, Pfizer, Inc.

Presentation to be AnnouncedMolecular Health

Luncheon Presentation: CiPA: How Comprehensive Does It Have to BeCharles River Discovery
James Kramer, Ph.D., Principal Scientist, Discovery, Charles River

IN VIVO TECHNIQUES FOR MONITORING DRUG TOXICITY

A Disruption of Autonomic Balance: Use of Heart Rate Variability (HRV) in Cardiovascular Safety Pharmacology
Carrie Northcott, Ph.D., Senior Principal Scientist, Global Safety Pharmacology, Pfizer Inc.

Whole-Body Imaging of Drug-Induced Toxicity
Ming Zhao, Ph.D., Associate Professor, Feinberg School of Medicine, Northwestern University

NEW IN VITRO SCREENING APPROACHES FOR SAFETY TESTING

Combination of Screening Assays for Assessing Drug-Induced Liver Injury in Humans
Christoph Funk, Ph.D., Vice Director, Pharmaceutical Sciences, F. Hoffmann-La Roche

In vitro Approach to Classify Drugs According to Their Idiosyncratic, Drug-Induced Liver Injury Liability
Robert A. Roth, Ph.D., DABT, Professor of Pharmacology and Toxicology and Director, Graduate Program in Environmental and Integrative Toxicological Sciences, Michigan State University

Generation of Complex Disease Phenotypes in 3D Bioprinted Human Liver Tissues for the Assessment of Drug-Induced InjuryOrganovo
Leah Norona, Doctoral Candidate Curriculum in Toxicology, University of North Carolina at Chapel Hill

Drug-Induced Vascular Injury (DIVI)- Historical Review of Non-Clinical DIVI and Development of an Early Screening Strategy
Todd Wisialowski, MS, Associate Research Fellow, Global Safety Pharmacology, Pfizer Inc.

Day 2

INTERACTIVE BREAKOUT DISCUSSION GROUPS

TOPIC: Safety Assessments for Biologics
Moderator: Susan M.G. Goody, Ph.D., Senior PrincipalScientist, Global Safety Pharmacology, Pfizer, Inc.

TOPIC: Translation of Preclinical Findings to Clinic
Moderators: Carrie Northcott, Ph.D., Senior Principal Scientist, Global Safety Pharmacology, Pfizer Inc. Ming Zhao, Ph.D., Associate Professor, Feinberg School of Medicine, Northwestern University

TOPIC: Using iPSC for Drug Safety Screening
Moderators: Paul W. Burridge, Ph.D., Assistant Professor, Department of Pharmacology, Center for Pharmacogenomics, Northwestern University Feinberg School ofMedicine Xi Yang, Ph.D., DABT, Principal Investigator, Division of Systems Biology, National Center for Toxicological Research (NCTR), U.S. FDA

TOPIC: Key Issues Related to Drug Transporters in a Pharma R&D Setting
Moderator: Christoph Funk, Ph.D., Vice Director, Pharmaceutical Sciences, F. Hoffmann La-Roche

USE OF iPS CELLS FOR DRUG TOXICITY SCREENING

Utilization of iPSCs in Developing Human-on-a-Chip Systems for Phenotypic Screening Applications
James J. Hickman, Ph.D., Founding Director, NanoScience Technology Center; Professor, Nanoscience Technology, Chemistry, Biomolecular Science, Material Science and Electrical Engineering, University of Central Florida

Human-Induced Pluripotent Stem Cells Recapitulate Breast Cancer Patients’ Predilection to Doxorubicin-Induced Cardiotoxicity
Paul W. Burridge, Ph.D., Assistant Professor, Department of Pharmacology, Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine

Utilization of Induced Pluripotent Stem Cells to Understand Tyrosine Kinase Inhibitors (TKIs)-Induced Hepatotoxicity
Qiang Shi, Ph.D., Principal Investigator, Division of Systems Biology, National Center for Toxicological Research (NCTR), U.S. FDA

UNDERSTANDING MECHANISMS TO BETTER PREDICT DRUG TOXICITY

Predict Tyrosine Kinase Inhibitors (TKIs)-Induced Cardiotoxicity Using Induced Pluripotent Stem Cell-Derived Cardiomyocytes
Xi Yang, Ph.D., DABT, Principal Investigator, Division of Systems Biology, National Center for Toxicological Research (NCTR), U.S. FDA

Prediction of Transporter-Related Drug-Induced Liver Injury (DILI) Using Integrated Approaches
Mingxiang Liao, Ph.D., Senior Scientist I, DMPK, Takeda Pharmaceutical Intl. Company

Bridging Luncheon Presentation: Case Studies in Cardiac and Neuro Safety / Toxicity Assessment Using Human iPSC-Derived Cell SystemsAxioGenesis
Greg Luerman, Ph.D., Head, Applications Development, Axiogenesis Inc.

Plenary Sessions

June 16, 1:45-2:45 pm
PLENARY KEYNOTE PRESENTATIONS:

 

INSIGHTS ON INNOVATIVE APPROACHES TO TRANSFORM DRUG DISCOVERY

This year’s Plenary Keynote Presentations feature two prominent thought-leaders who are playing an important role in innovating drug discovery. They share their experiences and their perspectives on what has changed and what can be changed to improve preclinical research, help translate preclinical findings to the clinic, and to foster effective communication and collaboration. Attendees will have an opportunity to ask questions and gain valuable insights from their learnings.

Keynote Speakers:
Anthony CoyleAnthony J. Coyle Ph.D., Chief Scientific Officer and Senior Vice President, Centers for Therapeutic Innovation, Pfizer Inc.

 

James WilsonJames Wilson, M.D., Ph.D., Professor, Department of Pathology and Laboratory Medicine, Perelman School of Medicine; Director, Orphan Disease Center and Director, Gene Therapy Program, University of Pennsylvania

 

June 16, 2:45-3:30 pm
PLENARY KEYNOTE PANEL:

 

INSIGHTS ON INNOVATIVE TECHNOLOGIES ENABLING PRECLINICAL RESEARCH

This year’s Plenary Keynote Panel features a group of technical experts from life science technology and service companies, who share their perspectives on various trends and tools that will likely change the way in which we traditionally approach preclinical drug discovery and development. Attendees will have an opportunity to ask questions and understand the impact of recent technical advances.

Panelists:
Matthew GevaertMatt Gevaert, Ph.D., CEO and Co-founder, KIYATEC

 

Amit VasanjiAmit Vasanji, Ph.D., CTO & CSO, ImageIQ

 

Biographies:

Dr. Anthony Coyle is the founding CSO of the Centers for Therapeutic Innovation (CTI) and is responsible for CTI’s strategy and scientific direction. Before leading CTI, Dr. Coyle was the Vice President and Global Head of Respiratory, Inflammation, and Autoimmunity Research at MedImmune Biologics, a Division of AstraZeneca. At MedImmune, Dr. Coyle advanced a biologic portfolio from discovery to Phase II in the areas of respiratory and autoimmune diseases, specifically targeting lupus, asthma and COPD. Prior to his work at MedImmune, Dr. Coyle was Director of Research at Millennium Pharmaceuticals, where he led a group responsible for the identification of novel target genes, as well as for late stage lead optimization and delivery of both small-molecule and biologic development candidates. Dr. Coyle has been Associate Professor in the Department of Pathology and Experimental Therapeutics at McMaster University in Ontario since 1992. He has authored more than 200 manuscripts. Dr. Coyle holds a BSc (with honors) and a Ph.D. from Kings College, University of London. Dr. Coyle is a member of the scientific board for the Alliance for Lupus Research, the C4 NCATS consortium and the Boston Children’s Hospital Technology Fund Advisory Board.

Dr. James M. Wilson is a Professor in the Perelman School of Medicine at the University of Pennsylvania where he has led an effort to develop the field of gene therapy. Dr. Wilson began his work in gene therapy during his graduate studies at the University of Michigan over 30 years ago. He then moved to Boston to do a residency in Internal Medicine at the Massachusetts General Hospital and continued his work in gene therapy at MIT. He created the first and largest academic-based program in gene therapy after being recruited to University of Pennsylvania in 1993. He initially focused on the clinical translation of existing gene transfer technologies but soon redirected his efforts to the development of second and third generation gene transfer platforms; the first of which was licensed to a biotechnology company he founded that resulted in the first, and only, commercially approved gene therapy in the western hemisphere. More recently, his laboratory discovered a family of viruses from primates that could be engineered to be very effective gene transfer vehicles. These so called “vectors” have become the technology platform of choice and have set the stage for the recent resurgence of the field of gene therapy. Dr. Wilson has also been active in facilitating the commercial development of these new gene therapy platforms through the establishment of several biotechnology companies. Throughout his career, the focus of Dr. Wilson’s research has been rare inherited diseases, ranging from cystic fibrosis to dyslipidemias to a variety of metabolic disorders. Dr. Wilson has published over 550 papers, reviews, commentaries and editorials in the peer-reviewed literature and is an inventor on over 117 patents.

Dr. Matthew (Matt) Gevaert is the CEO of KIYATEC Inc., a life sciences company in Greenville, SC. KIYATEC specializes in ex vivo 3D cell culture and tissue systems that more accurately replicate in vivo human biology and function, with a focus on methods to accurately predict individual cancer patients’response to drugs by culturing and treating live patient derived primary cells. Dr. Gevaert co-founded the company and has served as CEO since 2007. Possessing a background which combines both business and technology, before his role at KIYATEC Dr. Gevaert led the commercialization of Clemson University’s biomedical and biotechnology intellectual property portfolio for nearly 5 years, working with both entrepreneurial start-ups and large, industry leading corporations. He has previous experience with Merck, 3M and Dow Chemica l, and has been published in Science magazine and the journal of the US National Academy of Engineering. Currently he serves as a board member of SCBIO, the state of South Carolina’s life science industry organization, and a board member of NEXT, which provides entrepreneur services and infrastructure to high-growth ventures in Greenville and Upstate South Carolina. Dr. Gevaert grew up the fifth of six children on a farm in Ontario, Canada and graduated from the University of Waterloo with a bachelor’s degree in Applied Chemistry. He also holds a master’s degree and a doctorate in Bioengineering from Clemson University. He maintains current appointments as adjunct professor in the Clemson University Department of Bioengineering and as a lecturer in the Clemson MBA in Entrepreneurship & Innovation.

Dr. Amit Vasanji has over 17 years of experience with basic and clinical research image acquisition, processing, analysis, visualization and biomedical software engineering. He was the founder of Cleveland Clinic’s Biomedical Imaging and Analysis Center, and served as its Executive Director. During his tenure at the Cleveland Clinic, he authored over 50 publications — many in high impact journals, participated in the writing of numerous federally funded grants, served as a consultant and/or co-investigator on many federal, state, corporate, and institutional grants, presented at national scientific meetings, and won various awards for innovation and service. Dr. Vasanji received a BS in Biomedical Engineering from the University of Miami, and a Ph.D. in biomedical engineering from Case Western Reserve University

Read Full Post »

Humanized Mice May Revolutionize Cancer Drug Discovery

Curator: Stephen J. Williams, Ph.D.

Humanized Mice May Revolutionize Cancer Drug Discovery

Word Cloud by Zach Day

Decades ago cancer research and the process of oncology drug discovery was revolutionized by the development of mice deficient in their immune system, allowing for the successful implantation of human-derived tumors. The ability to implant human tumors without rejection allowed researchers to study how the kinetics of human tumor growth in its three-dimensional environment, evaluate potential human oncogenes and drivers of oncogenesis, and evaluate potential chemotherapeutic therapies. Indeed, the standard preclinical test for antitumor activity has involved the subcutaneous xenograft model in immunocompromised (SCID or nude athymic) mice. More detail is given in the follow posts in which I describe some early pioneers in this work as well as the development of large animal SCID models:

Heroes in Medical Research: Developing Models for Cancer Research

The SCID Pig: How Pigs are becoming a Great Alternate Model for Cancer Research

The SCID Pig II: Researchers Develop Another SCID Pig, And Another Great Model For Cancer Research

This strategy (putting human tumor cells into immunocompromised mice and testing therapeutic genes and /or compounds) has worked extremely well for most cytotoxic chemotherapeutics (those chemotherapeutic drugs with mechanisms of action related to cell kill, vital cell functions, and cell cycle). For example the NCI 60 panel of human tumor cell lines has proved predictive for the chemosensitivity of a wide range of compounds.

Even though the immunocompromised model has contributed greatly to the chemotherapeutic drug discovery process. using these models to develop the new line of immuno-oncology products has been met with challenges three which I highlight below with curated database of references and examples.

From a practical standpoint development of a mouse which can act as a recipient for human tumors yet have a humanized immune system allows for the preclinical evaluation of antitumoral effect of therapeutic antibodies without the need to use neutralizing antibodies to the comparable mouse epitope,   thereby reducing the complexity of the study and preventing complications related to pharmacokinetics.

Champions Oncology Files Patents for Use of PDX Platform in Immune-Oncology

Hackensack, NJ – August 17, 2015 – Champions Oncology, Inc. (OTC: CSBR), engaged in the development of advanced technology solutions and services to personalize the development and use of oncology drugs, today announced that it has filed two patent applications with the United States Patent and Trademark Office (USPTO) relating to the development and use of mice with humanized immune systems to test immune-oncology drugs and therapeutic cancer vaccines.

Dr. David Sidransky, the founder and Chairman of Champions Oncology commented, “Drug development ‎in the immune-oncology space is fundamentally changing our approach to cancer treatment. These patents represent potentially invaluable tools for developing and personalizing immune therapy based on cutting edge sequence analysis, bioinformatics and our unique in vivo models.”

Joel Ackerman, Chief Executive Officer of Champions Oncology stated, “Developing intellectual property related to our Champions TumorGraft® platform has been an important component of strategy. The filing of these patents is an important milestone in leveraging our research and development investment to expand our platform and create proprietary tools for use by our pharmaceutical partners. We continue to look for additional revenue streams to supplement our fee-for-service business and we believe these patents will help us capture more of the value we create for our customers in the future.”

The first patent filing covers the methodology used by the Company to create a mouse model, containing a humanized immune system and a human tumor xenograft, which is capable of testing the efficacy of immune-oncology agents, both as single agents and in combination with anti-neoplastic drugs. The second patent filing relates to the detection of neoantigens and their role in the development of anti-cancer vaccines.

Keren Pez, Chief Scientific Officer, explained, “In the last few years, there has been a significant increase in cancer research that focuses on exploring the power of the human immune system to attack tumors. However, it’s challenging to test immune-oncology agents in traditional animal models due to the major differences between human and murine immune systems. The Champions ImmunoGraft™ platform has the unique ability of mimicking a human adaptive immune response in the mice, which allows us to specifically evaluate a variety of cancer therapeutics that modulate human immunity.

“Therapeutic vaccines that trigger the immune system to mount a response against a growing tumor are another area of intense interest. The development of an effective vaccine remains challenging but has an outstanding curative potential. Tumors harbor mutations in DNA that result in the translation of aberrant proteins. While these proteins have the potential to provoke an immune response that destructs early-stage cancer development, often the immune response becomes insufficient. Vaccines can trigger it by proactively challenging the system with these specific mutated peptides. Nevertheless, developing anti-cancer vaccines that effectively inhibit tumor growth has been complicated, partially due to challenges in finding the critical mutations, among others difficulties. With the more recent advances in genome sequencing, it’s now possible to identify tumor-specific antigens, or neoantigens, that naturally develop as an individual’s tumor grows and mutates,” she continued.

Traumatic spinal cord injury in mice with human immune systems.

Carpenter RS, Kigerl KA, Marbourg JM, Gaudet AD, Huey D, Niewiesk S, Popovich PG.

Exp Neurol. 2015 Jul 17;271:432-444. doi: 10.1016/j.expneurol.2015.07.011. [Epub ahead of print]

Inflamm Bowel Dis. 2015 Jul;21(7):1652-73. doi: 10.1097/MIB.0000000000000446.

Use of Humanized Mice to Study the Pathogenesis of Autoimmune and Inflammatory Diseases.

Koboziev I1, Jones-Hall Y, Valentine JF, Webb CR, Furr KL, Grisham MB.

Author information

Abstract

Animal models of disease have been used extensively by the research community for the past several decades to better understand the pathogenesis of different diseases and assess the efficacy and toxicity of different therapeutic agents. Retrospective analyses of numerous preclinical intervention studies using mouse models of acute and chronic inflammatory diseases reveal a generalized failure to translate promising interventions or therapeutics into clinically effective treatments in patients. Although several possible reasons have been suggested to account for this generalized failure to translate therapeutic efficacy from the laboratory bench to the patient’s bedside, it is becoming increasingly apparent that the mouse immune system is substantially different from the human. Indeed, it is well known that >80 major differences exist between mouse and human immunology; all of which contribute to significant differences in immune system development, activation, and responses to challenges in innate and adaptive immunity. This inconvenient reality has prompted investigators to attempt to humanize the mouse immune system to address important human-specific questions that are impossible to study in patients. The successful long-term engraftment of human hematolymphoid cells in mice would provide investigators with a relatively inexpensive small animal model to study clinically relevant mechanisms and facilitate the evaluation of human-specific therapies in vivo. The discovery that targeted mutation of the IL-2 receptor common gamma chain in lymphopenic mice allows for the long-term engraftment of functional human immune cells has advanced greatly our ability to humanize the mouse immune system. The objective of this review is to present a brief overview of the recent advances that have been made in the development and use of humanized mice with special emphasis on autoimmune and chronic inflammatory diseases. In addition, we discuss the use of these unique mouse models to define the human-specific immunopathological mechanisms responsible for the induction and perpetuation of chronic gut inflammation.

J Immunother Cancer. 2015 Apr 21;3:12. doi: 10.1186/s40425-015-0056-2. eCollection 2015.

Human tumor infiltrating lymphocytes cooperatively regulate prostate tumor growth in a humanized mouse model.

Roth MD1, Harui A1.

Author information

Abstract

BACKGROUND:

The complex interactions that occur between human tumors, tumor infiltrating lymphocytes (TIL) and the systemic immune system are likely to define critical factors in the host response to cancer. While conventional animal models have identified an array of potential anti-tumor therapies, mouse models often fail to translate into effective human treatments. Our goal is to establish a humanized tumor model as a more effective pre-clinical platform for understanding and manipulating TIL.

METHODS:

The immune system in NOD/SCID/IL-2Rγnull (NSG) mice was reconstituted by the co-administration of human peripheral blood lymphocytes (PBL) or subsets (CD4+ or CD8+) and autologous human dendritic cells (DC), and animals simultaneously challenged by implanting human prostate cancer cells (PC3 line). Tumor growth was evaluated over time and the phenotype of recovered splenocytes and TIL characterized by flow cytometry and immunohistochemistry (IHC). Serum levels of circulating cytokines and chemokines were also assessed.

RESULTS:

A tumor-bearing huPBL-NSG model was established in which human leukocytes reconstituted secondary lymphoid organs and promoted the accumulation of TIL. These TIL exhibited a unique phenotype when compared to splenocytes with a predominance of CD8+ T cells that exhibited increased expression of CD69, CD56, and an effector memory phenotype. TIL from huPBL-NSG animals closely matched the features of TIL recovered from primary human prostate cancers. Human cytokines were readily detectible in the serum and exhibited a different profile in animals implanted with PBL alone, tumor alone, and those reconstituted with both. Immune reconstitution slowed but could not eliminate tumor growth and this effect required the presence of CD4+ T cell help.

CONCLUSIONS:

Simultaneous implantation of human PBL, DC and tumor results in a huPBL-NSG model that recapitulates the development of human TIL and allows an assessment of tumor and immune system interaction that cannot be carried out in humans. Furthermore, the capacity to manipulate individual features and cell populations provides an opportunity for hypothesis testing and outcome monitoring in a humanized system that may be more relevant than conventional mouse models.

Methods Mol Biol. 2014;1213:379-88. doi: 10.1007/978-1-4939-1453-1_31.

A chimeric mouse model to study immunopathogenesis of HCV infection.

Bility MT1, Curtis A, Su L.

Author information

Abstract

Several human hepatotropic pathogens including chronic hepatitis C virus (HCV) have narrow species restriction, thus hindering research and therapeutics development against these pathogens. Developing a rodent model that accurately recapitulates hepatotropic pathogens infection, human immune response, chronic hepatitis, and associated immunopathogenesis is essential for research and therapeutics development. Here, we describe the recently developed AFC8 humanized liver- and immune system-mouse model for studying chronic hepatitis C virus and associated human immune response, chronic hepatitis, and liver fibrosis.

PMID:

25173399

[PubMed – indexed for MEDLINE]

PMCID:

PMC4329723

Free PMC Article

Immune humanization of immunodeficient mice using diagnostic bone marrow aspirates from carcinoma patients.

Werner-Klein M, Proske J, Werno C, Schneider K, Hofmann HS, Rack B, Buchholz S, Ganzer R, Blana A, Seelbach-Göbel B, Nitsche U, Männel DN, Klein CA.

PLoS One. 2014 May 15;9(5):e97860. doi: 10.1371/journal.pone.0097860. eCollection 2014.

From 2015 AACR National Meeting in Philadelphia

LB-050: Patient-derived tumor xenografts in humanized NSG mice: a model to study immune responses in cancer therapy
Sunday, Apr 19, 2015, 3:20 PM – 3:35 PM
Minan Wang1, James G. Keck1, Mingshan Cheng1, Danying Cai1, Leonard Shultz2, Karolina Palucka2, Jacques Banchereau2, Carol Bult2, Rick Huntress2. 1The Jackson Laboratory, Sacramento, CA; 2The Jackson Laboratory, Bar Harbor, ME

References

  1. Paull KD, Shoemaker RH, Hodes L, Monks A, Scudiero DA, Rubinstein L, Plowman J, Boyd MR. J Natl Cancer Inst. 1989;81:1088–1092. [PubMed]
  2. Shi LM, Fan Y, Lee JK, Waltham M, Andrews DT, Scherf U, Paull KD, Weinstein JN. J Chem Inf Comput Sci. 2000;40:367–379. [PubMed]
  3. Monks A, Scudiero D, Skehan P, Shoemaker R, Paull K, Vistica D, Hose C, Langley J, Cronise P, Vaigro-Wolff A, et al. J Natl Cancer Inst. 1991;83:757–766. [PubMed]
  4. Potti A, Dressman HK, Bild A, et al. Genomic signatures to guide the use of chemotherapeutics. Nat Med. 2006;12:1294–1300. [PubMed]
  5. Baggerly KA, Coombes KR. Deriving chemosensitivity from cell lines: forensic bioinformatics and reproducible research in high-throughput biology. Ann Appl Stat. 2009;3:1309–1334.
  6. Carlson, B. Putting Oncology Patients at Risk Biotechnol Healthc. 2012 Fall; 9(3): 17–21.
  7. Salter KH, Acharya CR, Walters KS, et al. An Integrated Approach to the Prediction of Chemotherapeutic Response in Patients with Breast Cancer. Ouchi T, ed. PLoS ONE. 2008;3(4):e1908. NOTE RETRACTED PAPER

Other posts on this site on Animal Models, Disease and Cancer Include:

Heroes in Medical Research: Developing Models for Cancer Research

Guidelines for the welfare and use of animals in cancer research

Model mimicking clinical profile of patients with ovarian cancer @ Yale School of Medicine

Vaccines, Small Peptides, aptamers and Immunotherapy [9]

Immunotherapy in Cancer: A Series of Twelve Articles in the Frontier of Oncology by Larry H Bernstein, MD, FCAP

Mouse With ‘Humanized Version’ Of Human Language Gene Provides Clues To Language Development

The SCID Pig: How Pigs are becoming a Great Alternate Model for Cancer Research

The SCID Pig II: Researchers Develop Another SCID Pig, And Another Great Model For Cancer Research

Read Full Post »

 

The SCID Pig:  How Pigs are becoming a Great Alternate Model for Cancer Research[1]

Author/Writer: Stephen J. Williams, Phd

Article ID #80: The SCID Pig: How Pigs are becoming a Great Alternate Model for Cancer Research. Published on 10/10/2013

WordCloud Image Produced by Adam Tubman

UPDATED 3/14/2020

The need for alternate models of human cancer

Many worldwide regulatory bodies are in agreement that proper choice of animal model is necessary for adequate extrapolation of toxicity and efficacy data from animal to human, considering the varied classes of therapeutics now being developed for oncology.  The inability of screens, reliant on human xenografts grown in immunocompromised mice to evaluate host-immune and species-dependent effects, has made development of alternative animal-models a priority.   This is evident in the fact that ninety percent of new anticancer drugs which showed anti-tumor efficacy in mouse preclinical models failed in human clinical studies. A recently developed “humanized” mouse model may assist in testing the metabolism of cancer drugs but still relies on older “immunosuppression” mouse models (http://stehlin.org/mouse-model-development/). This inadequacy of older, accepted models is clearly evident when evaluating safety and efficacy of adenoviral based gene therapies such as oncolytic conditionally-replicative adenovirus (CRAd).  Although new-generation CRAds present with a relative safe profile[2, 3], adenoviral particles, especially the Ad5 based virus used for most CRAds, have the tendency to replicate in non-tumor tissue, such as liver and lung, resulting in tissue-specific toxicities[4-7].  The manifestation of these toxicities is only evident in species permissive for viral replication, such as the pig. Indeed, one of the first clinical trials with older adenovirus gene therapy, resulting in severe hepatic toxicity and fatality, may have been prevented if more appropriate preclinical screens were conducted.  Thereafter, strict regulatory guidelines for adenoviral-based clinical trials have been issued, with particular emphasis on vector dosage, safety and toxicity[8]. Indeed, at Schering-Plough, a toxicology program was initiated to evaluate SCH 58500, and adenoviral gene therapy directed against p53, which involved use of non-immunogenic rats compared with testing in Yorkshire pigs made immunoreactive to the vector[9, 10].  In fact, data from the pig study revealed a faster clearance of virus as well as toxicities not seen in non-immunogenic, non-permissive hosts such as rat and mouse.

Therefore, development of a porcine model of cancer would permit both testing of both the efficacy and safety of these therapies in the same animal.

Development of large animal models of cancer

To date, large animal tumor models have been used for studying spontaneously formed tumors in dogs and cats [11](Vail, 2000, Cancer Invest), the most common being canine [12] and feline non-Hodgkin’s lymphoma [13]. The advantages of these companion models are the outbred nature of the animals, comparable size and kinetics to human tumors [14-18], and high incidence rates. Allografts of the outbred-canine transplanted venereal tumor have been used to test the ability to detect tumors using X-ray computed tomography and MRI with the ultimate goal of imaging-guided intervention. Researchers have recently utilized the spontaneously arising canine and feline soft tissue sarcomas to study effects of hyperthermia on chemotherapy pharmocokinetics, development of hypoxic cell markers, and cancer imaging techniques [15, 19-26]

Although it appears that, for a select number of tumor types, spontaneously arising tumors in large outbred animals can be useful to model the human disease, it is disappointing these spontaneous arising tumors are limited to discrete tumor types. However, due to recent advances in sequencing of several domestic animal genomes and the development of new cloning strategies, it is now very feasible to utilize other animal models more relevant to human disease, notably the miniature pig.

gottingen minipigThe Gottingen mini-pig

Large animals in medical research: Advantages of the minipig

Due to recent advances in sequencing of several domestic animal genomes [27, 28] and the development of new organism cloning technologies [29-31], it is now very feasible to utilize other species to model human disease, notably the pig. The development of porcine models of human disease has gained much interest lately. Advantages include the resemblance in anatomy, physiology, and genetic makeup with the human, as well as new methods to manipulate the pig genome [32, 33]. To date, porcine models of human metabolic syndrome [34] and diabetes [35], aortic aneurism [36], infectious disease resistance [32, 37], seizure [38], neurologic syndromes [33], and pancreatitis [39] have been developed. Recently, a genetically-engineered porcine model of cystic fibrosis was produced in collaboration with investigators at University of Iowa and Exemplar Genetics [40-42]. Additionally, Cho et al. successfully transplanted spontaneously transformed leukemic and lymphatic tumor cells in a major histocompatibility complex (MHC)-defined inbred miniature swine model [43], suggesting feasibility of an ex vivo strategy to develop a porcine tumor model. Porcine models have, also, been used to develop, test and refine surgical [44, 45] and laparoscopic techniques [46, 47], radio- and cryoablation protocols of tissues [48-52] and robotic surgery using the da Vinci Surgical SystemÒ [53, 54].  In addition, because of the size of porcine organs and their resemblance to the human (in genetics) the minipig is very useful and abundant of a source to isolate specific cell types for in vitro studies.  Below is a figure showing the comparable size of human and porcine ovaries to the mouse and  ability to purify  porcine ovarian epithelial cells and their similarity to human and mouse ovarian epithelial cells.

newslidemousehumanpigovarysizejpeg

Figure 1.  The human and pig ovary have similar size and can yield a greater number of isolated cells than one can get from a mouse ovary.

posehosemosepicforpostjpg

Figure 2.  Isolation and morphology of ovarian epithelial cells from three sources:

A) Devonshire/Yorkshire pig

B) normal human ovary

c) SV129/BL6  mouse

note cobblestone epithelial morphology from all three sources©

To date, there has been no allograft or xenograft model of cancer in pigs. The consensus amongst many surgeons suggests development of a minipig tumor model would be an invaluable tool for developing surgical skills. 

A recent advancement in porcine tumor modeling was made by collaboration between researchers from the laboratories of Dr. Stefan Bossmann and Deryl Troyer at Kansas State and Iowa State, respectively[1].  The joint collaboration resulted in the development of the first severe combined immunodeficient pig line (SCID pig) which was shown to be able to accept human tumor xenografts.  The line of immunodeficient pig was discovered when Yorkshire pigs were bred for increased feed efficiency and a line of pigs exhibited SCID-like symptoms including:

  • Decreased levels of circulating lymphocytes
  • Atrophied thymus and lymph nodes

The SCID phenotype in mice have been ascribed to defects in a DNA-dependent protein kinase gene which prevents variable-diversity-joining [V(D)J] gene region recombination[55].  There have been multiple genetic defects found in humans resulting in SCID, including defects in adenylate kinase2, Janus kinase 3, the IL2 receptor, and the IL-7 receptor[56]. The SCID phenotype in this pig line has a simple autosomal recessive inheritance pattern which, as described below in an interview with the authors, allows for the propagation of this porcine line.

An important feature of SCID models is the ability of these animals to act as a recipient of human tumorigenic cell lines.  In fact, growth of cell lines in SCID mice is a common test for tumorigenicity.  Therefore, to test if these pigs could act as recipients for human cancer cell lines, the authors inoculated the SCID Yorkshire pigs with 4 million A3755M human melanoma cells or PANC1 human pancreatic carcinoma cells subcutaneously in the left and right ears respectively of three pigs.  Some features of the results include:

  • All injection sites showed evidence (either histologic or palpable) of tumor growth
  • Tumors showed characteristic histologic features of malignant neoplasm including
  1. Bizarre and atypical mitotic figures
  2. Anisocytosis (different cell sizes and shapes; feature of malignancy)
  3. Anisokaryosis (different size and shape of nucleus)
  • tumors stained with anti-human mitochondrial antibody (a marker of epithelial cancer cells) showed strong cytoplasmic staining of neoplastic cells
  • interestingly no necrotic regions in the tumor

 

scidpigfig1Figure 3. Visual evidence of human tumor cells growing in SCID pig ear (day 20). B) Same picture as A) but circle outlines growth.  From reference 1. Basel et al., used with permission from Mary Liebert.

It is interesting to note that these tumors only grew roughly 10 x 5.5 mm, which is genrally large enough to do preclinical studies but may be too expensive to be of use for xenograft studies.  However it would be very feasible to conduct allograft studies in these SCID pigs.

Dr. Jack Dekkers, C.F. Curtiss Distinguished Professor and Section Leader of Animal Breeding and Genetics at Iowa State University, was kind to answer a few questions about the SCID pig model.

Question: You had mentioned this line was identified after breeding Yorkshire pigs for increased feed efficiency.  Have you identified or hypothesize which altered pathway or molecular defect which results in a SCID phenotype?  Is this SCID phenotype a result of a metabolic syndrome these pigs could have?

Dr. Dekkers: We indeed identified the SCID phenotype in a line of pigs that we had selected for increased feed efficiency. However, I don’t think this phenotype has anything to do with the selection we practiced; it was either already present in the founders of the line or it was a random mutation that occurred in the line, independent of the selection for feed efficiency. We have narrowed the mutation that causes the SCID in our pigs down to a chromosomal region and have a very strong candidate gene in that region that we are currently pursuing.

Question: In your opinion, is it possible to produce a highly inbred immunocompromised strain of pig such as a Gottingen minipig?

Dr. Dekkers: We are working on breeding the SCID mutation into mini pigs. But in the meantime, we have used bone marrow transfer to create a male that is homozygous SCID (it’s an autosomal recessive) and reproducing. This allows us to produce litters that are 50% SCID and 50% normal (carriers) by mating him to carrier females.

REFERENCES

1.         Basel MT, Balivada S, Beck AP, Kerrigan MA, Pyle MM, Dekkers JC, Wyatt CR, Rowland RR, Anderson DE, Bossmann SH et al: Human xenografts are not rejected in a naturally occurring immunodeficient porcine line: a human tumor model in pigs. BioResearch open access 2012, 1(2):63-68.

2.         Dobbelstein M: Replicating adenoviruses in cancer therapy. Curr Top Microbiol Immunol 2004, 273:291-334.

3.         Lichtenstein DL, Wold WS: Experimental infections of humans with wild-type adenoviruses and with replication-competent adenovirus vectors: replication, safety, and transmission. Cancer Gene Ther 2004, 11(12):819-829.

4.         Volpers C, Kochanek S: Adenoviral vectors for gene transfer and therapy. J Gene Med 2004, 6 Suppl 1:S164-171.

5.         Brand K, Arnold W, Bartels T, Lieber A, Kay MA, Strauss M, Dorken B: Liver-associated toxicity of the HSV-tk/GCV approach and adenoviral vectors. Cancer Gene Ther 1997, 4(1):9-16.

6.         Lieber A, He CY, Meuse L, Schowalter D, Kirillova I, Winther B, Kay MA: The role of Kupffer cell activation and viral gene expression in early liver toxicity after infusion of recombinant adenovirus vectors. J Virol 1997, 71(11):8798-8807.

7.         Keedy V, Wang W, Schiller J, Chada S, Slovis B, Coffee K, Worrell J, Thet LA, Johnson DH, Carbone DP: Phase I study of adenovirus p53 administered by bronchoalveolar lavage in patients with bronchioloalveolar cell lung carcinoma: ECOG 6597. J Clin Oncol 2008, 26(25):4166-4171.

8.         Assessment of adenoviral vector safety and toxicity: report of the National Institutes of Health Recombinant DNA Advisory Committee. Hum Gene Ther 2002, 13(1):3-13.

9.         Morrissey RE, Horvath C, Snyder EA, Patrick J, Collins N, Evans E, MacDonald JS: Porcine toxicology studies of SCH 58500, an adenoviral vector for the p53 gene. Toxicol Sci 2002, 65(2):256-265.

10.       Morrissey RE, Horvath C, Snyder EA, Patrick J, MacDonald JS: Rodent nonclinical safety evaluation studies of SCH 58500, an adenoviral vector for the p53 gene. Toxicol Sci 2002, 65(2):266-275.

11.       Vail DM, MacEwen EG: Spontaneously occurring tumors of companion animals as models for human cancer. Cancer Invest 2000, 18(8):781-792.

12.       Leifer CE, Matus RE: Canine lymphoma: clinical considerations. Semin Vet Med Surg (Small Anim) 1986, 1(1):43-50.

13.       MacEwen EG: Spontaneous tumors in dogs and cats: models for the study of cancer biology and treatment. Cancer Metastasis Rev 1990, 9(2):125-136.

14.       Schwyn U, Crompton NE, Blattmann H, Hauser B, Klink B, Parvis A, Ruslander D, Kaser-Hotz B: Potential tumour doubling time: determination of Tpot for various canine and feline tumours. Vet Res Commun 1998, 22(4):233-247.

15.       Zeman EM, Calkins DP, Cline JM, Thrall DE, Raleigh JA: The relationship between proliferative and oxygenation status in spontaneous canine tumors. Int J Radiat Oncol Biol Phys 1993, 27(4):891-898.

16.       LaRue SM, Fox MH, Withrow SJ, Powers BE, Straw RC, Cote IM, Gillette EL: Impact of heterogeneity in the predictive value of kinetic parameters in canine osteosarcoma. Cancer Res 1994, 54(14):3916-3921.

17.       Vail DM, Kisseberth WC, Obradovich JE, Moore FM, London CA, MacEwen EG, Ritter MA: Assessment of potential doubling time (Tpot), argyrophilic nucleolar organizer regions (AgNOR), and proliferating cell nuclear antigen (PCNA) as predictors of therapy response in canine non-Hodgkin’s lymphoma. Exp Hematol 1996, 24(7):807-815.

18.       Guglielmino R, Canese MG, Miniscalco B, Geuna M: Comparison of clinical, morphological, immunophenotypical and cytochemical characteristics of LGL leukemia/lymphoma in dog, cat and human. Eur J Histochem 1997, 41 Suppl 2:23-24.

19.       Cline JM, Thrall DE, Rosner GL, Raleigh JA: Distribution of the hypoxia marker CCI-103F in canine tumors. Int J Radiat Oncol Biol Phys 1994, 28(4):921-933.

20.       Thrall DE, McEntee MC, Cline JM, Raleigh JA: ELISA quantification of CCI-103F binding in canine tumors prior to and during irradiation. Int J Radiat Oncol Biol Phys 1994, 28(3):649-659.

21.       Raleigh JA, La Dine JK, Cline JM, Thrall DE: An enzyme-linked immunosorbent assay for hypoxia marker binding in tumours. Br J Cancer 1994, 69(1):66-71.

22.       Thrall DE, Larue SM, Pruitt AF, Case B, Dewhirst MW: Changes in tumour oxygenation during fractionated hyperthermia and radiation therapy in spontaneous canine sarcomas. Int J Hyperthermia 2006, 22(5):365-373.

23.       Siddiqui F, Li CY, Larue SM, Poulson JM, Avery PR, Pruitt AF, Zhang X, Ullrich RL, Thrall DE, Dewhirst MW et al: A phase I trial of hyperthermia-induced interleukin-12 gene therapy in spontaneously arising feline soft tissue sarcomas. Mol Cancer Ther 2007, 6(1):380-389.

24.       Sostman HD, Prescott DM, Dewhirst MW, Dodge RK, Thrall DE, Page RL, Tucker JA, Harrelson JM, Reece G, Leopold KA et al: MR imaging and spectroscopy for prognostic evaluation in soft-tissue sarcomas. Radiology 1994, 190(1):269-275.

25.       Dennis R: Imaging features of orbital myxosarcoma in dogs. Vet Radiol Ultrasound 2008, 49(3):256-263.

26.       Mueller F, Fuchs B, Kaser-Hotz B: Comparative biology of human and canine osteosarcoma. Anticancer Res 2007, 27(1A):155-164.

27.       Cockett NE: Current status of the ovine genome map. Cytogenet Genome Res 2003, 102(1-4):76-78.

28.       Schook LB, Beever JE, Rogers J, Humphray S, Archibald A, Chardon P, Milan D, Rohrer G, Eversole K: Swine Genome Sequencing Consortium (SGSC): A Strategic Roadmap for Sequencing The Pig Genome. Comp Funct Genomics 2005, 6(4):251-255.

29.       Wilmut I, Schnieke AE, McWhir J, Kind AJ, Campbell KH: Viable offspring derived from fetal and adult mammalian cells. Nature 1997, 385(6619):810-813.

30.       Cibelli JB, Stice SL, Golueke PJ, Kane JJ, Jerry J, Blackwell C, Ponce de Leon FA, Robl JM: Cloned transgenic calves produced from nonquiescent fetal fibroblasts. Science 1998, 280(5367):1256-1258.

31.       Polejaeva IA, Chen SH, Vaught TD, Page RL, Mullins J, Ball S, Dai Y, Boone J, Walker S, Ayares DL et al: Cloned pigs produced by nuclear transfer from adult somatic cells. Nature 2000, 407(6800):86-90.

32.       Lunney JK: Advances in swine biomedical model genomics. Int J Biol Sci 2007, 3(3):179-184.

33.       Schook LB, Kuzmuk K, Adam S, Rund L, Chen K, Rogatcheva M, Mazur M, Pollock C, Counter C: DNA-based animal models of human disease: from genotype to phenotype. Dev Biol (Basel) 2008, 132:15-25.

34.       Spurlock ME, Gabler NK: The development of porcine models of obesity and the metabolic syndrome. J Nutr 2008, 138(2):397-402.

35.       Palin MF, Labrecque B, Beaudry D, Mayhue M, Bordignon V, Murphy BD: Visfatin expression is not associated with adipose tissue abundance in the porcine model. Domest Anim Endocrinol 2008, 35(1):58-73.

36.       Sadek M, Hynecek RL, Goldenberg S, Kent KC, Marin ML, Faries PL: Gene expression analysis of a porcine native abdominal aortic aneurysm model. Surgery 2008, 144(2):252-258.

37.       Saetre T, Hoiby EA, Aspelin T, Lermark G, Lyberg T: Acute serogroup A streptococcal shock: A porcine model. J Infect Dis 2000, 182(1):133-141.

38.       Marchi N, Angelov L, Masaryk T, Fazio V, Granata T, Hernandez N, Hallene K, Diglaw T, Franic L, Najm I et al: Seizure-promoting effect of blood-brain barrier disruption. Epilepsia 2007, 48(4):732-742.

39.       Tao J, Gong D, Ji D, Xu B, Liu Z, Li L: Improvement of monocyte secretion function in a porcine pancreatitis model by continuous dose dependent veno-venous hemofiltration. Int J Artif Organs 2008, 31(8):716-721.

40.       Rogers CS, Abraham WM, Brogden KA, Engelhardt JF, Fisher JT, McCray PB, Jr., McLennan G, Meyerholz DK, Namati E, Ostedgaard LS et al: The porcine lung as a potential model for cystic fibrosis. Am J Physiol Lung Cell Mol Physiol 2008, 295(2):L240-263.

41.       Rogers CS, Stoltz DA, Meyerholz DK, Ostedgaard LS, Rokhlina T, Taft PJ, Rogan MP, Pezzulo AA, Karp PH, Itani OA et al: Disruption of the CFTR gene produces a model of cystic fibrosis in newborn pigs. Science 2008, 321(5897):1837-1841.

42.       Rogers CS, Hao Y, Rokhlina T, Samuel M, Stoltz DA, Li Y, Petroff E, Vermeer DW, Kabel AC, Yan Z et al: Production of CFTR-null and CFTR-DeltaF508 heterozygous pigs by adeno-associated virus-mediated gene targeting and somatic cell nuclear transfer. J Clin Invest 2008, 118(4):1571-1577.

43.       Cho PS, Lo DP, Wikiel KJ, Rowland HC, Coburn RC, McMorrow IM, Goodrich JG, Arn JS, Billiter RA, Houser SL et al: Establishment of transplantable porcine tumor cell lines derived from MHC-inbred miniature swine. Blood 2007, 110(12):3996-4004.

44.       Hammond I, Taylor J, Obermair A, McMenamin P: The anatomy of complications workshop: an educational strategy to improve the training and performance of fellows in gynecologic oncology. Gynecol Oncol 2004, 94(3):769-773.

45.       Schneider C, Jung A, Reymond MA, Tannapfel A, Balli J, Franklin ME, Hohenberger W, Kockerling F: Efficacy of surgical measures in preventing port-site recurrences in a porcine model. Surg Endosc 2001, 15(2):121-125.

46.       Rassweiler JJ, Henkel TO, Potempa DM, Frede T, Stock C, Gunther M, Alken P: [Laparoscopic training in urology. An essential principle of laparoscopic interventions in the retroperitoneum]. Urologe A 1993, 32(5):393-402.

47.       Orvieto MA, Zorn KC, Lyon MB, Tolhurst SR, Rapp DE, Seip R, Sanghvi N, Shalhav A: High intensity focused ultrasound renal tissue ablation: a laparoscopic porcine model. J Urol 2009, 181(2):861-866.

48.       Ng KK, Lam CM, Poon RT, Shek TW, To JY, Wo YH, Ho DW, Fan ST: Comparison of systemic responses of radiofrequency ablation, cryotherapy, and surgical resection in a porcine liver model. Ann Surg Oncol 2004, 11(7):650-657.

49.       Alemany R, Balague C, Curiel DT: Replicative adenoviruses for cancer therapy. Nat Biotechnol 2000, 18(7):723-727.

50.       Kahlenberg MS, Volpe C, Klippenstein DL, Penetrante RB, Petrelli NJ, Rodriguez-Bigas MA: Clinicopathologic effects of cryotherapy on hepatic vessels and bile ducts in a porcine model. Ann Surg Oncol 1998, 5(8):713-718.

51.       Long JP, Faller GT: Percutaneous cryoablation of the kidney in a porcine model. Cryobiology 1999, 38(1):89-93.

52.       Scott DM, Young WN, Watumull LM, Lindberg G, Fleming JB, Rege RV, Brown RJ, Jones DB: Development of an in vivo tumor-mimic model for learning radiofrequency ablation. J Gastrointest Surg 2000, 4(6):620-625.

53.       Molpus KL, Wedergren JS, Carlson MA: Robotically assisted endoscopic ovarian transposition. Jsls 2003, 7(1):59-62.

54.       Hanly EJ, Marohn MR, Bachman SL, Talamini MA, Hacker SO, Howard RS, Schenkman NS: Multiservice laparoscopic surgical training using the daVinci surgical system. Am J Surg 2004, 187(2):309-315.

55.       Finnie NJ, Gottlieb TM, Blunt T, Jeggo PA, Jackson SP: DNA-dependent protein kinase defects are linked to deficiencies in DNA repair and V(D)J recombination. Philosophical transactions of the Royal Society of London Series B, Biological sciences 1996, 351(1336):173-179.

56.       Notarangelo LD: Primary immunodeficiencies. The Journal of allergy and clinical immunology 2010, 125(2 Suppl 2):S182-194.

UPDATED 3/14/2020

A recent Research Article and Research Article Summary in Science discusses, by the primary author of her study that describes the utility of the pig as an excellent surrogate model of the human brain and human brain function.  The study, by Dr. Evelina Sjostdedt et al., was an integrative analysis of porcine, mouse, and human transcriptomic, genomic, and proteomic data from discrete anatomical regions of the brain.  The global analysis suggested that there is similar regional organization and expression patterns among the three mammalian species.  The authors found interspecies variability with respect for many neurotransmitter receptors.

However, for some regions of the brain, such as the cerebellum and hypothalamus, the human global expression profile is closer to that of the pig than of the mouse, suggesting that the pig might be considered a preferred animal model to study many brain processes.

In addition, interestingly, the authors found that many signature genes canonically thought to only be expressed in certain brain cells (astrocytes, microglia, oligodendrocytes) are expressed in higher levels in peripheral organs as well as immune cells.

Please go to the full article in Science,

An atlas of the protein-coding genes in the human, pig, and mouse brain, 

Science  06 Mar 2020:
Vol. 367, Issue 6482, eaay5947
DOI: 10.1126/science.aay5947

to access the data used in this study, which includes high resolution images and metadata have also been made publicly available in the open-access Human Protein Atlas (HPA) Brain Atlas. at www.proteinatlas.org.

Other articles on this site pertaining to Alternate Animal Models and Cancer and Disease include:

Guidelines for the welfare and use of animals in cancer research

Demythologizing sharks, cancer, and shark fins

Predicting Drug Toxicity for Acute Cardiac Events

FDA Guidelines For Developmental and Reproductive Toxicology (DART) Studies for Small Molecules

Read Full Post »