Download as pdf or txt
Download as pdf or txt
You are on page 1of 53

Cancer Stem Cells Methods and

Protocols Methods in Molecular


Biology 2777 Papaccio
Visit to download the full and correct content document:
https://textbookfull.com/product/cancer-stem-cells-methods-and-protocols-methods-in
-molecular-biology-2777-papaccio/
More products digital (pdf, epub, mobi) instant
download maybe you interests ...

Cancer Cytogenetics Methods and Protocols Methods in


Molecular Biology 1541 Desconocido

https://textbookfull.com/product/cancer-cytogenetics-methods-and-
protocols-methods-in-molecular-biology-1541-desconocido/

Hepatic Stem Cells: Methods and Protocols Naoki


Tanimizu

https://textbookfull.com/product/hepatic-stem-cells-methods-and-
protocols-naoki-tanimizu/

Skin Stem Cells: Methods and Protocols Kursad Turksen

https://textbookfull.com/product/skin-stem-cells-methods-and-
protocols-kursad-turksen/

Stem Cell Renewal and Cell Cell Communication Methods


and Protocols Methods in Molecular Biology 2346 Kursad
Turksen Editor

https://textbookfull.com/product/stem-cell-renewal-and-cell-cell-
communication-methods-and-protocols-methods-in-molecular-
biology-2346-kursad-turksen-editor/
Neurobiology Methods and Protocols Methods in Molecular
Biology 2746 Sebastian Dworkin

https://textbookfull.com/product/neurobiology-methods-and-
protocols-methods-in-molecular-biology-2746-sebastian-dworkin/

Xylem Methods and Protocols Methods in Molecular


Biology 2722 Javier Agusti

https://textbookfull.com/product/xylem-methods-and-protocols-
methods-in-molecular-biology-2722-javier-agusti/

Teratogenicity Testing Methods and Protocols Methods in


Molecular Biology 2753 Luís Félix

https://textbookfull.com/product/teratogenicity-testing-methods-
and-protocols-methods-in-molecular-biology-2753-luis-felix/

Mucins Methods and Protocols Methods in Molecular


Biology 2763 1st Edition Kameyama

https://textbookfull.com/product/mucins-methods-and-protocols-
methods-in-molecular-biology-2763-1st-edition-kameyama/

Computational Stem Cell Biology Methods and Protocols


Patrick Cahan

https://textbookfull.com/product/computational-stem-cell-biology-
methods-and-protocols-patrick-cahan/
Methods in
Molecular Biology 2777

Federica Papaccio
Gianpaolo Papaccio Editors

Cancer
Stem Cells
Methods and Protocols
Second Edition
METHODS IN MOLECULAR BIOLOGY

Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, UK

For further volumes:


http://www.springer.com/series/7651
For over 35 years, biological scientists have come to rely on the research protocols and
methodologies in the critically acclaimed Methods in Molecular Biology series. The series was
the first to introduce the step-by-step protocols approach that has become the standard in all
biomedical protocol publishing. Each protocol is provided in readily-reproducible step-by
step fashion, opening with an introductory overview, a list of the materials and reagents
needed to complete the experiment, and followed by a detailed procedure that is supported
with a helpful notes section offering tips and tricks of the trade as well as troubleshooting
advice. These hallmark features were introduced by series editor Dr. John Walker and
constitute the key ingredient in each and every volume of the Methods in Molecular Biology
series. Tested and trusted, comprehensive and reliable, all protocols from the series are
indexed in PubMed.
Cancer Stem Cells

Methods and Protocols

Second Edition

Edited by

Federica Papaccio
Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno,
Baronissi, Italy

Gianpaolo Papaccio
Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
Editors
Federica Papaccio Gianpaolo Papaccio
Department of Medicine, Surgery and Department of Experimental Medicine
Dentistry “Scuola Medica Salernitana” University of Campania “Luigi Vanvitelli”
University of Salerno Naples, Italy
Baronissi, Italy

ISSN 1064-3745 ISSN 1940-6029 (electronic)


Methods in Molecular Biology
ISBN 978-1-0716-3729-6 ISBN 978-1-0716-3730-2 (eBook)
https://doi.org/10.1007/978-1-0716-3730-2
© The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Science+Business Media, LLC, part
of Springer Nature 2024
This work is subject to copyright. All rights are solely and exclusively licensed by the Publisher, whether the whole or part
of the material is concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation,
broadcasting, reproduction on microfilms or in any other physical way, and transmission or information storage and
retrieval, electronic adaptation, computer software, or by similar or dissimilar methodology now known or hereafter
developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication does not imply,
even in the absence of a specific statement, that such names are exempt from the relevant protective laws and regulations
and therefore free for general use.
The publisher, the authors, and the editors are safe to assume that the advice and information in this book are believed to
be true and accurate at the date of publication. Neither the publisher nor the authors or the editors give a warranty,
expressed or implied, with respect to the material contained herein or for any errors or omissions that may have been
made. The publisher remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

This Humana imprint is published by the registered company Springer Science+Business Media, LLC, part of Springer
Nature.
The registered company address is: 1 New York Plaza, New York, NY 10004, U.S.A.

Paper in this product is recyclable.


Preface

This second edition of the book Cancer Stem Cells: Methods and Protocols follows the
great success of its first edition, published in 2018.
In this second edition, a great effort has been made to provide readers with all the
scientific advances in methods and protocols related to cancer stem cells. They will, there-
fore, find both the most important updated chapters from the previous edition and new
chapters on hot topics.
The aim of the book is always to try to be a comprehensive collection of all methods,
protocols, and procedures used for the identification, characterization, and selection of
cancer stem cells. The new chapters focus on the latest technologies that have improved
our knowledge in this field. Among the newly included chapters, we feature contributions
on organoids, machine learning, nanoparticles, and other recent advances.
Regarding the topics of the new chapters, we believe that organoids, in particular,
represent a great challenge, and the related techniques are of great importance for cancer
stem cells thanks to the many applications they can have. At this point, it is very important to
be rigorous and understand the difference between spheroids and organoids.
We hope that the hard work we put into producing this book will be useful to scientists
who want to read it.

Baronissi, Italy Federica Papaccio


Naples, Italy Gianpaolo Papaccio

v
Contents

Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
1 Cancer Stem Cells: Current Challenges and Future Perspectives . . . . . . . . . . . . . . 1
Muhammad Vaseem Shaikh, Stefan Custers, Alisha Anand,
Petar Miletic, Chitra Venugopal, and Sheila K. Singh
2 Immunohistochemistry for Cancer Stem Cell Detection: Principles
and Methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
Giuseppa Zannini, Renato Franco, and Federica Zito Marino
3 Isolating Cancer Stem Cells from Solid Tumors . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35
Vitale Del Vecchio, Marcella La Noce, and Virginia Tirino
4 Surface Markers for the Identification of Cancer Stem Cells . . . . . . . . . . . . . . . . . . 51
Tasfik Ul Haque Pronoy, Farhadul Islam, Vinod Gopalan,
and Alfred King-yin Lam
5 CD44-Based Detection of CSCs: CD44 Immunodetection
by Flow Cytometry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 71
Lornella Seeneevassen, Anissa Zaafour, and Christine Varon
6 ALDH Activity Assay: A Method for Cancer Stem Cell (CSC)
Identification and Isolation. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
Vitale Del Vecchio, Marcella La Noce, and Virginia Tirino
7 In Vitro Tumorigenic Assay: A Tumor Sphere Assay for Cancer
Stem Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 91
Amani Yehya, Hisham Bahmad, and Wassim Abou-Kheir
8 Multicellular Tumoroids for Investigating Cancer Stem-Like
Cells in the Heterogeneous Tumor Microenvironment . . . . . . . . . . . . . . . . . . . . . . 99
Kathleen M. Burkhard and Geeta Mehta
9 Generation, Expansion, and Biobanking of Gastrointestinal
Patient-Derived Organoids from Tumor and Normal Tissues. . . . . . . . . . . . . . . . . 123
Manuel Cabeza-Segura, Blanca Garcia-Mico, Andrés Cervantes,
and Josefa Castillo
10 Prostate Cancer Organoids for Tumor Modeling and Drug Screening. . . . . . . . . 135
Amani Yehya, Fatima Ghamlouche, Sana Hachem, and Wassim Abou-
Kheir
11 Mimicking the Tumor Niche: Methods for Isolation, Culture,
and Characterization of Cancer Stem Cells and Multicellular Spheroids. . . . . . . . 145
Laura De Lara-Peña, Cristiano Farace, Andrea Pisano,
Julia Lopez de Andrés, Grazia Fenu, Federica Etzi,
Carmen Griñán-Lison, Juan Antonio Marchal, and Roberto Madeddu

vii
viii Contents

12 Detection of Cancer Stem Cells in Normal and Dysplastic/Leukemic


Human Blood . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 163
Alessia De Stefano, Alessandra Cappellini, Irene Casalin, Stefania Paolini,
Sarah Parisi, Maria Vittoria Marvi, Antonietta Fazio, Irene Neri,
Foteini-Dionysia Koufi, Stefano Ratti, Carlo Finelli, Antonio Curti,
Lucia Manzoli, Lucio Cocco, and Matilde Y. Follo
13 Methods to Study the Role of Mechanical Signals in the Induction
of Cancer Stem Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 177
Alessandro Gandin, Paolo Contessotto, and Tito Panciera
14 Co-Delivery Polymeric Poly(Lactic-Co-Glycolic Acid) (PLGA)
Nanoparticles to Target Cancer Stem-Like Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . 191
Catherine S. Snyder, Taylor Repetto, Kathleen M. Burkhard,
Anish Tuteja, and Geeta Mehta
15 Isolating Circulating Cancer Stem Cells (CCSCs) from Human
Whole Blood . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 205
Carla Kantara and Pomila Singh
16 Generation of Cancer Stem Cells by Co-Culture Methods . . . . . . . . . . . . . . . . . . . 219
Biswajit Das and Chanakya Nath Kundu
17 Deep Learning of Cancer Stem Cell Morphology . . . . . . . . . . . . . . . . . . . . . . . . . . . 231
Hiroyuki Kameda, Hiroaki Ishihata, and Tomoyasu Sugiyama

Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 257
Contributors

WASSIM ABOU-KHEIR • Department of Anatomy, Cell Biology, and Physiological Sciences,


Faculty of Medicine, American University of Beirut, Beirut, Lebanon
ALISHA ANAND • Department of Biochemistry and Biomedical Sciences, McMaster University,
Hamilton, ON, Canada
HISHAM BAHMAD • Arkadi M. Rywlin M.D. Department of Pathology and Laboratory
Medicine, Mount Sinai Medical Center, Miami Beach, FL, USA
KATHLEEN M. BURKHARD • Department of Biomedical Engineering, University of Michigan,
Ann Arbor, MI, USA
MANUEL CABEZA-SEGURA • Department of Medical Oncology, Hospital Clı́nico Universitario
de Valencia, INCLIVA, Biomedical Research Institute, University of Valencia, Valencia,
Spain
ALESSANDRA CAPPELLINI • Cellular Signalling Laboratory, Department of Biomedical and
Neuromotor Sciences, University of Bologna, Bologna, Italy
IRENE CASALIN • Cellular Signalling Laboratory, Department of Biomedical and
Neuromotor Sciences, University of Bologna, Bologna, Italy
JOSEFA CASTILLO • Department of Medical Oncology, Hospital Clı́nico Universitario de
Valencia, INCLIVA, Biomedical Research Institute, University of Valencia, Valencia,
Spain; Centro de Investigacion Biomédica en Red (CIBERONC), Instituto de Salud
Carlos III, Madrid, Spain; Department of Biochemistry and Molecular Biology,
Universitat de Valencia, Burjassot, Spain
ANDRÉS CERVANTES • Department of Medical Oncology, Hospital Clı́nico Universitario de
Valencia, INCLIVA, Biomedical Research Institute, University of Valencia, Valencia,
Spain; Centro de Investigacion Biomédica en Red (CIBERONC), Instituto de Salud
Carlos III, Madrid, Spain
LUCIO COCCO • Cellular Signalling Laboratory, Department of Biomedical and
Neuromotor Sciences, University of Bologna, Bologna, Italy
PAOLO CONTESSOTTO • Department of Molecular Medicine, University of Padua, Padua,
Italy
ANTONIO CURTI • IRCCS – Azienda Ospedaliero-Universitaria di Bologna, Istituto di
Ematologia “Sera ` gnoli”, Bologna, Italy
STEFAN CUSTERS • Department of Biochemistry and Biomedical Sciences, McMaster
University, Hamilton, ON, Canada
BISWAJIT DAS • Cancer Biology Division, School of Biotechnology, Kalinga Institute of
Industrial Technology (KIIT)-Deemed to be University, Bhubaneswar, Odisha, India
JULIA LÓPEZ DE ANDRÉS • Biopathology and Regenerative Medicine Institute (IBIMER),
Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain;
Instituto de Investigacion Biosanitaria ibs.GRANADA, University Hospitals of Granada-
University of Granada, Granada, Spain; Excellence Research Unit ‘Modeling Nature’
(MNat), University of Granada, Granada, Spain; Department of Human Anatomy and
Embryology, Faculty of Medicine, University of Granada, Granada, Spain; BioFab i3D
Lab- Biofabrication and 3D (bio)printing Singular Laboratory, University of Granada,
Granada, Spain

ix
x Contributors

LAURA DE LARA-PEÑA • Biopathology and Regenerative Medicine Institute (IBIMER),


Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain;
Instituto de Investigacion Biosanitaria ibs.GRANADA, University Hospitals of Granada-
University of Granada, Granada, Spain; Excellence Research Unit ‘Modeling Nature’
(MNat), University of Granada, Granada, Spain; Department of Human Anatomy and
Embryology, Faculty of Medicine, University of Granada, Granada, Spain; BioFab i3D
Lab- Biofabrication and 3D (bio)printing Singular Laboratory, University of Granada,
Granada, Spain
ALESSIA DE STEFANO • Cellular Signalling Laboratory, Department of Biomedical and
Neuromotor Sciences, University of Bologna, Bologna, Italy
VITALE DEL VECCHIO • Department of Experimental Medicine, Histology and Embryology
Section, University of Campania “L. Vanvitelli”, Naples, Italy
FEDERICA ETZI • Department of Biomedical Sciences-Histology, University of Sassari, Sassari,
Italy; I.N.B.B. National Institute of Biostructure and Biosystem, Rome, Italy
CRISTIANO FARACE • Department of Biomedical Sciences-Histology, University of Sassari,
Sassari, Italy; I.N.B.B. National Institute of Biostructure and Biosystem, Rome, Italy
ANTONIETTA FAZIO • Cellular Signalling Laboratory, Department of Biomedical and
Neuromotor Sciences, University of Bologna, Bologna, Italy
GRAZIA FENU • Department of Biomedical Sciences-Histology, University of Sassari, Sassari,
Italy
CARLO FINELLI • IRCCS – Azienda Ospedaliero-Universitaria di Bologna, Istituto di
Ematologia “Sera ` gnoli”, Bologna, Italy
MATILDE Y. FOLLO • Cellular Signalling Laboratory, Department of Biomedical and
Neuromotor Sciences, University of Bologna, Bologna, Italy
RENATO FRANCO • Pathology Unit, Dipartimento di Salute Mentale, Fisica e Medicina
Preventiva, Università degli Studi della Campania ‘Luigi Vanvitelli’, Naples, Italy
ALESSANDRO GANDIN • Department of Industrial Engineering (DII), University of Padua,
Padua, Italy
BLANCA GARCIA-MICÓ • Department of Medical Oncology, Hospital Clı́nico Universitario de
Valencia, INCLIVA, Biomedical Research Institute, University of Valencia, Valencia,
Spain
FATIMA GHAMLOUCHE • Department of Anatomy, Cell Biology, and Physiological Sciences,
Faculty of Medicine, American University of Beirut, Beirut, Lebanon
VINOD GOPALAN • Cancer Molecular Pathology, School of Medicine and Dentistry, Griffith
University, Gold Coast, QLD, Australia
CARMEN GRIÑÁN-LISÓN • Biopathology and Regenerative Medicine Institute (IBIMER),
Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain;
Instituto de Investigacion Biosanitaria ibs.GRANADA, University Hospitals of Granada-
University of Granada, Granada, Spain; Excellence Research Unit ‘Modeling Nature’
(MNat), University of Granada, Granada, Spain; BioFab i3D Lab- Biofabrication and
3D (bio)printing Singular Laboratory, University of Granada, Granada, Spain;
Department of Biochemistry and Molecular Biology 2, School of Pharmacy, University of
Granada, Granada, Spain; GENYO, Centre for Genomics and Oncological Research,
Pfizer/University of Granada/Andalusian Regional Government, Granada, Spain
SANA HACHEM • Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of
Medicine, American University of Beirut, Beirut, Lebanon
HIROAKI ISHIHATA • School of Computer Science, Tokyo University of Technology, Tokyo, Japan
Contributors xi

FARHADUL ISLAM • Department of Biochemistry and Molecular Biology, University of


Rajshahi, Rajshahi, Bangladesh
HIROYUKI KAMEDA • School of Bioscience and Biotechnology, Tokyo University of Technology,
Tokyo, Japan; School of Computer Science, Tokyo University of Technology, Tokyo, Japan
CARLA KANTARA • Departments of Neuroscience and Cell Biology and Biochemistry and
Molecular Biology, The University of Texas Medical Branch, Galveston, TX, USA
FOTEINI-DIONYSIA KOUFI • Cellular Signalling Laboratory, Department of Biomedical and
Neuromotor Sciences, University of Bologna, Bologna, Italy
CHANAKYA NATH KUNDU • Cancer Biology Division, School of Biotechnology, Kalinga
Institute of Industrial Technology (KIIT)-Deemed to be University, Bhubaneswar, Odisha,
India
MARCELLA LA NOCE • Department of Experimental Medicine, Histology and Embryology
Section, University of Campania “L. Vanvitelli”, Naples, Italy
ALFRED KING-YIN LAM • Cancer Molecular Pathology, School of Medicine and Dentistry,
Griffith University, Gold Coast, QLD, Australia
ROBERTO MADEDDU • Department of Biomedical Sciences-Histology, University of Sassari,
Sassari, Italy; I.N.B.B. National Institute of Biostructure and Biosystem, Rome, Italy
LUCIA MANZOLI • Cellular Signalling Laboratory, Department of Biomedical and
Neuromotor Sciences, University of Bologna, Bologna, Italy
JUAN ANTONIO MARCHAL • Biopathology and Regenerative Medicine Institute (IBIMER),
Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain;
Instituto de Investigacion Biosanitaria ibs.GRANADA, University Hospitals of Granada-
University of Granada, Granada, Spain; Excellence Research Unit ‘Modeling Nature’
(MNat), University of Granada, Granada, Spain; Department of Human Anatomy and
Embryology, Faculty of Medicine, University of Granada, Granada, Spain; BioFab i3D
Lab- Biofabrication and 3D (bio)printing Singular Laboratory, University of Granada,
Granada, Spain
MARIA VITTORIA MARVI • Cellular Signalling Laboratory, Department of Biomedical and
Neuromotor Sciences, University of Bologna, Bologna, Italy
GEETA MEHTA • Department of Biomedical Engineering, University of Michigan, Ann
Arbor, MI, USA; Department of Materials Science and Engineering, University of
Michigan, Ann Arbor, MI, USA; Macromolecular Science and Engineering, University of
Michigan, Ann Arbor, MI, USA; Rogel Cancer Center, University of Michigan, Ann
Arbor, MI, USA; Precision Health, University of Michigan, Ann Arbor, MI, USA
GEETA MEHTA • Department of Materials Science and Engineering, University of Michigan,
Ann Arbor, MI, USA; Department of Biomedical Engineering, University of Michigan,
Ann Arbor, MI, USA; Macromolecular Sciences and Engineering, University of Michigan,
Ann Arbor, MI, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI,
USA; Precision Health, University of Michigan, Ann Arbor, MI, USA
PETAR MILETIC • Department of Surgery, McMaster University, Hamilton, ON, Canada
IRENE NERI • Cellular Signalling Laboratory, Department of Biomedical and Neuromotor
Sciences, University of Bologna, Bologna, Italy
TITO PANCIERA • Department of Molecular Medicine, University of Padua, Padua, Italy
STEFANIA PAOLINI • IRCCS – Azienda Ospedaliero-Universitaria di Bologna, Istituto di
Ematologia “Sera ` gnoli”, Bologna, Italy
SARAH PARISI • IRCCS – Azienda Ospedaliero-Universitaria di Bologna, Istituto di
Ematologia “Sera ` gnoli”, Bologna, Italy
xii Contributors

ANDREA PISANO • Department of Biomedical Sciences-Histology, University of Sassari,


Sassari, Italy
TASFIK UL HAQUE PRONOY • Department of Biochemistry and Molecular Biology, University
of Rajshahi, Rajshahi, Bangladesh
STEFANO RATTI • Cellular Signalling Laboratory, Department of Biomedical and
Neuromotor Sciences, University of Bologna, Bologna, Italy
TAYLOR REPETTO • Department of Materials Science and Engineering, University of
Michigan, Ann Arbor, MI, USA
LORNELLA SEENEEVASSEN • INSERM U1312 BRIC Bordeaux Institute of Oncology,
University of Bordeaux, Bordeaux, France
MUHAMMAD VASEEM SHAIKH • Department of Surgery, McMaster University, Hamilton,
ON, Canada
POMILA SINGH • Departments of Neuroscience and Cell Biology and Biochemistry and
Molecular Biology, The University of Texas Medical Branch, Galveston, TX, USA
SHEILA K. SINGH • Department of Surgery, McMaster University, Hamilton, ON, Canada;
Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton,
ON, Canada
CATHERINE S. SNYDER • Department of Materials Science and Engineering, University of
Michigan, Ann Arbor, MI, USA
TOMOYASU SUGIYAMA • School of Bioscience and Biotechnology, Tokyo University of Technology,
Tokyo, Japan
VIRGINIA TIRINO • Department of Experimental Medicine, Histology and Embryology
Section, University of Campania “L. Vanvitelli”, Naples, Italy
ANISH TUTEJA • Department of Materials Science and Engineering, University of Michigan,
Ann Arbor, MI, USA; Macromolecular Sciences and Engineering, University of Michigan,
Ann Arbor, MI, USA; Department of Chemical Engineering, University of Michigan, Ann
Arbor, MI, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
CHRISTINE VARON • INSERM U1312 BRIC Bordeaux Institute of Oncology, University of
Bordeaux, Bordeaux, France
CHITRA VENUGOPAL • Department of Surgery, McMaster University, Hamilton, ON,
Canada
AMANI YEHYA • Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of
Medicine, American University of Beirut, Beirut, Lebanon
ANISSA ZAAFOUR • INSERM U1312 BRIC Bordeaux Institute of Oncology, University of
Bordeaux, Bordeaux, France
GIUSEPPA ZANNINI • Pathology Unit, Dipartimento di Salute Mentale, Fisica e Medicina
Preventiva, Università degli Studi della Campania ‘Luigi Vanvitelli’, Naples, Italy
FEDERICA ZITO MARINO • Pathology Unit, Dipartimento di Salute Mentale, Fisica
e Medicina Preventiva, Universita` degli Studi della Campania ‘Luigi Vanvitelli’, Naples,
Italy
Chapter 1

Cancer Stem Cells: Current Challenges and Future


Perspectives
Muhammad Vaseem Shaikh, Stefan Custers, Alisha Anand, Petar Miletic,
Chitra Venugopal, and Sheila K. Singh

Abstract
Despite major advances in health care including improved diagnostic tools, robust chemotherapeutic
regimens, advent of precision, adjuvant and multimodal therapies, there is a major proportion of patients
that still go on to experience tumor progression and recurrence. Cancer stem cells (CSCs) are shown to be
responsible for tumor persistence and relapse. This subpopulation of cancer cells possess normal stem
cell like traits of self-renewal, proliferation, and multilineage differentiation. Currently, they are isolated and
enriched based on the cell surface markers that can be detected and sorted through fluorescence and
magnetic-based cell sorting. In this chapter, we review the current challenges and limitations often
encountered in CSC research, including the identification of universal markers, therapy resistance, and
new drug development. Current and future perspectives are discussed to address these challenges including
utilization of cutting-edge technologies such as next-generation sequencing to elucidate the genome,
epigenome, and transcriptome on a single-cell level and genome-wide CRISPR-Cas9 screens to identify
novel pathway-based targeted therapies. Further, we discuss the future of precision medicine and the need
for the improvement of clinical trial designs.

Key words Cancer stem cells, Tumor initiating cells, Standard of care, Intratumoral heterogeneity,
Cell surface makers, Fluorescent activated cell sorting, CRISPR-Cas9 system, Single cell RNA
sequencing, Minimal residual disease.

1 Introduction

Cancer is the second leading cause of death worldwide, accounting


for nearly 10 million deaths in 2020 [1]. Although there have been
significant improvements in the prognosis of patients due to
advancements in diagnostic tools, intensive chemotherapeutic
regimens, and the advent of precision and multimodal therapies
[2–4], most patients are faced with relapse and disease

Muhammad Vaseem Shaikh and Stefan Custers contributed equally with all other contributors.

Federica Papaccio and Gianpaolo Papaccio (eds.), Cancer Stem Cells: Methods and Protocols, Methods in Molecular Biology,
vol. 2777, https://doi.org/10.1007/978-1-0716-3730-2_1,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2024

1
2 Muhammad Vaseem Shaikh et al.

progression and ultimately death. Moreover, there have been little


to no improvements in prognosis for some advanced and aggressive
cancers [5].
This therapeutic failure can be partly attributed to extensive
intratumoral heterogeneity (ITH) present at the genetic, transcrip-
tomic, and functional levels [6, 7]. Several studies have shown
correlation between tumor heterogeneity and poorer clinical out-
comes [8–10]. ITH was traditionally explained by the clonal evolu-
tion model, wherein carcinogenesis was believed to be a result of
sequential accumulation of mutations and the subsequent gradual
selection of clones under various attractor states [11]. Recently,
several studies indicate heterogeneity to be driven by distinct sub-
populations of cells commonly referred as cancer stem cells (CSCs)
that possess traits of normal stem cells, such as multilineage differ-
entiation, self-renewal, and therapy evasion [12]. CSCs are believed
to adapt and survive the intensive standard of care (SoC) regimen,
become chemoradiotherapy-resistant, and seed the formation of
therapy-resistant fulminant relapse [13]. CSCs are also referred to
as tumor-initiating cells (TICs), as they are believed to initiate
tumorigenesis in a hierarchical manner, with CSCs at the apex and
rapidly proliferating cells forming the bulk of the tumor [14].
In 1997, Bonnet and Dick were the first to isolate and charac-
terize cancer stem cell subpopulations from acute myeloid leukemia
(AML) patients [15]. They demonstrated that a finite number of
cells termed leukemic stem cells (LSCs), characterized by the pres-
ence of cell surface phenotype CD34+CD38-, when injected into
immunodeficient mice were able to proliferate and differentiate,
giving rise to disease similar to the patient donor. They were able to
demonstrate the self-renewal capacity of CSCs by serial transplan-
tation of cells into secondary recipient mice, resulting in
re-establishment of AML. Moreover, they demonstrated an
enhanced tumorigenic ability of these cells by limiting dilution
assay. Later, Al-Hajj et al. demonstrated the presence of CSCs in
solid tumors, when they demonstrated CD44+/CD24- cells in
breast cancer tumors had enhanced tumorigenic capacity
[16]. CSCs have since then been characterized in several cancers,
viz., brain, lung, liver, pancreatic, and melanoma [17–21].
Understanding the biology of CSCs is instrumental for the
development of novel therapeutics that can target CSCs. In doing
so we recognize the challenges that accompany CSC research which
include the origins of CSCs, inconsistent markers, drug resistance,
and drug development. To circumvent these barriers, novel tools
and techniques such as single-cell RNA sequencing, genome-wide
CRISPR-Cas9 screens, and modifications in precision medicine and
clinical trial designs are being incorporated into CSC research.
Cancer Stem Cells: Current Challenges and Future Perspectives 3

2 Challenges in CSC Research

The concept of the CSC has progressed from being a highly specu-
lative and intensively debated concept to a fairly defined and better
understood biological entity in many tumor types, including AML,
breast, brain, colon, lung, and other cancers. Despite the advances
made in understanding CSCs, many challenges lie ahead and will be
discussed below.

2.1 The CSC CSCs by definition are a subset of tumor cells that contain both
Definition Is a self-renewal and multilineage differentiation capacities that fuel
Functional Trait Rather tumor initiation, treatment resistance, and recurrence [22]. This
than a Specific Cell definition is based on the shared functional tumor-initiation trait
among CSCs. Considering that CSC phenotypes are complex, vary
between tumors, and are affected by abnormalities arising from
neoplastic transformation, defining CSCs outside the scope of the
shared functional trait of tumor initiation can be difficult
[23]. Importantly, this functional CSC definition can make data
interpretation complex as the gold standard assay for CSC identifi-
cation is in vivo serial transplantation, a functional read-out which
can underestimate total CSC numbers and has a bias towards more
malignant CSC types. Moreover, serial implantation risks isolating
artifacts and may limit true heterogeneity of the original tumor.
Due to this limiting effect a more refined definition has been
proposed: CSCs are tumor cells that can be proven to generate
long-term progeny, either by direct engraftment or genetic
tracing [22].

2.2 Molecular Historically, CSCs have been isolated by Hoechst staining, utilizing
Markers Used to the ATP-binding cassette (ABC) transporter protein family, which
Identify the CSC Do Not was later replaced and standardized by fluorescence-activated cell
Specifically Nor sorting (FACS) based on cell surface markers. Although marker-
Uniformly Mark CSCs based CSC isolation is easier to perform, the markers used are not
always reliable, leading to challenges in CSC identification and data
interpretation [22].
First, the markers currently used do not specifically identify
CSCs, as these markers play central roles in normal stem cell biol-
ogy and are also expressed in other normal cell types. For some
markers, CSC specificity can be achieved by targeting splice var-
iants. For example, full-length CD144 is widely expressed, while
the CD44v6 splice variant is more indicative of CSCs [24–26]. The
use of marker combinations has alleviated this problem to some
extent; however, the lack of specificity remains a hurdle for reliable
CSC detection [22, 23]. On top of this, markers may have roles in
other mechanisms, which may be favorably selected for during
in vivo serial transplantation. For example, CD44, CD90, and
CD34 are involved in cell adhesion and attachment and are thus
4 Muhammad Vaseem Shaikh et al.

favorably selected for during in vivo engraftment. Together these


observations indicate that markers that support the CSC phenotype
need to be identified [27].
Second, CSC markers are not universal for CSC identification
due to inter- and intratumoral heterogeneity, as CSC biology dif-
fers substantially both within tumors and among cancer types
[22, 23]. Proper use of existing markers is highly tumor dependent;
for example, ALDH1 seems to be a reliable CSC marker in certain
cancers such as breast cancer [28], but its validity is debated in
other cancer types such as liver and pancreatic cancer [29]. How-
ever, there is evidence indicating that deletion of ALDH1 enhances
stemness in breast cancer, making its validity as a CSC marker
highly contested only in certain contexts [30]. The identification
of consensus markers, capable of uniformly detecting CSCs across
and within tumors, is warranted to improve CSC research.
Third, CSC markers are not universally applicable, as antibodies
targeting CSC markers may not be suitable for every research
method [22]. This is the case for CD133, which can be used to
sort CSCs by FACS, but its suitability for mRNA- or immunohis-
tochemistry (IHC)-based applications is limited [31, 32].
Fourth, current CSC markers may not be constitutively
expressed. CD133 is an example of a marker that is downregulated
during the G0/G1 phase of the cell cycle [33]. Additionally, inter-
and intratumor heterogeneity may make markers unreliable as they
might be downregulated, potentially leaving certain subclones
undetectable [22], as is the case for CD133 inactivation by CpG
methylation in colorectal cancer and glioblastoma (GBM) [34].
In conclusion, consensus markers are needed that are function-
ally linked to stemness mechanisms and assure reliable and uniform
expression on CSCs. Research into enzymatic and signaling
mechanisms is a starting point on the functional CSC road of
discovery. Additional work should be performed to identify cell
surface markers that are selectively induced by signaling pathways
involved in normal stem cell biology (Wnt, TGF-b, Hedgehog).
Stringent validation in human primary tumor samples is warranted
for these potential CSC markers. It is expected that these markers
will be different between tumor types and potentially between
clones [22].

2.3 The Origins of One of the most debated topics of the CSC is its cell of origin.
CSCs Tumors are the result of genetic and epigenetic alterations accu-
mulated over many years. The presence of stem cell markers seems
indicative of a stem cell origin. Another possibility is that CSCs arise
from transit-amplifying/progenitor cells that increase mutational
burden during multistep tumorigenesis and introduce their
acquired mutations into corresponding stem cell pools via a process
of spontaneous dedifferentiation [23, 35, 36].
Cancer Stem Cells: Current Challenges and Future Perspectives 5

Realizing these two hypotheses may not exclude one another,


the rigid hierarchical CSC model has progressed into the more
flexible model of phenotypic reversion. Phenotypic reversion indi-
cates that there is a balance between CSCs and differentiated cell
states which can interconvert and hereby link stochastic and hierar-
chical properties [22].
Stemness is acquired via CSC intrinsic changes in gene expres-
sion programs and underlying regulatory mechanisms of self-
renewal and differentiation. On top of these intrinsic mechanisms,
studies propose a prominent role of the cancer microenvironment,
or CSC niche, generating feed-forward and feed-backward
mechanisms. For example, gliomas can be initiated from terminally
differentiated neurons by reverting back into the CSC state, a
mechanism stimulated by Notch secreting endothelial cells
[37, 38]. Additionally, studies suggest that epithelial-to-mesenchy-
mal transitioning (EMT) signaling can induce CSCs from differ-
entiated tumor cells [39]. Not all CSCs express EMT markers [40]
and EMT signaling factors in some cases can reduce CSC
qualities [41].
Molecular understanding of how CSC-intrinsic and environ-
mental signaling dictate CSC hierarchies and how they govern
effective interconversion between tumor states remains limited.
In summary, current models capable of capturing microenvi-
ronmental interactions with CSCs remain suboptimal. Molecular
understanding of these mechanisms will provide further insights in
self-renewal and differentiation cues and thus tumor cell hierarchy
dependencies on the CSC niche.
Single cells would need to be followed in vitro and in vivo using
genetic tracing and live imaging. Lineage tracing animal models are
needed to determine whether CSC reversion occurs in vivo and to
assess its frequency of occurrence. Moreover, this would help
understand to what extent differentiation becomes irreversible.

2.4 Drug Resistance A major hurdle in cancer treatment is CSC-mediated treatment


resistance to SoC and initiation of relapse. These cells are causa-
tively correlated with poor prognosis, treatment resistance, and
subsequent relapse. Unraveling these mechanisms and effective
targeting of CSCs will be a decisive victory in the treatment of
cancer as a whole [42].
CSCs are mostly quiescent and abundantly express ABC2 trans-
porter proteins and anti-apoptotic proteins. Together these proper-
ties aid in evading apoptosis and drug-induced cytotoxicity when
compared to differentiated tumor cells [23, 43–45]. As described
above, research has indicated the involvement of (partial) EMT
programs in the initiation and sustainment of CSCs, and secreted
EMT signaling molecules from stromal cells have been shown to
induce chemo- and radiotherapy resistance [46–48]. Further
insights into treatment resistance are warranted for more efficient
therapeutic targeting of CSCs.
6 Muhammad Vaseem Shaikh et al.

2.5 Drug Another challenge is the development of drugs targeting CSCs.


Development Currently, there are two approaches considered: the direct target-
ing of CSCs and the targeting of the CSC niche.
Targeting CSCs is dependent on its genetics and environment,
resulting in context-dependent signaling pathways and unique
challenges per tumor type. Drug screens can be technically difficult
due to the rarity of CSCs in most tumors and limited means of
propagating them in vitro. Means to circumvent these issues
include the use of enrichment media used for brain cancers such
as medulloblastoma and GBM [17, 49]. Another method that has
been employed is genetically enforcing an EMT-like state in epithe-
lial tumors [23].
Heterotypical signals arising from tumor-associated stroma are
often responsible for activating EMT. Targeting paracrine signaling
pathways that activate and sustain CSC programs should provide
insights for targeting CSCs. There have been successes with anti-
bodies targeting tropical signals. For example, macrophage M-CSF
receptor antibodies have been developed and effectively reduced
macrophage homing tumors in syngeneic mouse models [50]. In
addition, stimulating paracrine signaling molecules favoring
non-CSCs could be used to push the CSC/non-CSC balance
towards the non-CSC side. Derivates of molecules such as secreted
frizzled-related protein 1 (SFRP) and Dickkopf WNT signaling
pathway inhibitor 1 (DKK) could be developed and employed to
induce SoC susceptibility [23, 40].
CSCs exist in a niche formed by multiple cell types and distinct
signaling molecules (growth factors, cytokines, extracellular matrix
components). Studying biological properties of CSCs in isolation
or performing screens in the absence of a surrounding relevant
niche is unlikely to yield translatable results in vivo. Currently,
in vivo models depend on immunodeficient mice or syngeneic
mouse models. Immunodeficient models lack a full microenviron-
ment as the adaptive immune system is lacking and syngeneic
settings are not fully translational as they are artificially induced
by knocking out one or two genes. Neither model recapitulates the
clinical setting to a full extent and are therefore limited in translat-
ing therapies to the clinic.

3 Future Perspectives

To overcome the challenges encountered in CSC research, an


enormous amount of work is being done to better characterize
the CSCs, understand resistance mechanisms, develop precision
medicines and multimodal therapy regimens, and enhance clinical
trial design (Fig. 1).
Cancer Stem Cells: Current Challenges and Future Perspectives 7

Fig. 1 Schematic representation of target discovery for the development of CSC-specific therapies

3.1 Single-Cell RNA Indeed, next-generation sequencing (NGS) technologies have


Sequencing helped us to sequence large amounts of DNA and RNA and have
led to a better understanding of cancer biology and intertumoral
heterogeneity [51]. However, cancer is a heterogenous ecosystem
comprising bulk cancer cells, tumor immune microenviron-
ment (TIME), and CSCs [52], and bulk sequencing obscures the
accurate understanding of all the different cellular subpopulations
[53]. Rapidly evolving methods of single-cell RNA sequencing
(scRNA seq), which can profile genome, epigenome, and transcrip-
tome of an individual cell, can help us better elucidate the drivers of
tumor progression, metastasis, and therapy resistance [54, 55].
scRNA seq technology not only illuminates intratumoral het-
erogeneity, but also highlights the plasticity and dynamic nature of
8 Muhammad Vaseem Shaikh et al.

various cellular states. Neftel et al. profiled glioma stem cells


(GSCs) using scRNA seq and classified GBM into four major sub-
types: oligodendrocyte progenitor cells (OPC-like), astrocytes
(AC-like), neural progenitor cells (NPC-like), and mesenchymal
(MES-like) [7]. They discovered that a single GBM tumor com-
prises multiple cellular states in stark contrast to a singular cellular
state as initially believed [7]. Through lineage tracing experiments,
they further discovered that when cells of a particular type are
engrafted in mice, they not only form the tumor of that specific
state but re-establish the heterogeneity seen in the primary tumor
through lineage tracing experiments [7]. Additionally, when they
investigated the commonly known GSC marker and transcription
factors, they found each of the four states was enriched for specific
markers; for example, CD133 was enriched in OPC-like and CD44
was enriched in MES-like state [7].
Additionally, scRNA seq has aided in profiling circulating
tumor cells, identifying prognostic markers, understanding the
TIME, and studying tumor resistance mechanisms, thereby
providing a new perspective in CSC research. Aceto et al. [56]
identified oligoclonal circulating tumor clusters to have increased
propensity for breast-to-lung metastasis, and abundance of circu-
lating tumor cells (CTC) correlated with poorer outcomes. scRNA
seq helped identify the cell junction component plakoglobin as
differentially expressed, and knockdown of plakoglobin led to sig-
nificant reduction in metastasis. Lei et al. [57] employed scRNA seq
to profile breast cancer stem cells (BCSCs) isolated from two
HER2+ breast cancer patients and compared the transcriptomic
profile with breast cancer cells, mammary cells, and CD44+ mam-
mary cells. They were able to identify 404 genes differentially
expressed in BCSCs, and additionally, they identified CA12 as a
novel prognostic marker in HER2+ breast cancer patients. Chung
et al. [58] performed scRNA seq in 11 patients with primary breast
cancer. At a single-cell resolution, the immune population was
characterized in three distinct clusters of T lymphocytes, B lym-
phocytes, and macrophages. T lymphocyte and macrophages dis-
played T cell regulatory and M2 phenotype, respectively, which
promotes an immunosuppressive and pro-tumorigenic environ-
ment. To understand resistance, Zhang et al. carried out scRNA
seq in samples before and after treatment in high-grade serous
ovarian cancer. They identified a stress-associated genetic signature
present before treatment which was subsequently enriched post-
treatment and is correlated with poor patient prognosis. In addition
to stress-associated genetic signature genes associated with stem-
ness, pro-survival and pro-inflammation were also enriched, further
protecting the cells from chemotherapy [59].
Research over the past decade has concentrated on characteriz-
ing CSCs as their elimination might translate to clinical benefit.
scRNA seq has helped to deconvolute the CSC theory, ITH, drug
Cancer Stem Cells: Current Challenges and Future Perspectives 9

resistance mechanisms, and identify prognostic markers. The vast


amount of data generated can be used to identify vulnerabilities of
CSCs and develop targeted therapies or multimodal therapies to
synchronously attack the susceptible targets.

3.2 Genome-Wide Comprehensive genetic studies in the past two decades have
CRISPR Screens emphasized and helped us understand correlations between the
disease phenotype and the mutations seen in cancers; however,
gene editing technologies that could allow us to study the driving
forces of cancer and correct mutations in the genome were needed
to drive further breakthroughs in precision medicine. Discovery of
zinc finger nucleases (ZFNs) [60], TALE domains in transcription
activator-like effector nucleases (TALENs) [61], and CRISPR/Cas
(clustered regularly interspaced palindromic repeats/CRISPR-
associated) technology have allowed for the precise editing of the
eukaryotic genome. The CRISPR/Cas system has revolutionized
the gene editing technology due to its ability to precisely manipu-
late genes even if they are silenced or in a dense location and have
allowed for ease of design, affordability, and scalability compared to
other techniques [62, 63]. With the advent of genome-wide loss-
of-function (LOF) and gain-of-function (GOF) single-guide RNA
(sgRNA) libraries, it is now possible to screen and discover new
therapeutic vulnerabilities in CSCs.
LOF screens are carried out by using genome-wide knockdown
libraries, in the presence or absence of therapies in order to identify
the fitness genes and biological processes that make the cells resis-
tant. Shalem et al. [64] constructed a genome-wide CRISPR/Cas9
knockout library (GecKO) and employed it to mice for genes that
confer resistance to a human melanoma cell line when treated with
vemurafenib. Hart et al. [65] developed an improved CRISPR loss-
of-function library known as the Toronto KnockOut (TKO) library
targeting over 18,000 genes, identifying ~2000 fitness genes which
is 4–5 times more than RNAi screens. Chen et al. [66] employed a
genome-wide CRISPR LOF screen (mGeCKOa) comprising
67,405 sgRNAs targeting 20,611 protein-coding genes and 1175
microRNA precursors to study the evolution of cancer and metas-
tasis in a mouse model.
LOF screens have been routinely used to discover novel and
adjuvant therapeutic targets as well as studying the TIME. CRISPR
LOF screen conducted on CSCs from patient-derived colon cancer
lines identified 44 essential genes including genes involved in the
cholesterol biosynthesis pathway. The authors validated these find-
ings by demonstrating enhanced antitumor efficacy in vivo when
treated with combinatorial treatment with cholesterol biosynthesis
inhibitors and 5-FU [67]. Wei et al. [68] discovered loss of
Regnase-1 led to increased fitness in CD8 + Tcells, prolonging
their persistence and their ability to infiltrate and fight cancer
cells. Recently, CRISPR Cas9 screens carried out on GSCs
10 Muhammad Vaseem Shaikh et al.

identified SOX transcription family, SOCS3, USP8, and DOT1L,


and protein UFMylation as essential genes responsible for temozo-
lomide resistance [69]. Xu et al. [70] identified that NMDAR as an
adjuvant therapeutic target with tyrosine kinase inhibitors (TKIs) in
hepatocellular carcinoma (HCC), co-treatment with an NMDAR
antagonist, ifenprodil, with TKI, sorafenib, suppressed genes asso-
ciated with the WNT signaling pathway and stemness and reduced
self-renewal ability and proliferation of HCC cells.
In contrast CRISPR activation (CRISPRa) screens are a potent
tool for testing gain of function of endogenous genes by transcrip-
tional upregulation of target genes through the selective activation
using transcriptional activators targeting the promoter sites of tar-
get genes guided by sgRNA and a dead CAS9 system
[71, 72]. CRISPRa screens have tremendous potential in the eluci-
dation of drug resistance mechanisms, which are normally believed
to be gain-of-function events in response to therapy. Recently, a
reciprocal CRISPRa and CRIPR LOF screen was carried out on
GSCs and chimeric antigen receptor-T (CAR-T) revealing RELA
and NPLOC4 to be essential in GSCs for resistance against immu-
notherapy and also elucidated that the knockout of TLE4 and
IKZF2 led to superior effector function and reduced the exhaus-
tion of CAR-Ts [73]. Moreover, CRISPRa screens carried out on
pancreatic ductal adenocarcinoma lines identified a cellular resis-
tance mechanism to gemcitabine, 5-fluorouracil, irinotecan, and
oxaliplatin, revealing overexpression of ABCG2, a drug efflux
pump, as a common drug resistance mechanism to all the four
drugs [74].
Taken together these studies suggest the immense promise for
CRISPR technology in the development of new precision oncoly-
tics by identifying novel targets and facilitating existing therapies
through the elucidation of resistance mechanisms.

3.3 Precision Pathway-based medicines have been developed using our deeper
Medicine understanding of cancer biology from the genomic, proteomic, and
functional data generated from CRISPR screens and single-cell
sequencing. These therapies could not only target aberrant cellular
populations but also help to spare healthy cells. Imatinib, which
targets gain-of-function mutations in the genes encoding platelet-
derived growth factor receptor-α (PDGFRα) in gastrointestinal
stromal tumors, was the first clinical success story for targeted
cancer treatment [75].
Since then, several targeted therapies have been designed
against CSCs owing to the increased clinical significance of target-
ing these functionally important cells. Hedgehog signaling pathway
expression is correlated with chemoresistance, metastatic spread,
and CSC maintenance. Hh regulates the target gene expression
through the transmembrane protein Smoothened (Smo)
[76]. Glasdegib, vismodegib, and sonidegib showed significant
Cancer Stem Cells: Current Challenges and Future Perspectives 11

activity in locally advanced and metastatic basal cell carcinoma and


AML and were approved by the US Food and Drug Administration
(FDA) [77, 78]. MK-0752 a γ-secretase inhibitor that targets the
Notch signaling pathway, which is implicated for maintenance of
CSCs [79], showed clinical benefit in refractory central nervous
tumors in pediatric patients [80]. Gao et al. used CRISPR LOF
screens on CSCs that identified 44 genes in the cholesterol biosyn-
thesis pathway to be essential for stemness and chemoresistance in
colon cancer. They also further demonstrated that lovastatin and
5-flurouracil combination treatment showed significant preclinical
efficacy [67].
However, clinical approval of new drugs is a lengthy and an
expensive process, on average taking 13 years of research and 2–-
3 billion USD for a drug to reach from bench to market
[81]. Hence, recently, drug repurposing (“teaching old dogs new
tricks”) is receiving lot of traction in the research community,
wherein the strategy is to use next-generation biological data and
leverage it for the usage of inhibitors that have already received
regulatory approval. This pathway greatly reduces development
costs and time as well as alleviates business risks as safety, toxicity,
and pharmacokinetic profiles of the investigational drug have
already been established, allowing drugs to be directly fast-tracked
to Phase II and III of the clinical trial process [82].
Machine learning and data mining are vital to tap into the
unprecedented resources of approved drugs. Connectivity
Mapping (CMap) and the Library of Integrated Network-based
Cellular Signatures (LINCS) are extensive databases of aberrant
gene expression signatures and putative drugs that could poten-
tially have an inhibitory or potentiating effect on these genetic
profiles [83, 84]. These tools have greatly facilitated and acceler-
ated the process for identification of new molecules. Using CMap,
Singh et al. identified apomorphine, a dopamine antagonist, to
prevent brain metastasis in mouse models by targeting pre-
metastasis-associated genes KIF16B, SEPW1, and TESK2 in
BTICs [85]. Recently, a clinical trial conducted in patients with
pancreatic cancer showed significant survival benefit in patients
treated with metformin, a common antidiabetic drug [86]. This
strategy has also shown clinical benefit in elderly non-fit AML
patients, and the addition of all-trans retinoic acid to decitabine
showed significant higher remission rates as compared to the single
treatment arm [87]. These studies highlight the window of oppor-
tunity that drug repurposing has opened.
Carcinogenesis is believed to be strongly related to a dysfunc-
tional immune system as well as immune surveillance [88]. Cancer’s
cellular landscape evolves with cancer progression and cancer cells
remodel the TIME to their advantage [89], while the dysregulated
immune system activates stem cell signaling pathways responsible
for therapy resistance, antigen escape, and recurrence
12 Muhammad Vaseem Shaikh et al.

[89, 90]. Based on our understanding of the TIME, over the past
decade, novel immunotherapeutic strategies such as immune
checkpoint inhibitors and CAR-T cell therapy have emerged. In a
first of its kind study, Vora et al. carried out a head-to-head com-
parison of three immunotherapeutic modalities (antibodies, bispe-
cific T cell engagers [BITEs], and CAR-T) targeting CD133 on
GSCs and found the CAR-T-based treatment modality to be most
efficacious [91]. Several immune checkpoint inhibitors including
CTLA4 (ipilimumab) [92], PD-1 (cemiplimab) [93], and PDL-1
(avelumab) [94] have been evaluated clinically in various cancers
and have shown efficacy. CD19 and CD22 CAR-T cell therapy have
shown clinical success in liquid tumors [95–97] and several trials
testing their efficacy are underway for solid tumors
(NCT02915445, NCT02465983, NCT03585764).

3.4 Clinical Trial Improved therapeutic regimens have allowed complete remission in
Design patients with most cancers and thereby improving prognosis
[98, 99]. Complete remission is defined as reduction in the tumor
burden by 99% as measured by advanced radiological techniques
(PET, CT, or MRI) [100]. However, small therapy-resistant sub-
populations of tumor cells survive aggressive treatment regimens to
drive tumor recurrence [101, 102]. These cells are traditionally
called “minimal residual disease” (MRD) [103]. Recent studies
indicate that tumor recurrence is not driven by innate mutational
mechanisms [104, 105], rather by enrichment of a small population
of cells (CSCs) that are inherently resistant to anticancer therapy
[102, 106].
There are several conceptual advantages of treating patients at
MRD rather than waiting for clinical relapse to initiate further
therapy: first, the fact that cancer which is characterized by exten-
sive ITH will be at its lowest subclonal complexity and most homo-
geneous state which would increase the likelihood of designing a
tailored drug, targeting a rare minority of CSCs enriched at MRD
[107]. Additionally, due to the decrease in tumor burden there is
also reduction in the ability of the cells to remodel the surrounding
environment, or reprogram the TIME leading to a chemoprotec-
tive milieu [108]. Moreover, at MRD patients are clinically well and
generally off SoC therapy and hence more likely to tolerate any
potential therapeutic side effects compared to being treated at
relapse [109].
Identification, characterization, validation, and developing tar-
geted therapies against MRD will allow for the development of
precision treatment modalities that would most likely improve
patient outcomes and may lead to a potential cure [110]. Unfortu-
nately, it comes with a new set of clinical and technical challenges in
sampling, especially in solid tumors wherein repeated invasive pro-
cedures would lead to patient discomfort and reduce clinical appli-
cability [111]. To overcome this situation there is a need to develop
Cancer Stem Cells: Current Challenges and Future Perspectives 13

in vitro and in vivo models that would recapitulate human disease.


Recently, Qazi et al. [109] used a unique model of human GBM
recurrence along with barcoding technology to track the BTIC
clonal population through SoC revealing that MRD can be driven
by either prior fitness of the subclones or prior equipotency result-
ing in selection of subclones under therapeutic selection pressure.
Additionally, they profiled GBM temporally by bulk sequencing
and scRNA-seq which could be used for potential target mining
as well as predicting and anticipating recurrence. Also, several
patient-derived xenograft models have been used to study MRD
for breast cancer, AML, ALL, and melanomas.
Monitoring patients at MRD using iterative sampling especially
in liquid cancers is being used as a standard practice to tailor the
anticipation of relapse and to refine clinical decisions [112]. Detec-
tion of MRD above or below a threshold is used as a tool for
escalating or tapering off therapy, though still it is not being used
to select targeted therapies to truly exploit the vulnerabilities of
MRD. Additionally, there are several studies indicating the use of
adjuvant therapies after complete remission resulting in improved
clinical outcomes [113–115]. Clinical trials in colorectal cancer are
underway wherein MRD is being used to inform which patients
should be enrolled for an adjuvant therapy. Targeting CSCs at
MRD appears to be a tractable strategy for efficiently eliminating
the surviving CSCs after SoC and preventing relapse.

References

1. Sung H, Ferlay J, Siegel RL, Laversanne M, 6. El-Sayes N, Vito A, Mossman K (2021)


Soerjomataram I, Jemal A et al (2021) Global Tumor heterogeneity: a great barrier in the
cancer statistics 2020: GLOBOCAN esti- age of cancer immunotherapy. Cancers
mates of incidence and mortality worldwide 13(4):806
for 36 cancers in 185 countries. CA Cancer J 7. Neftel C, Laffy J, Filbin MG, Hara T, Shore
Clin 71(3):209–249 ME, Rahme GJ et al (2019) An integrative
2. Skaane P, Gullien R, Bjørndal H, Eben EB, model of cellular states, plasticity and genetics
Ekseth U, Haakenaasen U et al (2012) Digital for glioblastoma. Cell 178(4):835–849.e21
breast tomosynthesis (DBT): initial experi- 8. Jamal-Hanjani M, Quezada SA, Larkin J,
ence in a clinical setting. Acta Radiol 53(5): Swanton C (2015) Translational implications
524–529 of tumor heterogeneity. Clin Cancer Res
3. Romond EH, Perez EA, Bryant J, Suman VJ, 21(6):1258–1266
Geyer CE, Davidson NE et al (2005) Trastu- 9. Patel AP, Tirosh I, Trombetta JJ, Shalek AK,
zumab plus adjuvant chemotherapy for oper- Gillespie SM, Wakimoto H et al (2014)
able HER2-positive breast cancer. N Engl J Single-cell RNA-seq highlights intratumoral
Med 353(16):1673–1684 heterogeneity in primary glioblastoma. Sci-
4. Baselga J, Cortés J, Kim SB, Im SA, Hegg R, ence 344(6190):1396–1401
Im YH et al (2012) Pertuzumab plus Trastu- 10. Mroz EA, Rocco JW (2013) MATH, a novel
zumab plus Docetaxel for metastatic breast measure of intratumor genetic heterogeneity,
cancer. N Engl J Med 366(2):109–119 is high in poor-outcome classes of head and
5. Oberstein PE, Olive KP (2013) Pancreatic neck squamous cell carcinoma. Oral Oncol
cancer: why is it so hard to treat? Ther Adv 49(3):211–215
Gastroenterol 6(4):321–337
14 Muhammad Vaseem Shaikh et al.

11. Greaves M, Maley CC (2012) Clonal evolu- characterization of human colorectal cancer
tion in cancer. Nature 481(7381):306–313 stem cells. Proc Natl Acad Sci U S A
12. Steinbichler TB, Dudás J, Skvortsov S, 104(24):10158–10163
Ganswindt U, Riechelmann H, Skvortsova II 25. Al-Hajj M, Wicha MS, Benito-Hernandez A,
(2018) Therapy resistance mediated by cancer Morrison SJ, Clarke MF (2003) Prospective
stem cells. Semin Cancer Biol 53:156–167 identification of tumorigenic breast cancer
13. Baisiwala S, Hall RR, Saathoff MR, cells. Proc Natl Acad Sci U S A 100(7):
Shireman J, Park C, Budhiraja S et al (2020) 3983–3988
LNX1 modulates Notch1 signaling to pro- 26. Snyder EL, Bailey D, Shipitsin M, Polyak K,
mote expansion of the glioma stem cell popu- Loda M (2009) Identification of CD44v6+/
lation during Temozolomide therapy in CD24- breast carcinoma cells in primary
glioblastoma. Cancers 12(12):E3505 human tumors by quantum dot-conjugated
14. Walcher L, Kistenmacher AK, Suo H, Kitte R, antibodies. Lab Investig 89(8):857–866
Dluczek S, Strauß A et al (2020) Cancer stem 27. Kemper K, Grandela C, Medema JP (2010)
cells—origins and biomarkers: perspectives Molecular identification and targeting of
for targeted personalized therapies. Front colorectal cancer stem cells. Oncotarget
Immunol 11:1280 1(6):387–395
15. Bonnet D, Dick JE (1997) Human acute 28. Ginestier C, Hur MH, Charafe-Jauffret E,
myeloid leukemia is organized as a hierarchy Monville F, Dutcher J, Brown M et al
that originates from a primitive hematopoietic (2007) ALDH1 is a marker of normal and
cell. Nat Med 3(7):730–737 malignant human mammary stem cells and a
16. Al-Hajj M, Wicha MS, Benito-Hernandez A, predictor of poor clinical outcome. Cell Stem
Morrison SJ, Clarke MF (2003) Prospective Cell 1(5):555–567
identification of tumorigenic breast cancer 29. Deng S, Yang X, Lassus H, Liang S, Kaur S, Ye
cells. Proc Natl Acad Sci 100(7):3983–3988 Q et al (2010) Distinct expression levels and
17. Singh SK, Hawkins C, Clarke ID, Squire JA, patterns of stem cell marker, aldehyde dehy-
Bayani J, Hide T et al (2004) Identification of drogenase isoform 1 (ALDH1), in human
human brain tumour initiating cells. Nature epithelial cancers. PLoS One 5(4):e10277
432(7015):396–401 30. Ginestier C, Wicinski J, Cervera N,
18. Eramo A, Lotti F, Sette G, Pilozzi E, Monville F, Finetti P, Bertucci F et al (2009)
Biffoni M, Di Virgilio A et al (2008) Identifi- Retinoid signaling regulates breast cancer
cation and expansion of the tumorigenic lung stem cell differentiation. Cell Cycle 8(20):
cancer stem cell population. Cell Death Differ 3297–3302
15(3):504–514 31. Kemper K, Sprick MR, de Bree M,
19. Ma S, Chan KW, Hu L, Lee TKW, Wo JYH, Scopelliti A, Vermeulen L, Hoek M et al
Ng IOL et al (2007) Identification and char- (2010) The AC133 epitope, but not the
acterization of tumorigenic liver cancer stem/ CD133 protein, is lost upon cancer stem cell
progenitor cells. Gastroenterology 132(7): differentiation. Cancer Res 70(2):719–729
2542–2556 32. Mak AB, Blakely KM, Williams RA, Penttil€a
20. Li C, Heidt DG, Dalerba P, Burant CF, PA, Shukalyuk AI, Osman KT et al (2011)
Zhang L, Adsay V et al (2007) Identification CD133 protein N-glycosylation processing
of pancreatic cancer stem cells. Cancer Res contributes to cell surface recognition of the
67(3):1030–1037 primitive cell marker AC133 epitope. J Biol
21. Schatton T, Murphy GF, Frank NY, Chem 286(47):41046–41056
Yamaura K, Waaga-Gasser AM, Gasser M 33. Sun Y, Kong W, Falk A, Hu J, Zhou L, Pollard
et al (2008) Identification of cells initiating S et al (2009) CD133 (Prominin) negative
human melanomas. Nature 451(7176): human neural stem cells are clonogenic and
345–349 tripotent. PLoS One 4(5):e5498
22. Medema JP (2013) Cancer stem cells: the 34. Yi JM, Tsai HC, Glöckner SC, Lin S, Ohm JE,
challenges ahead. Nat Cell Biol 15(4): Easwaran H et al (2008) Abnormal DNA
338–344 methylation of CD133 in colorectal and glio-
23. Pattabiraman DR, Weinberg RA (2014) Tack- blastoma tumors. Cancer Res 68(19):
ling the cancer stem cells – what challenges do 8094–8103
they pose? Nat Rev Drug Discov 13(7): 35. Chaffer CL, Brueckmann I, Scheel C, Kaestli
497–512 AJ, Wiggins PA, Rodrigues LO et al (2011)
24. Dalerba P, Dylla SJ, Park IK, Liu R, Wang X, Normal and neoplastic nonstem cells can
Cho RW et al (2007) Phenotypic spontaneously convert to a stem-like state.
Cancer Stem Cells: Current Challenges and Future Perspectives 15

Proc Natl Acad Sci U S A 108(19): stem cells by high-throughput screening. Cell
7950–7955 138(4):645–659
36. Gupta PB, Fillmore CM, Jiang G, Shapira SD, 47. Farmer P, Bonnefoi H, Anderle P,
Tao K, Kuperwasser C et al (2011) Stochastic Cameron D, Wirapati P, Becette V et al
state transitions give rise to phenotypic equi- (2009) A stroma-related gene signature pre-
librium in populations of cancer cells. Cell dicts resistance to neoadjuvant chemotherapy
146(4):633–644 in breast cancer. Nat Med 15(1):68–74
37. Borovski T, Verhoeff JJC, ten Cate R, 48. Kurrey NK, Jalgaonkar SP, Joglekar AV, Gha-
Cameron K, de Vries NA, van Tellingen O nate AD, Chaskar PD, Doiphode RY et al
et al (2009) Tumor microvasculature sup- (2009) Snail and slug mediate radioresistance
ports proliferation and expansion of glioma- and chemoresistance by antagonizing
propagating cells. Int J Cancer 125(5): p53-mediated apoptosis and acquiring a
1222–1230 stem-like phenotype in ovarian cancer cells.
38. Friedmann-Morvinski D, Bushong EA, Ke E, Stem Cells 27(9):2059–2068
Soda Y, Marumoto T, Singer O et al (2012) 49. Venugopal C, Hallett R, Vora P,
Dedifferentiation of neurons and astrocytes Manoranjan B, Mahendram S, Qazi MA et al
by oncogenes can induce gliomas in mice. (2015) Pyrvinium targets CD133 in human
Science 338(6110):1080–1084 glioblastoma brain tumor–initiating cells.
39. Vermeulen L, De Sousa E, Melo F, van der Clin Cancer Res 21(23):5324–5337
Heijden M, Cameron K, de Jong JH, Bor- 50. Sica A, Allavena P, Mantovani A (2008) Can-
ovski T et al (2010) Wnt activity defines cer related inflammation: the macrophage
colon cancer stem cells and is regulated by connection. Cancer Lett 267(2):204–215
the microenvironment. Nat Cell Biol 12(5): 51. Garziera M, Roncato R, Montico M, De
468–476 Mattia E, Gagno S, Poletto E et al (2019)
40. Scheel C, Eaton EN, Li SHJ, Chaffer CL, New challenges in tumor mutation heteroge-
Reinhardt F, Kah KJ et al (2011) Paracrine neity in advanced ovarian cancer by a targeted
and autocrine signals induce and maintain next-generation sequencing (NGS) approach.
mesenchymal and stem cell states in the Cell 8(6):E584
breast. Cell 145(6):926–940 52. Sathe A, Grimes SM, Lau BT, Chen J,
41. Bruna A, Greenwood W, Le Quesne J, Suarez C, Huang RJ et al (2020) Single-cell
Teschendorff A, Miranda-Saavedra D, Rueda genomic characterization reveals the cellular
OM et al (2012) TGFβ induces the formation reprogramming of the gastric tumor microen-
of tumour-initiating cells in claudinlow breast vironment. Clin Cancer Res 26(11):
cancer. Nat Commun 3(1):1055 2640–2653
42. Chen J, Li Y, Yu TS, McKay RM, Burns DK, 53. Sun G, Li Z, Rong D, Zhang H, Shi X, Yang
Kernie SG et al (2012) A restricted cell popu- W et al (2021) Single-cell RNA sequencing in
lation propagates glioblastoma growth after cancer: applications, advances, and emerging
chemotherapy. Nature 488(7412):522–526 challenges. Mol Ther Oncolytics 21:183–206
43. Roesch A, Fukunaga-Kalabis M, Schmidt EC, 54. Tang F, Barbacioru C, Wang Y, Nordman E,
Zabierowski SE, Brafford PA, Vultur A et al Lee C, Xu N et al (2009) mRNA-Seq whole-
(2010) A temporarily distinct subpopulation transcriptome analysis of a single cell. Nat
of slow-cycling melanoma cells is required for Methods 6(5):377–382
continuous tumor growth. Cell 141(4): 55. Navin N, Kendall J, Troge J, Andrews P,
583–594 Rodgers L, McIndoo J et al (2011) Tumour
44. Grimm M, Krimmel M, Polligkeit J, evolution inferred by single-cell sequencing.
Alexander D, Munz A, Kluba S et al (2012) Nature 472(7341):90–94
ABCB5 expression and cancer stem cell 56. Aceto N, Bardia A, Miyamoto DT, Donald-
hypothesis in oral squamous cell carcinoma. son MC, Wittner BS, Spencer JA et al (2014)
Eur J Cancer 48(17):3186–3197 Circulating tumor cell clusters are oligoclonal
45. Moitra K, Lou H, Dean M (2011) Multidrug precursors of breast cancer metastasis. Cell
efflux pumps and cancer stem cells: insights 158(5):1110–1122
into multidrug resistance and therapeutic 57. Lei B, Zhang XY, Zhou JP, Mu GN, Li YW,
development. Clin Pharmacol Ther 89(4): Zhang YX et al (2016) Transcriptome
491–502 sequencing of HER2-positive breast cancer
46. Gupta PB, Onder TT, Jiang G, Tao K, stem cells identifies potential prognostic
Kuperwasser C, Weinberg RA et al (2009) marker. Tumour Biol 37(11):14757–14764
Identification of selective inhibitors of cancer
16 Muhammad Vaseem Shaikh et al.

58. Chung W, Eum HH, Lee HO, Lee KM, Lee CRISPR–Cas9 screen identifies ifenprodil as
HB, Kim KT et al (2017) Single-cell RNA-seq an adjunct to sorafenib for liver cancer treat-
enables comprehensive tumour and immune ment. Cancer Res 81(24):6219–6232
cell profiling in primary breast cancer. Nat 71. Decker CE, Young T, Pasnikowski E, Chiu J,
Commun 8:15081 Song H, Wei Y et al (2019) Genome-scale
59. Zhang K, Erkan EP, Jamalzadeh S, Dai J, CRISPR activation screen uncovers tumor-
Andersson N, Kaipio K et al (2022) Longitu- intrinsic modulators of CD3 bispecific anti-
dinal single-cell RNA-seq analysis reveals body efficacy. Sci Rep 9(1):20068
stress-promoted chemoresistance in meta- 72. Joung J, Kirchgatterer PC, Singh A, Cho JH,
static ovarian cancer. Sci Adv 8(8):eabm1831 Nety SP, Larson RC et al (2022) CRISPR
60. Bibikova M, Beumer K, Trautman JK, Carroll activation screen identifies BCL-2 proteins
D (2003) Enhancing gene targeting with and B3GNT2 as drivers of cancer resistance
designed zinc finger nucleases. Science to T cell-mediated cytotoxicity. Nat Commun
300(5620):764–764 13(1):1606
61. Boch J, Scholze H, Schornack S, Landgraf A, 73. Wang D, Prager BC, Gimple RC, Aguilar B,
Hahn S, Kay S et al (2009) Breaking the code Alizadeh D, Tang H et al (2021) CRISPR
of DNA binding specificity of TAL-type III screening of CAR T cells and cancer stem
effectors. Science 326(5959):1509–1512 cells reveals critical dependencies for cell-
62. Knight SC, Xie L, Deng W, Guglielmi B, Wit- based therapies. Cancer Discov 11(5):
kowsky LB, Bosanac L et al (2015) Dynamics 1192–1211
of CRISPR-Cas9 genome interrogation in liv- 74. Ramaker RC, Hardigan AA, Gordon ER,
ing cells. Science 350(6262):823–826 Wright CA, Myers RM, Cooper SJ (2021)
63. Zhang F, Wen Y, Guo X (2014) CRISPR/ Pooled CRISPR screening in pancreatic can-
Cas9 for genome editing: progress, implica- cer cells implicates co-repressor complexes as a
tions and challenges. Hum Mol Genet 23 cause of multiple drug resistance via regula-
(R1):R40–R46 tion of epithelial-to-mesenchymal transition.
64. Shalem O, Sanjana NE, Hartenian E, Shi X, BMC Cancer 21(1):632
Scott DA, Mikkelson T et al (2014) Genome- 75. Dagher R, Cohen M, Williams G,
scale CRISPR-Cas9 knockout screening in Rothmann M, Gobburu J, Robbie G et al
human cells. Science 343(6166):84–87 (2002) Approval summary: imatinib mesylate
65. Hart T, Chandrashekhar M, Aregger M, in the treatment of metastatic and/or unre-
Steinhart Z, Brown KR, MacLeod G et al sectable malignant gastrointestinal stromal
(2015) High-resolution CRISPR screens tumors. Clin Cancer Res 8(10):3034–3038
reveal fitness genes and genotype-specific can- 76. Zhao Y, Tong C, Jiang J (2007) Hedgehog
cer liabilities. Cell 163(6):1515–1526 regulates smoothened activity by inducing a
66. Chen S, Sanjana NE, Zheng K, Shalem O, conformational switch. Nature 450(7167):
Lee K, Shi X et al (2015) Genome-wide 252–258
CRISPR screen in a mouse model of tumor 77. Sekulic A, Migden MR, Oro AE, Dirix L,
growth and metastasis. Cell 160(6): Lewis KD, Hainsworth JD et al (2012) Effi-
1246–1260 cacy and safety of vismodegib in advanced
67. Gao S, Soares F, Wang S, Wong CC, Chen H, basal-cell carcinoma. N Engl J Med 366(23):
Yang Z et al (2021) CRISPR screens identify 2171–2179
cholesterol biosynthesis as a therapeutic target 78. Norsworthy KJ, By K, Subramaniam S,
on stemness and drug resistance of colon can- Zhuang L, Del Valle PL, Przepiorka D et al
cer. Oncogene 40(48):6601–6613 (2019) FDA approval summary: glasdegib for
68. Wei J, Long L, Zheng W, Dhungana Y, Lim newly diagnosed acute myeloid leukemia. Clin
SA, Guy C et al (2019) Targeting REGNASE- Cancer Res 25(20):6021–6025
1 programs long-lived effector T cells for can- 79. Quail DF, Taylor MJ, Postovit LM (2012)
cer therapy. Nature 576(7787):471–476 Microenvironmental regulation of cancer
69. MacLeod G, Bozek DA, Rajakulendran N, stem cell phenotypes. Curr Stem Cell Res
Monteiro V, Ahmadi M, Steinhart Z et al Ther 7(3):197–216
(2019) Genome-wide CRISPR-Cas9 screens 80. Hoffman LM, Fouladi M, Olson J, Daryani
expose genetic vulnerabilities and mechanisms VM, Stewart CF, Wetmore C et al (2015)
of Temozolomide sensitivity in glioblastoma Phase I trial of weekly MK-0752 in children
stem cells. Cell Rep 27(3):971–986.e9 with refractory central nervous system malig-
70. Xu F, Tong M, Tong CSW, Chan BKC, Chu nancies: a pediatric brain tumor consortium
HY, Wong TL et al (2021) A combinatorial study. Childs Nerv Syst 31(8):1283–1289
Cancer Stem Cells: Current Challenges and Future Perspectives 17

81. Su M, Zhang Q, Bai X, Wu C, Li Y, Mossialos lenalidomide in patients with advanced can-


E et al (2017) Availability, cost, and prescrip- cers. Mol Cancer Ther 17(3):671–676
tion patterns of antihypertensive medications 93. Migden MR, Rischin D, Schmults CD,
in primary health care in China: a nationwide Guminski A, Hauschild A, Lewis KD et al
cross-sectional survey. Lancet 390(10112): (2018) PD-1 blockade with cemiplimab in
2559–2568 advanced cutaneous squamous-cell carci-
82. Maxmen A (2016) Busting the billion-dollar- noma. N Engl J Med 379(4):341–351
myth: how to slash the cost of drug develop- 94. Motzer RJ, Penkov K, Haanen J, Rini B,
ment. Nature 536(7617):388–390 Albiges L, Campbell MT et al (2019) Avelu-
83. Lamb J, Crawford ED, Peck D, Modell JW, mab plus axitinib versus sunitinib for
Blat IC, Wrobel MJ et al (2006) The advanced renal-cell carcinoma. N Engl J Med
connectivity map: using gene-expression sig- 380(12):1103–1115
natures to connect small molecules, genes, 95. Park JH, Rivière I, Gonen M, Wang X,
and disease. Science 313(5795):1929–1935 Sénéchal B, Curran KJ et al (2018) Long-
84. Vidović D, Koleti A, Schürer SC (2014) term follow-up of CD19 CAR therapy in
Large-scale integration of small molecule- acute lymphoblastic leukemia. N Engl J Med
induced genome-wide transcriptional 378(5):449–459
responses, Kinome-wide binding affinities 96. Maude SL, Frey N, Shaw PA, Aplenc R, Bar-
and cell-growth inhibition profiles reveal rett DM, Bunin NJ et al (2014) Chimeric
global trends characterizing systems-level antigen receptor T cells for sustained remis-
drug action. Front Genet 5:342 sions in leukemia. N Engl J Med 371(16):
85. Singh M, Venugopal C, Tokar T, 1507–1517
McFarlane N, Subapanditha MK, Qazi M 97. Fry TJ, Shah NN, Orentas RJ, Stetler-
et al (2018) Therapeutic targeting of the pre- Stevenson M, Yuan CM, Ramakrishna S et al
metastatic stage in human lung-to-brain (2018) CD22-targeted CAR T cells induce
metastasis. Cancer Res 78(17):5124–5134 remission in B-ALL that is naive or resistant
86. Chaiteerakij R, Petersen GM, Bamlet WR, to CD19-targeted CAR immunotherapy. Nat
Chaffee KG, Zhen DB, Burch PA et al Med 24(1):20–28
(2016) Metformin use and survival of patients 98. Kahl B (2008) Chemotherapy combinations
with pancreatic cancer: a cautionary lesson. with monoclonal antibodies in non-Hodg-
JCO 34(16):1898–1904 kin’s lymphoma. Semin Hematol 45(2):
87. Lübbert M, Grishina O, Schmoor C, Schlenk 90–94
RF, Jost E, Crysandt M et al (2020) Valproate 99. Stupp R, Mason WP, van den Bent MJ,
and retinoic acid in combination with Decita- Weller M, Fisher B, Taphoorn MJB et al
bine in elderly nonfit patients with acute mye- (2005) Radiotherapy plus concomitant and
loid leukemia: results of a multicenter, adjuvant temozolomide for glioblastoma. N
randomized, 2 × 2. Phase II Trial J Clin Engl J Med 352(10):987–996
Oncol 38(3):257–270 100. Adler S, Seidel J, Choyke P, Knopp MV,
88. Hiam-Galvez KJ, Allen BM, Spitzer MH Binzel K, Zhang J et al (2017) Minimum
(2021) Systemic immunity in cancer. Nat lesion detectability as a measure of PET sys-
Rev Cancer 21(6):345–359 tem performance. EJNMMI Phys 4(1):13
89. Prager BC, Xie Q, Bao S, Rich JN (2019) 101. Bao S, Wu Q, McLendon RE, Hao Y, Shi Q,
Cancer stem cells: the architects of the tumor Hjelmeland AB et al (2006) Glioma stem cells
ecosystem. Cell Stem Cell 24(1):41–53 promote radioresistance by preferential acti-
90. Lytle NK, Barber AG, Reya T (2018) Stem vation of the DNA damage response. Nature
cell fate in cancer growth, progression and 444(7120):756–760
therapy resistance. Nat Rev Cancer 18(11): 102. Liu G, Yuan X, Zeng Z, Tunici P, Ng H,
669–680 Abdulkadir IR et al (2006) Analysis of gene
91. Vora P, Venugopal C, Salim SK, Tatari N, expression and chemoresistance of CD133+
Bakhshinyan D, Singh M et al (2020) The cancer stem cells in glioblastoma. Mol Cancer
rational development of CD133-targeting 5(1):67
immunotherapies for glioblastoma. Cell 103. Rambow F, Rogiers A, Marin-Bejar O,
Stem Cell 26(6):832–844.e6 Aibar S, Femel J, Dewaele M et al (2018)
92. Sakamuri D, Glitza IC, Betancourt Cuellar Toward minimal residual disease-directed
SL, Subbiah V, Fu S, Tsimberidou AM et al therapy in melanoma. Cell 174(4):843–855.
(2018) Phase I dose-escalation study of anti- e19
CTLA-4 antibody Ipilimumab and
Another random document with
no related content on Scribd:
He was soon at the head of a large and powerful army. With this
he marched forward, defeated the English troops that advanced to
meet him, and, in three months after his arrival, he took Edinburgh,
the capital of Scotland.—​France now sent him aid, and, with a force
of 7000 men, he marched southward into England, and took the
town of Carlisle. At Preston Pans, he defeated an English army of
4000 strong; and such was his success, that the English
government, under King William, of Orange, trembled for their safety.
They therefore made great efforts, and in April, 1746, they sent a
large army against him, under the Duke of Cumberland. At Culloden,
the two armies met, and a terrible battle followed; Prince Edward
was defeated, and his army entirely dispersed. He was scarce able
to save his life by flight; and, indeed, he wandered about, from place
to place, among the wilds of Scotland, being every day in danger of
being seized and given up to the English government, who offered
$150,000 to anybody who would bring him to them. It seems strange
that so large a bribe could be resisted; but, such was the love that
the Scottish people bore him, and such their fidelity, that no one was
found to betray him, though many people were entrusted with the
secret of his being among them. Even the poor mountaineers
refused to give him up, though offered a sum of money that would
have made them very rich.
At last, a faithful Scottish nobleman, by the name of O’Neil, took
him in charge, and after wandering along the sea-shore in a skiff,
flying from island to island, and experiencing the greatest sufferings
and dangers, he was put on board a French frigate, that had been
sent for his rescue. He was now taken to France, and soon after,
giving up all hopes of seeing his family restored to the throne, he
settled in Italy, where he died in 1788, in the 68th year of his age. He
was the last of the Stuart line, and was called the Pretender, on
account of his pretending to set up claims to the throne of England.
Winter.

December has come! Winter is here! These are common-place


words, but they mean more, perhaps, than we are apt to consider.
Winter, then, means that the myriad leaves of the forest are
shrivelled and torn from the trees, and scattered in the valley: it
means that the sap of the trees has ceased to flow, and that these
giants of the vegetable world have passed into a state of stupor, in
which they must remain till spring again returns.
Winter means that the myriad races of annual weeds and plants
are dead, to revive again no more; that myriads of blossoms have
faded forever from the view; that the verdure of the forest has
passed away; that the gemmed garment of the meadow is
exchanged for the thin, brown mantle of leanness and poverty; that
the velvet of the lawn has given place to the scanty covering of dried
and faded grass.
Winter means that the minstrelsy of the birds is gone, and that the
field and forest, so lately cheered by a thousand forms and sounds
of happy existence are now silent, or rendered more dreary and
desolate by the moaning winds. It means that the birds are gone to
their southern retreats; that the myriad races of insects are dead;
that the whole generation of butterflies has perished; that the
grasshoppers have sung their last song; that even the pensive
cricket has gone to his long home. It means that death has breathed
on our portion of the world, and that nature herself, as if weary of her
efforts, has fallen into a cold and fearful slumber.
Winter means all these melancholy things; but it also means
something more. It means that the granary of the farmer is full; that
his barn is supplied; that there is good and ample store for the
beasts that look to man for support, and for man himself. It means,
too, that the comfortable fire will be kindled, around which the family
will assemble, and where, secure from the bitter blast without, there
will still be peace, comfort, and content. It means, too, that there is
such a thing as poverty, shivering, without fire, without food—
perhaps, without sufficient shelter; and it means that charity should
seek and save those who are suffering in such a condition.
And winter means something more than all this: it means, by its
examples of decay and death, to teach us that we, too, must pass
away; and that it is well for us to make preparation for the great
event. Winter also brings us to the end of the year, and suggests a
serious self-inquiry, and self-examination. It would ask us if the last
year has been one of profit or loss? Are we better, and wiser, than
when it began? Are we more kind, more just, more patient, more
faithful, more fond of truth?—Summer is the season for the harvest
of the field; winter is the season for the moral harvest of the heart.
Let it not pass with any of us as a barren and unproductive season,
in which we neither sow nor reap the fruits of wisdom and peace.
The Hand.

Every limb and member of the body is made for some good
purpose.
The eye is made to see with; the ear is made to hear with; the
nose is made to smell with; the mouth is made to eat and speak with.
The feet are made to run and walk with; the hands are made to
work with, to write with, and to do many other things.
But do you think children’s hands were ever made to strike their
brothers, or sisters, or playmates? Were your little hands ever made
to snatch away things from each other?
Who gave you hands? God gave them. Did he give you hands to
steal with? Did God give you hands that you might throw stones at
geese, or dogs, or hens, or cows, or any other innocent animals?
Did God give you hands to injure or wound any of the creatures
he has made?
Take care of your little hands, then, my children! Take care that
the hands God has given, do nothing that God disapproves.
Nuts to Crack.

The Word “Fast.”—This is as great a contradiction as we have in


the language. The river is fast, because the ice is immoveable; and
then the ice disappears fast for the contrary reason—it is loose. A
clock is called fast when it goes quicker than time; but a man is told
to stand fast, when he is desired to remain stationary. People fast
when they have nothing to eat, and eat fast when opportunity offers.

Military Courtesy.—Gen. Meadows, equally renowned for his


wit and bravery, being on a reconnoitring party, in the Mysore
country, a twenty-four pound shot struck the ground at some
distance from the General, and was passing in such a direction as
would have exposed him to danger had he continued on his route;
quick as lightning he stopped his horse, and, pulling off his hat very
gracefully, as the shot rolled on, good-humoredly said: “I beg you to
proceed, sir; I never dispute precedence with any gentleman of your
family.”

A doctor, in Scotland, was employed by a poor man to attend


his wife, who was dangerously ill. The doctor gave a hint, amounting
to the suspicion that he would not be paid. “I have,” says the man,
“five pounds; and if you kill, or cure her, you shall have it.” The
woman died, under the hands of the doctor, and, after a reasonable
time, he called for his five pounds. The man then said: “Did you kill
my wife?—did you cure her?” “No.” “Then,” said the poor man, “you
have no legal demand,” and turned upon his heel.
How to shake off Trouble.—Set about doing good to
somebody: put on your hat, and go and visit the sick and poor—
inquire into their wants, and minister to them; seek out the desolate
and oppressed, and tell them of the consolations of religion. I have
often tried this method, and have always found it the best medicine
for a heavy heart.

A Father’s Impulse.—When Lord Erskine made his debut at the


bar, his agitation almost overpowered him, and he was just going to
sit down: “At that moment,” said he, “I thought I felt my little children
tugging at my gown, and the idea roused me to an exertion, of which
I did not think myself capable.”

The Sublime.—Over the stall of a public writer, in Rue de Bac, at


Paris, is the following inscription: “M. Renard, public writer and
compiler—translates the tongues, explains the language of flowers,
and sells fried potatoes.”

Feeling for Another.—A Quaker, once hearing a person tell


how much he felt for a friend who needed his assistance, dryly
observed: “Friend, hast thou ever felt in thy pocket for him?”

“What are you writing such a thundering big hand for, Patrick?”
“Why, do you see, my grandmother is deaf, and I am writing a loud
lether to her.”

A Knotty Case.—Not many years ago, a man appeared in court,


whether as plaintiff, defendant, or witness, tradition does not inform
us. Be this as it may, the following dialogue ensued:—Court—“What
is your name, sir?” “My name is Knott Martin, your honor.” “Well,
what is it?” “It is Knott Martin.” “Not Martin, again! We do not ask you
what your name is not, but what it is. No contempt of court, sir.” “If
your honor will give me leave, I will spell my name.” “Well, spell it.”
“K-n-o-tt, Knott, M-a-r, Mar, t-i-n, tin—Knott Martin.” “O, well, Mr.
Martin, we see through it now; but it is one of the most knotty cases
we have had before us for some time.”

Good.—It was a judicious resolution of a father, as well as a most


pleasing compliment to his wife, when, on being asked by a friend
what he intended to do with his daughters, he replied: “I intend to
apprentice them to their mother, that they may become like her—
good wives, mothers, heads of families, and useful members of
society.”

A Learned Character.—“Give me ‘Venice Preserved,’” said a


gentleman, last week, on going to a celebrated bookseller’s at the
West-end. “We don’t sell preserves,” said an apprentice, newly-
imported from the country; “but you will get them next door, at Mr.
Brown’s, the confectioner.”

Ten To One.—Strict attention to office hours is a duty incumbent


upon every public officer. We heard of a case of an American consul,
in a foreign country, who was not remarkable for his attention to duty.
A gentleman, calling one day, found his office shut, and a label
sticking upon the door, with these words: “In from ten to one.” Having
called again several times within those hours, without finding him, he
wrote at the bottom of the label—“Ten to one he’s not in.”
To the Black-ey’d and Blue-ey’d Friends of
Robert Merry.

It is now about a twelvemonth since our acquaintance


commenced; and I hope the feeling is such between us, that there is
a mutual desire to continue it. I know that the young, the happy, and
the gay-hearted, are apt to think that we old fellows are sour and sad
—disposed to look with an evil eye upon childhood and its sports;
and more ready to preach than practise charity.
I will not pretend to deny that, now and then, a person gets cross
and crabbed as he grows old, and like cider too long kept, turns to
vinegar: but this is not my case, or, if it be, my ill-humor never
displays itself toward the young. They are to me the buds and
blossoms of life, and their presence ever brings the welcome
feelings that belong to sunshine and summer.
Old age has been often compared to winter—the close of the
year; the season of desolation; the period of storms and tempests;
the funeral-time of the vegetable world; the time when the leaves,
the fruits, and the flowers are laid in their tomb, and covered over
with a winding-sheet of snow. This is a sad picture at first view; and I
believe many a child is led to avoid old people from the habit of
regarding them in this light—from the idea that they are shrivelled,
frost-bitten, bitter, and disagreeable.
Now, I will not deny that there is some resemblance between
winter and old age: an old man has not the warm blood of youth; his
pulses are, perhaps, like the river, chilled and obstructed by ice; his
temper is sometimes capricious and gusty, like the winds of
December; and his head, bald, or covered with a few silvery hairs, is
like the oak, stripped of its covering, and having its boughs
powdered with snow.
All this may be true enough; but it is not good reason why the old
should be deserted by the young. I remember very well, that, when I
was a boy, there was a fine old walnut-tree, upon a hillside, not far
from where I lived. Now, I never thought or cared about this tree, till
the time when winter approached. Then, when the leaves were
scattered, the nuts were all ripe, then it was that the tree became an
object of interest to me. Then it was that I loved to visit it; to climb its
limbs and give it a shake, and hear the fruit rattle down like hail.
Never, in all my boyhood days, did I meet with anything more
delightful than this!
And let me tell you, my black-ey’d and blue-ey’d friends, that this
old walnut-tree was like many an old person you may meet with. You
will remark that, in this case, it was when winter had come, or was
near at hand, that the fruit was ripe, and ready for those who would
climb up for it and gather it. And let me tell you, that old people, like
this tree, have many a good nut to crack, many a good story to tell,
to those who will climb up in the lap and ask for it.
This is my view of the matter; and I hope that young people,
instead of running away from me, as a crusty, crabbed, one-legged
old chap, will treat me as I did the old walnut-tree—give it a shake,
and see if the nuts don’t rattle down!
I am not fond of making great promises; but, as I am anxious to
have my readers, who have set out on a journey with me, still keep
me company—at least for one year more—I am ready to engage to
do my best to please them. I shall, if I live, tell the rest of my own
story, and bring the history of Brusque to a close. The tale of the
Sable-Hunters, the travels of Thomas Trotter, the stories of the
Indians, will be continued and completed; and a variety of other
things are in store.
I can promise one thing more—and that is, some tales from the
pen of Peter Parley. That pleasant, kind-hearted old man is no more;
but I knew him better than anybody else, and all his papers are in my
hands. Among them are several tales, and I intend to publish them in
my magazine. My young readers, perhaps, do not know how
shabbily poor old Peter was treated. The fact was, that several
people in this country, as well as in others, wrote stories, and put his
name to them; thus pretending that they were actually his! Some of
these were very silly, and some were very improper. This cut Peter to
the heart, and it served greatly to shorten his days. I am sorry that,
even now, people are palming off trumpery works of their own as
Peter Parley’s.
But the tales that I propose to give, are genuine; there is no
mistake. They are by the same hand that wrote the tales about
Europe, Asia, Africa, and America; and I hope they may be as
acceptable as those were.
I return a thousand thanks to my many young friends, who have
written me letters, whether of criticism, advice, or commendation. I
am glad to know that so many of them like Bill Keeler: let them be
assured his whole story will come out in due time. I shall be very
glad to get the bear story, which L. S., of Vermont, offers to tell. The
Indiana legend of the Wolf and the Wild-cat, is received, and will
appear soon. Jane R—— will accept my thanks for—she knows
what! If she were not so many hundred miles off, I should ask her to
let me see whether she is a blue-eyed or black-eyed friend. The
basket of chestnuts were duly received from Alice D——, and were
very welcome. Ralph H—— will see that I have done as he
requested; I have given a portrait of the fine gray squirrel he sent
me, in this number. He is well, and as lively as ever.
Robert Merry.
WINTER—A SONG.
the words and music composed for
merry’s museum.

“Tell me what does winter mean!”


’Tis a drea-ry change of scene—
When the meadow yields its bloom,
And the blo-soms seek their tomb.
Winter is the time of storms,
When the cloud in angry forms,
O’er the land in terror sweeps,
And the sighing forest weeps.
’Tis the funeral time of flowers,
Withered in their lovely bowers;
While the zephyr sings in grief,
O’er each shrivelled stem and leaf.
’Tis the dreary time of snow,
Falling chill on all below,
As a winding-sheet it weaves
O’er the graves of myriad leaves.

Winter is a time of tears,


For the poor, in youth or years,—
Where the storm drives keenly in,
And the blanket’s brief and thin.
Winter is the time of wreck,
When the billow cleaves the deck,
And the mariners go down
Where the battling surges frown.
Transcriber’s Note:
This book was written in a period when many words had not
become standardized in their spelling. Words may have multiple
spelling variations or inconsistent hyphenation in the text. These
have been left unchanged unless indicated below. Obsolete and
alternative spellings were left unchanged. Misspelled words were not
corrected.
One Footnote was moved to the end of the chapter. Obvious
printing errors, such as backwards, upside down, or partially printed
letters and punctuation, were corrected. Final stops missing at the
end of sentences and abbreviations were added. Duplicate letters at
line endings or page breaks were removed. Quotation marks were
adjusted to common usage. Page numbers in the Table of Contents
were corrected to match book pages.
Links to audio files were added for music. The music files are the
music transcriber’s interpretation of the printed notation and are
placed in the public domain. At the time of this writing, music file
links will not work in mobile e-book formats like epub or Kindle/mobi.
Users who are reading the e-book in one of these formats can listen
to the music or download music files in the HTML version. Lyrics to
musical scores are presented as poetry following the illustration of
the music.
*** END OF THE PROJECT GUTENBERG EBOOK ROBERT
MERRY'S MUSEUM, VOLUMES I AND II (1841) ***

Updated editions will replace the previous one—the old editions


will be renamed.

Creating the works from print editions not protected by U.S.


copyright law means that no one owns a United States copyright
in these works, so the Foundation (and you!) can copy and
distribute it in the United States without permission and without
paying copyright royalties. Special rules, set forth in the General
Terms of Use part of this license, apply to copying and
distributing Project Gutenberg™ electronic works to protect the
PROJECT GUTENBERG™ concept and trademark. Project
Gutenberg is a registered trademark, and may not be used if
you charge for an eBook, except by following the terms of the
trademark license, including paying royalties for use of the
Project Gutenberg trademark. If you do not charge anything for
copies of this eBook, complying with the trademark license is
very easy. You may use this eBook for nearly any purpose such
as creation of derivative works, reports, performances and
research. Project Gutenberg eBooks may be modified and
printed and given away—you may do practically ANYTHING in
the United States with eBooks not protected by U.S. copyright
law. Redistribution is subject to the trademark license, especially
commercial redistribution.

START: FULL LICENSE


THE FULL PROJECT GUTENBERG LICENSE
PLEASE READ THIS BEFORE YOU DISTRIBUTE OR USE THIS WORK

To protect the Project Gutenberg™ mission of promoting the


free distribution of electronic works, by using or distributing this
work (or any other work associated in any way with the phrase
“Project Gutenberg”), you agree to comply with all the terms of
the Full Project Gutenberg™ License available with this file or
online at www.gutenberg.org/license.

Section 1. General Terms of Use and


Redistributing Project Gutenberg™
electronic works
1.A. By reading or using any part of this Project Gutenberg™
electronic work, you indicate that you have read, understand,
agree to and accept all the terms of this license and intellectual
property (trademark/copyright) agreement. If you do not agree to
abide by all the terms of this agreement, you must cease using
and return or destroy all copies of Project Gutenberg™
electronic works in your possession. If you paid a fee for
obtaining a copy of or access to a Project Gutenberg™
electronic work and you do not agree to be bound by the terms
of this agreement, you may obtain a refund from the person or
entity to whom you paid the fee as set forth in paragraph 1.E.8.

1.B. “Project Gutenberg” is a registered trademark. It may only


be used on or associated in any way with an electronic work by
people who agree to be bound by the terms of this agreement.
There are a few things that you can do with most Project
Gutenberg™ electronic works even without complying with the
full terms of this agreement. See paragraph 1.C below. There
are a lot of things you can do with Project Gutenberg™
electronic works if you follow the terms of this agreement and
help preserve free future access to Project Gutenberg™
electronic works. See paragraph 1.E below.
1.C. The Project Gutenberg Literary Archive Foundation (“the
Foundation” or PGLAF), owns a compilation copyright in the
collection of Project Gutenberg™ electronic works. Nearly all the
individual works in the collection are in the public domain in the
United States. If an individual work is unprotected by copyright
law in the United States and you are located in the United
States, we do not claim a right to prevent you from copying,
distributing, performing, displaying or creating derivative works
based on the work as long as all references to Project
Gutenberg are removed. Of course, we hope that you will
support the Project Gutenberg™ mission of promoting free
access to electronic works by freely sharing Project
Gutenberg™ works in compliance with the terms of this
agreement for keeping the Project Gutenberg™ name
associated with the work. You can easily comply with the terms
of this agreement by keeping this work in the same format with
its attached full Project Gutenberg™ License when you share it
without charge with others.

1.D. The copyright laws of the place where you are located also
govern what you can do with this work. Copyright laws in most
countries are in a constant state of change. If you are outside
the United States, check the laws of your country in addition to
the terms of this agreement before downloading, copying,
displaying, performing, distributing or creating derivative works
based on this work or any other Project Gutenberg™ work. The
Foundation makes no representations concerning the copyright
status of any work in any country other than the United States.

1.E. Unless you have removed all references to Project


Gutenberg:

1.E.1. The following sentence, with active links to, or other


immediate access to, the full Project Gutenberg™ License must
appear prominently whenever any copy of a Project
Gutenberg™ work (any work on which the phrase “Project
Gutenberg” appears, or with which the phrase “Project
Gutenberg” is associated) is accessed, displayed, performed,
viewed, copied or distributed:

This eBook is for the use of anyone anywhere in the United


States and most other parts of the world at no cost and with
almost no restrictions whatsoever. You may copy it, give it
away or re-use it under the terms of the Project Gutenberg
License included with this eBook or online at
www.gutenberg.org. If you are not located in the United
States, you will have to check the laws of the country where
you are located before using this eBook.

1.E.2. If an individual Project Gutenberg™ electronic work is


derived from texts not protected by U.S. copyright law (does not
contain a notice indicating that it is posted with permission of the
copyright holder), the work can be copied and distributed to
anyone in the United States without paying any fees or charges.
If you are redistributing or providing access to a work with the
phrase “Project Gutenberg” associated with or appearing on the
work, you must comply either with the requirements of
paragraphs 1.E.1 through 1.E.7 or obtain permission for the use
of the work and the Project Gutenberg™ trademark as set forth
in paragraphs 1.E.8 or 1.E.9.

1.E.3. If an individual Project Gutenberg™ electronic work is


posted with the permission of the copyright holder, your use and
distribution must comply with both paragraphs 1.E.1 through
1.E.7 and any additional terms imposed by the copyright holder.
Additional terms will be linked to the Project Gutenberg™
License for all works posted with the permission of the copyright
holder found at the beginning of this work.

1.E.4. Do not unlink or detach or remove the full Project


Gutenberg™ License terms from this work, or any files
containing a part of this work or any other work associated with
Project Gutenberg™.
1.E.5. Do not copy, display, perform, distribute or redistribute
this electronic work, or any part of this electronic work, without
prominently displaying the sentence set forth in paragraph 1.E.1
with active links or immediate access to the full terms of the
Project Gutenberg™ License.

1.E.6. You may convert to and distribute this work in any binary,
compressed, marked up, nonproprietary or proprietary form,
including any word processing or hypertext form. However, if
you provide access to or distribute copies of a Project
Gutenberg™ work in a format other than “Plain Vanilla ASCII” or
other format used in the official version posted on the official
Project Gutenberg™ website (www.gutenberg.org), you must, at
no additional cost, fee or expense to the user, provide a copy, a
means of exporting a copy, or a means of obtaining a copy upon
request, of the work in its original “Plain Vanilla ASCII” or other
form. Any alternate format must include the full Project
Gutenberg™ License as specified in paragraph 1.E.1.

1.E.7. Do not charge a fee for access to, viewing, displaying,


performing, copying or distributing any Project Gutenberg™
works unless you comply with paragraph 1.E.8 or 1.E.9.

1.E.8. You may charge a reasonable fee for copies of or


providing access to or distributing Project Gutenberg™
electronic works provided that:

• You pay a royalty fee of 20% of the gross profits you derive from
the use of Project Gutenberg™ works calculated using the
method you already use to calculate your applicable taxes. The
fee is owed to the owner of the Project Gutenberg™ trademark,
but he has agreed to donate royalties under this paragraph to
the Project Gutenberg Literary Archive Foundation. Royalty
payments must be paid within 60 days following each date on
which you prepare (or are legally required to prepare) your
periodic tax returns. Royalty payments should be clearly marked
as such and sent to the Project Gutenberg Literary Archive
Foundation at the address specified in Section 4, “Information

You might also like