You are on page 1of 53

Anesthesia for Oncological Surgery 1st

Edition Jeffrey Huang


Visit to download the full and correct content document:
https://textbookfull.com/product/anesthesia-for-oncological-surgery-1st-edition-jeffrey-
huang/
More products digital (pdf, epub, mobi) instant
download maybe you interests ...

Imaging in Spine Surgery Jeffrey S. Ross

https://textbookfull.com/product/imaging-in-spine-surgery-
jeffrey-s-ross/

Evidence-Based Practice in Perioperative Cardiac


Anesthesia and Surgery Davy C.H. Cheng

https://textbookfull.com/product/evidence-based-practice-in-
perioperative-cardiac-anesthesia-and-surgery-davy-c-h-cheng/

Reproductive Surgery The Society of Reproductive


Surgeons Manual 1st Edition Jeffrey M. Goldberg

https://textbookfull.com/product/reproductive-surgery-the-
society-of-reproductive-surgeons-manual-1st-edition-jeffrey-m-
goldberg/

Retinal and Optic Nerve Stem Cell Surgery Jeffrey N.


Weiss

https://textbookfull.com/product/retinal-and-optic-nerve-stem-
cell-surgery-jeffrey-n-weiss/
Kaplan’s Cardiac Anesthesia: In Cardiac and Noncardiac
Surgery, 7e 7th Edition Joel A. Kaplan Md

https://textbookfull.com/product/kaplans-cardiac-anesthesia-in-
cardiac-and-noncardiac-surgery-7e-7th-edition-joel-a-kaplan-md/

Anesthesia for Cesarean Section 1st Edition Giorgio


Capogna (Eds.)

https://textbookfull.com/product/anesthesia-for-cesarean-
section-1st-edition-giorgio-capogna-eds/

Skin Cancer Surgery, An Issue of Facial Plastic Surgery


Clinics of North America, Volume 27-1 Jeffrey Moyer

https://textbookfull.com/product/skin-cancer-surgery-an-issue-of-
facial-plastic-surgery-clinics-of-north-america-
volume-27-1-jeffrey-moyer/

Essentials of Anesthesia for Neurotrauma First Edition


Kapoor

https://textbookfull.com/product/essentials-of-anesthesia-for-
neurotrauma-first-edition-kapoor/

Aid Effectiveness for Environmental Sustainability 1st


Edition Yongfu Huang

https://textbookfull.com/product/aid-effectiveness-for-
environmental-sustainability-1st-edition-yongfu-huang/
Jeffrey Huang
Jiapeng Huang
Henry Liu
Editors

Anesthesia
for Oncological
Surgery

123
Anesthesia for Oncological Surgery
Jeffrey Huang • Jiapeng Huang • Henry Liu
Editors

Anesthesia for Oncological


Surgery
Editors
Jeffrey Huang Jiapeng Huang
Department of Anesthesiology Department of Anesthesiology
Moffitt Cancer Center University of Louisville Hospital
Tampa, FL, USA Louisville, KY, USA

Henry Liu
Anesthesiology and Critical Care
University of Pennsylvania
Philadelphia, PA, USA

ISBN 978-3-031-50976-6    ISBN 978-3-031-50977-3 (eBook)


https://doi.org/10.1007/978-3-031-50977-3

© The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Nature Switzerland AG 2023
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 Springer imprint is published by the registered company Springer Nature Switzerland AG
The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland

Paper in this product is recyclable


Foreword

The global burden of new cancer cases reached an alarming 19.3 million in 2020, resulting in
nearly 10 million cancer-related deaths. However, amidst this concerning trend, there is a glim-
mer of progress. The 5-year survival rates for all cancers from 2012 to 2018 have nearly dou-
bled compared to the 1970s, primarily attributed to advancements in early detection and cancer
management.
A pivotal player in this progress is personalized medicine, a groundbreaking approach tai-
loring treatment plans to each patient’s unique genetic makeup, medical conditions, and spe-
cific cancer characteristics.
An often overlooked but crucial aspect of cancer care is anesthesia. Whether during diag-
nostic, therapeutic, or palliative procedures, personalized anesthesia care is vital, considering
factors such as comorbidities, medications, and treatment history. The success of oncological
surgeries hinges on well-managed anesthesia, contributing to improved recovery, fewer com-
plications, and better patient outcomes.
Given the dynamic evolution of cancer treatment approaches, surgical techniques, and med-
ical therapies, dedicated textbooks in this area become imperative. “Anesthesia for Oncological
Surgery,” edited by Professors Jeffrey Huang, Jiapeng Huang, and Henry Liu, emerges as a
standout resource. This concise yet comprehensive guide addresses the unique challenges of
anesthesia in cancer surgery, often overlooked by general anesthesia textbooks.
This textbook transcends the role of a mere manual for perioperative cancer patient care. It
delves into crucial aspects such as disease epidemiology, survival rates, pathology, risk factors,
screening, diagnosis, and latest treatment approaches for various cancer types across all
systems.
Central to the book is its exploration of how diseases and cancer treatments impact anes-
thetic planning. It provides detailed insights into preoperative evaluation, intraoperative man-
agement, and postoperative care, empowering anesthesia providers to make informed decisions
throughout the oncological surgical journey.
“Anesthesia for Oncological Surgery” serves a dual purpose, functioning as both a compre-
hensive textbook and a quick reference guide. Its versatility makes it an indispensable compan-
ion for anesthesia professionals, not just in the operating room but also in their ongoing
practice.
This book caters to anesthesia professionals of all experiences, offering an opportunity to
deepen their knowledge, enhance patient outcomes, and elevate the overall quality of practice
in the realm of oncological surgery.

Department of Anesthesiology and Perioperative Medicine Carin A. Hagberg


The University of Texas MD Anderson Cancer Center
Houston, TX, USA
Department of Anesthesiology
The University of Texas UTHealth McGovern Medical School
Houston, TX, USA

v
Preface

The steadily increasing prevalence of cancer patients in recent decades has subsequently led to
ever-increasing demands for more oncological surgical interventions, and more and more dedi-
cated oncological surgeons practice not only in major university/tertiary medical centers but
also in community hospitals. With the significant rise in the volume and the ever-expanding
range of oncological procedures, anesthesia care for these oncological procedures can be very
challenging; all these call for a reference book which addresses these challenges faced by
anesthesia providers for these oncological surgeries.
The book should cover all organ systems and all oncological procedures, especially those
newer and unique oncological procedures unfamiliar to anesthesia providers. The need for an
oncological anesthesia-focused reference book and the scarcity of such books led us to the
decision to edit this book entitled Anesthesia for Oncological Surgery. Recognizing that mod-
ern anesthesia care, especially for those newer oncological procedures, demands thorough,
updated medical and anesthetic knowledge to ascertain the quality of care for cancer patients
undergoing surgical interventions, because oncological surgical procedures are usually com-
plex and often involve diverse patient needs, which sometimes go beyond anesthesia practices
per se. Furthermore, cancer management can be very dynamic, medical interventions as che-
motherapy, radiation therapy, and immunotherapy can be integrated and interposed. Thus, it is
crucial for anesthesia providers to have a thorough understanding of dynamic oncological
management and its impact on anesthesia care. Adequate preoperative assessment, physical
and mental preparations, effective and appropriate intraoperative monitoring, timely manage-
ment of all potential adverse events, and satisfactory postoperative analgesia are all issues that
anesthesia providers will have to deal with. This book will focus on all these important issues.
Anesthesia for Oncological Surgery will serve as a comprehensive reference book and also
as a handy practice guide. The book intends to offer insights into anesthesia care specifically
for cancer patients, covering preoperative assessment, intraoperative management, and postop-
erative care. Each chapter provides pertinent oncological information, latest literature updates,
and evidence-based recommendations to enhance anesthesia providers’ understanding of the
unique challenges posed by oncological surgery.
We believe this book Anesthesia for Oncological Surgery will significantly contribute to the
knowledge in anesthesia care for oncological patients, enhance the expertise of anesthesia
providers, and ultimately improve the care delivery and clinical outcomes of perioperative
oncological patients.

Tampa, FL, USA Jeffrey Huang


Louisville, KY, USA Jiapeng Huang
Philadelphia, PA, USA Henry Liu

vii
Acknowledgments

I would like to express my gratitude to my wife, Frances Wu and my children Evan Huang and
Alexis Huang. Their unwavering support and belief, in me have been the foundation of my
accomplishments. I am also incredibly grateful for the colleagues I have had the privilege of
working with at Moffitt Cancer Center. The collaborative spirit within our hospital has fostered
an environment of innovation, research and exceptional care in the field of anesthesia and sur-
gery. It is my hope that this book will contribute to advancements, in onco-anesthesiology
through collaboration and the collective efforts of all the colleagues who have contributed to
its book.
Jeffrey Huang, MD, FASA

ix
Contents

Part I Basic Science of Oncology

Oncogenesis, What Is New?�����������������������������������������������������������������������������������������������   3


Humberto Trejo Bittar
Epidemiology of Cancer����������������������������������������������������������������������������������������������������� 11
Hui-Yi Lin and Jong Y. Park
Immunemodulation and Cancer��������������������������������������������������������������������������������������� 17
Jinhong Liu and Jeffrey Huang
Cancer Targeted Molecular Therapy ������������������������������������������������������������������������������� 27
Jinhong Liu
CAR T-Cell Therapy����������������������������������������������������������������������������������������������������������� 35
Vivian M. Irizarry Gatell, Jeffrey Huang, and Omar A. Castaneda Puglianini

Part II Perioperative Oncology


The Surgeon: Anesthesia Professional Relationship������������������������������������������������������� 47
Alexander S. Rosemurgy, Sharona B. Ross, and Iswanto Sucandy

Anesthetic Technique and Cancer Recurrence ��������������������������������������������������������������� 51
William M. Fowler, Kevin Thai, Thomas M. Kane, and John A. Hodgson
Prehabilitation��������������������������������������������������������������������������������������������������������������������� 57
Relin Yang, Troy Rush, and Charles Huang

Preoperative Evaluation and Preparations for Oncological Surgery���������������������������� 65
Julia K. Labovsky

Perioperative Surgical Home, Enhanced Recovery After Surgery,
and High-Risk Committee������������������������������������������������������������������������������������������������� 71
Kathleen J. Lee, Timothy D. Quinn, and Raymond Sroka

Blood Management for Oncological Surgery������������������������������������������������������������������� 79
Juan P. Cata

Acute Pain Service for Oncological Surgery ������������������������������������������������������������������� 87
Stephania Paredes Padilla, Chelsea Skinner, Sydney L. Keller, Surendrasingh
Chhabada, Ryu Komatsu, and Jijun Xu

Chronic Pain Clinic in Cancer Center and Oncological Services ��������������������������������� 101
Jessica Ibañez

Non pharmaceutics Therapy for Oncological Patients��������������������������������������������������� 111
Sahana Rajasekhara, Kristine A. Donovan, and Lora M. A. Thompson

xi
xii Contents

Part III Oncological Neurosurgery

Craniotomy for Meningioma��������������������������������������������������������������������������������������������� 121


Mian Shen
Craniotomy for Glioma ����������������������������������������������������������������������������������������������������� 125
Jerrad R. Businger and Brian J. Williams
Craniotomy for Brainstem Tumors����������������������������������������������������������������������������������� 129
Raja Jani, Aneeta Bhatia, Ajmal Zemmar, Akshitkumar Mistry, and Brian J.
Williams
Awake Craniotomy������������������������������������������������������������������������������������������������������������� 137
Maria Birzescu

Resection of Pituitary Gland Tumor��������������������������������������������������������������������������������� 143
Raja Jani, Brian J. Williams, Marina Varbanova, and Alexander Bautista
Posterior Fossa Tumor Resection ������������������������������������������������������������������������������������� 149
Matthew Protas, Satish Krishnamurthy, Fenghua Li, and Reza Gorji

Cervical Spine Cancer Surgery����������������������������������������������������������������������������������������� 157
Brianna Johnson, Nazar Dubchak, and Callum Dewar

Thoracic Spine Malignancy Surgery��������������������������������������������������������������������������������� 161
Daniel Haines and Bryant M. England

Lumbar Spine Surgery (Tumors in The Lumbar Skeletal Systems and Muscles)������� 165
Shawn W. Adams, Brian J. Williams, Carlos Perez Ruiz, and Alexander Bautista

Surgery for Spinal Cord Tumors��������������������������������������������������������������������������������������� 169
Jeremy Crane and Justin Zeien

Part IV Head and Neck Oncological Surgery


Oral Cavity, Larynx, and Tonsil Cancer Surgery ����������������������������������������������������������� 177
Melanie Townsend, Ramesh Mariyappa, and Emma C. Huang

Parotid Gland Cancer Surgery����������������������������������������������������������������������������������������� 183
Diana Hamann
Laryngeal Cancer Surgery������������������������������������������������������������������������������������������������� 187
James Miranda, S. Nini Malayaman, Joshua H. Atkins, and Henry Liu

Tracheostomy in Cancer Patients������������������������������������������������������������������������������������� 193
Kate Williams and Madeleine Strohl

Thyroid and Parathyroid Cancer Surgery����������������������������������������������������������������������� 199
Lin Tang and Samira M. Sadowski

Neck Dissection and Reconstruction��������������������������������������������������������������������������������� 207
Joshua Read and Brielle Klein

Part V Thoracic Oncological Surgery


Lobectomy for Lung Cancer��������������������������������������������������������������������������������������������� 215
William E. Rallya, Christopher Russo, and John Hodgson

Surgery for Pleural Malignancies������������������������������������������������������������������������������������� 221
Sandra M. Orfgen
Contents xiii


Surgery for Cardiac Malignancies ����������������������������������������������������������������������������������� 225
Tianyu Jiang and Jeffrey Huang

Surgery for Mediastinal Cancer ��������������������������������������������������������������������������������������� 229
Muhammad F. Sarwar, Jason M. Wallen, and Henry Liu
Surgery for Tracheal Cancer��������������������������������������������������������������������������������������������� 233
Melissa A. Burger

Surgery for Mainstem Bronchial Cancer������������������������������������������������������������������������� 241
Melissa A. Burger

Part VI Gastrointestinal Cancer Surgery

Esophageal Cancer Surgery����������������������������������������������������������������������������������������������� 251


Rana K. Latif, Prejesh Philips, Zachary J. Senders, and Sean P. Clifford
Gastric Cancer Surgery����������������������������������������������������������������������������������������������������� 257
Amber F. Gallanis, Andrew J. Mannes, and Jeremy L. Davis
Liver Cancer Surgery��������������������������������������������������������������������������������������������������������� 263
Michael Leclerc, Sean Stokes, Daniel Saenz Anaya, and Jeffrey Huang
Pancreatic Cancer��������������������������������������������������������������������������������������������������������������� 269
Jeffrey Huang
Colon/Rectal Cancer Surgery ������������������������������������������������������������������������������������������� 273
Brendan L. Hagerty, Anthony Dakwar, and Kathleen J. Lee
HIPEC��������������������������������������������������������������������������������������������������������������������������������� 279
Shadin Ghabra, Andrew M. Blakely, Andrew Mannes, and Ning Miao

Biliary System Cancer Surgery����������������������������������������������������������������������������������������� 287
Jeffrey Huang, Benjamin Powers, and Renee Mapes
Appendiceal Cancer Surgery��������������������������������������������������������������������������������������������� 293
Vicente Ramos-Santillan, Gary Mann, and Timothy Quinn
Retroperitoneal Tumor Surgery ��������������������������������������������������������������������������������������� 297
Korina E. Sandoval and Richard L. Burgan

Part VII Urological Oncology

Renal Cancer Surgery ������������������������������������������������������������������������������������������������������� 303


Ryan Grell and Mohammed Said
Bladder Cancer Surgery����������������������������������������������������������������������������������������������������� 307
Daisy Sangroula, Kellen B. Choi, and Sean P. Clifford
Prostate Cancer Surgery ��������������������������������������������������������������������������������������������������� 313
Daniel Nethala and Andrew J. Mannes
Urothelial Cancer Surgery������������������������������������������������������������������������������������������������� 317
Mark M. Hanna, Taylor Peak, Herney Andrés García-­Perdomo, Gagan Prakash,
Andrea Necchi, and Philippe E. Spiess
Testicular/Penile Cancer Surgery������������������������������������������������������������������������������������� 323
Tianyu Jiang, Taylor Peak, Philippe Spiess, and Jeffrey Huang
xiv Contents

Part VIII Endocrine and Metabolic Oncological Surgery

Surgery for Adrenal Tumors ��������������������������������������������������������������������������������������������� 331


Shadin Ghabra, Kenneth Luberice, Naris Nilubol, Andrew Mannes, and Xiaowei Lu

Surgery for Carcinoid Syndrome ������������������������������������������������������������������������������������� 341
Shadin Ghabra, Tracey Pu, Naris Nilubol, Andrew Mannes, and Ning Miao
Neuroendocrine/CREST Cancer Surgery ����������������������������������������������������������������������� 349
Andrew C. Baek, Kenny Wise, and Emanuela C. Peshel

Part IX Gynecological Cancer Surgery

Ovarian Cancer Surgery ��������������������������������������������������������������������������������������������������� 357


Brittany Maggard, Sarah Todd, Faizan Ahmed, Sean Clifford, Jiapeng Huang,
and Rana Latif
Uterine Cancer Surgery����������������������������������������������������������������������������������������������������� 363
Monica Avila and Rohini Kotha
Cervical Cancer Surgery ��������������������������������������������������������������������������������������������������� 369
Allyn O. Toles, Briana Rice, Jordyn Tumas, and Henry Liu
Perineal Cancer Surgery ��������������������������������������������������������������������������������������������������� 373
Andrewston Ting, Monica Avila, and Jeffrey Huang

Part X Surgery for Skeletal and Muscular Malignancies


Surgery for Bone Sarcoma������������������������������������������������������������������������������������������������� 381
Raymond Evans, Andrew Serdiuk, Douglas Letson, and Jeffrey Huang
Surgery for Rhabdomyosarcoma��������������������������������������������������������������������������������������� 387
Jamie Hoffman, Rachel Voss, and Jeffrey Huang

Part XI Surgery for Breast Cancer and Cutaneous Cancer


Surgery for Breast Cancer������������������������������������������������������������������������������������������������� 395
Cindy B. Yeoh, Kelly Elleson, Todd Schultz, Brielle Weinstein, Nicholas Panetta,
and Marie Catherine Lee
Cutaneous Cancer Surgery ����������������������������������������������������������������������������������������������� 403
Matthew Benesch, Julia Faller, and Joseph Skitzki

Part XII Pediatric Cancer Surgery


Anesthesia for Pediatric Procedures Outside of the Operating Room��������������������������� 411
Ashley Bocanegra and Christopher Setiawan
Wilms Tumor and Hepatoblastoma����������������������������������������������������������������������������������� 417
Alex Y. Chung
Pheochromocytoma������������������������������������������������������������������������������������������������������������� 423
Neethu Chandran
Medulloblastoma����������������������������������������������������������������������������������������������������������������� 427
John Zhong
Pilocytic Astrocytoma��������������������������������������������������������������������������������������������������������� 431
John Zhong
Contents xv

Part XIII Other Oncological Surgical Procedures


Intra-arterial Therapy for Primary and Secondary Liver Cancer ������������������������������� 437
Hakob Kocharyan, Altan Ahmed, and Nainesh Parikh

Percutaneous Ablative Techniques for Liver and Kidney Cancer��������������������������������� 441
Altan F. Ahmed, Hakob Kocharyan, Andrei Lojec, Kenny Le, and Nainesh Parikh

Interventional Diagnostic and Therapeutic Procedures in Surgical Oncology������������� 447
Kara M. Barnett, Victoria Brennan, Suken H. Shah, Elizabeth F. Rieth,
and Marisa A. Kollmeier

Intensive Care of Cancer Patients������������������������������������������������������������������������������������� 457
Aditi Balakrishna, Daniel Nahrwold, and Christopher Hughes

Palliative Care Surgery of Cancer Patients ��������������������������������������������������������������������� 471
Zhaosheng Jin, Vincent Bargnes, Alexandra Tsivitis, Jonathan B. Oster, and Jun Lin

Hospice Care and Palliative Care in Cancer Patients����������������������������������������������������� 477
Hui Liu, Lin Chen, Lauren Hollifield, James E. Miranda, Brian Entler,
Nini Malayaman, and Henry Liu
Index������������������������������������������������������������������������������������������������������������������������������������� 483
Part I
Basic Science of Oncology

Huang Jeffrey
Oncogenesis, What Is New?

Humberto Trejo Bittar

Nothing has revolutionized the field of cancer treatment in growth factor receptor), HER2, among many others), tumor
recent years more than the establishment of personalized suppressor genes, those that inhibit cell growth and their
medicine. Personalized cancer treatment uses therapies/ loss of function allows uncontrolled cell growth (p53 the
treatments targeted to the patient’s tumor profile, intending most commonly mutated gene in cancers and RB, etc.),
to provide a more precise and targeted therapy than conven- apoptotic genes, those regulate apoptosis promoting cancer
tional chemotherapy, particularly in advanced/metastatic cells survival (the BCL2 family of genes), and lastly genes
cancers. Pathologic assessment is an essential component of that affect the interactions between cancer cells and host
personalized cancer treatment by providing accurate cancer cells (checkpoint inhibitors genes would be in this category).
diagnoses and proper testing of tumors for those specific All the gene alterations act together to allow the cancer cells
molecular and immunohistochemical biomarkers used to to proliferate without the need for growth signals and avoid
guide targeted treatment strategies. To design targeted thera- inhibitory/apoptotic signals, to have altered cell metabolism
pies, one needs to understand the pathophysiology behind that supports continued cell growth, to guarantee blood sup-
oncogenesis. While a detailed review of oncogenesis ply, to evade the host immune system and to allow for inva-
deserves its book, in this book chapter, I focus on the impor- sion and metastasis, essentially becoming immortal [1].
tant issues of oncogenesis and personalized cancer treat- Some significant genetic changes/mutations in cancer
ment, using lung cancer as an example as I am a thoracic include point mutations, a change in a single nucleotide in
pathologist with minor references to the generalities of a gene sequence, for example, in the KRAS gene in lung
oncogenesis. adenocarcinomas. Gene rearrangements change the struc-
ture of a chromosome(s) by inversion or translocation, as it
occurs in ALK translocated lung adenocarcinomas. Gene
1 Oncogenesis and Personalized Cancer deletions, in which a portion of the chromosome is lost,
Treatment often affect tumor suppressor genes. Gene amplifications,
meaning producing multiple copies of the same gene with
Oncogenesis is a very complex and multifactorial process overexpression and activation, can be seen with HER2
related to not only the more well-known oncogenes and amplification in breast cancer. Aneuploidy is when a cancer
tumor suppressor genes but also the effect that the environ- cell has more than the standard number of chromosomes
ment has on them and the overall physiology of the normal and thus copies numbers of oncogenes. microRNAs are
cells and heritable conditions (not discussed in this chapter). small single-stranded RNA fragments that can bind to mes-
Genes are the center of oncogenesis, and simply said, the senger RNA after transcription and facilitate its degrada-
origin of cancer is related to genetic and epigenetic changes tion. This process specifically targets tumor suppressor
and mutations that alter the function of genes. There are genes, leading to downregulation of the corresponding pro-
oncogenes, those that promote cancer cells growth (RAS is teins and potentially enabling uncontrolled cell prolifera-
the most commonly mutated oncogene, EGFR (Epidermal tion, as observed in lung cancer cases [2].
Notably, a single mutation/genetic change cannot cause
the transformation of normal cells to cancer cells. Instead, in
H. T. Bittar (*) almost all cancers, multiple mutations accumulate (double/
Department of Pathology, Moffitt Cancer Center, Tampa, FL, USA
multiple hit hypothesis) in a single cell to transform it. It is
Department of Oncologic Sciences, University of South Florida, also essential to recognize that while theoretically tumors are
Tampa, FL, USA
made of proliferation from a single mutated cell
e-mail: Humberto.TrejoBittar@moffitt.org

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 3


J. Huang et al. (eds.), Anesthesia for Oncological Surgery, https://doi.org/10.1007/978-3-031-50977-3_1
4 H. T. Bittar

(­ monoclonality), in reality, tumors are heterogenous in their genes). There is currently no role for biomarkers testing in
cell composition. This is because, within the life of a tumor, small cell carcinoma.
new mutations can occur (because of the genetic instability
present in tumors), leading to new subclones of tumor cells,
which are often selected to proliferate better and become 2 The Increasing Role
resistant to treatments. This is the origin of tumor progres- of Immunotherapy
sion and the development of treatment resistance.
Changes in the integrity of the end of chromosomes, so-­ Immunotherapy in cancer treatment refers to using medica-
called telomeres that protect chromosomes from degrada- tions to improve the person’s immune system’s capacity to
tion, are a standard component of aging and have also been attack neoplastic cells.
implicated in the development of many conditions, including Immune checkpoint is a normal physiologic phenomenon
cancers. The length of telomeres is maintained by the tran- aimed to prevent the building of immune responses against
scription enzyme complex telomerase (some of its vital com- healthy/non-neoplastic cells. Generally speaking, it prevents
ponents are TERT (telomerase reverse transcriptase) and T-cells from activating and promoting cell death. In the cen-
TERC (telomerase RNA component)). Shortening of telo- ter of these complicated pathways are the proteins program
meres is a normal phenomenon that occurs with each cell death-1 (PD-1) and the program death-1 ligand (PD-L1).
cycle (prevented by the telomerase) or an abnormal one due PD-L1 is a transmembrane protein that downregulates
to excess reactive oxygen species (or lack of antioxidants). immune responses by binding to its two inhibitory PD-1 and
When telomeres shorten beyond a critical point, the cell B7–1 (CD80). PD-1 is an inhibitory receptor expressed on
becomes senescent and undergoes apoptosis/cell death with T-cells following their activation, which is sustained in states
the associated loss of tissue function (as it occurs in normal of chronic stimulation such as in chronic infection or cancer.
aging). To become immortal, an important step in tumori- The binding of PD-L1 with PD-1 inhibits T-cell prolifera-
genesis, cancer cells must irreversibly prevent the shortening tion, cytokine production, and cytolytic activity, leading to
of telomeres, most often by constitutively activating telomer- the functional inactivation or exhaustion of T cells. PD-L1
ase (frequently by amplification and mutations in the pro- expression has been observed in immune cells and tumor
moter regions of the TERT and TERC genes), thus avoiding cells. Aberrant expression of PD-L1 on tumor cells has been
the DNA damage cell death pathways [3]. reported to impede anti-tumor immunity, resulting in immune
The recent advances in lung cancer treatment are a perfect evasion. Therefore, interruption of the PD-L1/PD-1 pathway
example of successful personalized cancer treatment, par- represents a novel strategy to reactivate the tumor-specific
ticularly in lung adenocarcinomas, for which potentially tar- T-cell mediated immunity suppressed by the expression of
getable oncogenic mutations are present in over 75% of PD-L1 in the tumor microenvironment.
cases. In the United States, KRAS is the most frequently PD-L1/PD-1 inhibitors (collectively called immune
mutated oncogene in lung adenocarcinoma, followed by checkpoint inhibitors) are nowadays the cornerstone of the
EGFR [4, 5]. EGFR (upstream tyrosine kinase receptor) and treatment of many cancer types. PD-L1 is expressed in a
KRAS (downstream signaling molecule) are part of the broad range of cancer cells, including lung, melanoma, uro-
MAP-kinase pathway that regulates cell growth, prolifera- thelial, kidney, ovarian, breast, and colorectal cancer [8]. For
tion, and survival. Not surprisingly, most of the targeted many of the FDA-approved indications, immune checkpoint
therapies available for lung adenocarcinomas affect this inhibitors have proven to be a superior therapy with or with-
pathway. There are multiple guidelines [6] aligning with the out concurrent chemoradiation (depending on the cancer
current state of drugs approved by the U.S. Food and Drug stage and history of prior therapies) compared to chemora-
Administration (FDA), helping clinicians in their choice of diation alone [9].
biomarkers testing (see below) and targeted treatment strate- It is impossible to discuss immunotherapy without incor-
gies. Some recommendations include molecular testing of porating T-cell transfer therapies. The main principle is
nonsquamous lung non-small cell carcinomas for KRAS, removing the patient’s T-cells and, in the laboratory, select-
EGFR, HER2, and BRAF mutations; ALK, ROS1, RET, ing and conditioning them to better attack the patient’s can-
NTRK translocations, MET exon 14 alterations, and of cer cells [10]. There are two main types of T-cells transfer
course, PD-L1 status. In squamous cell carcinomas, the most therapies.
common genetic changes include those in p53, PI3KCA,
SOX2, and FGFR1. There is currently no FDA-approved tar- • Tumor-infiltrating lymphocytes (TIL) therapy uses the
geted therapy (or need for biomarkers testing) in these lymphocytes that are enriched on the patient’s tumor
tumors [7] unless the patient is young and has never been a (requires resection of the tumor to obtain the TILs) but
smoker. Lastly, for small cell carcinomas, the most common that is not sufficient or activated enough to destroy the
(essentially all cases) genetic alterations involve downregu- tumor, and in the laboratory, select and expand those
lating of p53 and Rb proteins (by the genetic loss of their T-lymphocytes that best recognize the tumor cells (refer
Oncogenesis, What Is New? 5

to as TIL products). The TIL product is then infused back immunostain is finally evaluated under the microscope for
into the patient with the improved capacity to attack the the presence or absence and the semiquantitative estimation
tumor cells. TIL therapy has been used to treat melanoma of the targeted protein.
[11] and is currently being studied and used experimen- Important examples of the use of immunohistochemistry
tally to treat other solid tumors like cervical squamous include the assessment of PD-L1 expression in many cancer
cell carcinoma, cholangiocarcinoma, and lung carcino- types. For instance, hormone receptors status in breast can-
mas [12]. cer, mismatch repair enzymes expression status in colon and
• The other technique is CAR-T cell therapy, which uses endometrial cancer, and ALK expression in ALK translo-
the patient’s T-lymphocytes after being genetically modi- cated lung adenocarcinomas, among many others. It is
fied in the lab to express a chimeric antigen receptor important to know that there are certain preanalytical require-
(CAR) that is a modified receptor specifically designed to ments that must be met for the immunostains to be properly
attach to the patient’s tumor cells. CAR-T cell therapy is interpreted. Many of these requirements are regulated by the
FDA-approved to treat many advanced forms of leuke- Food and Drug Administration (FDA) and many pathology
mias and lymphomas [13]. While very promising, CAR-T and oncology professional organizations. Some examples of
cell therapy is not free of secondary effects and is notori- these requirements include:
ously very costly.
• For PD-L1 expression in lung non-small cell carcinoma,
at least 100 viable tumor cells must be present on the
slide, and tissue should not be over 3 years old. This can
3 The Importance of Tissue Adequacy be easily obtained by small biopsies and cytology speci-
in Biomarker/Ancillary Studies mens [6, 14–16]. Following testing guidelines is
Testing extremely important to avoid misclassifying patients
who could have potentially benefited from
Biomarkers/prognostics testing in the personalized treatment immunotherapies.
of cancer can be done by multiple techniques (see Table 1) • For hormone receptors (estrogen, progesterone, and
with different degrees of complexity. The easiest and quick- HER2) expression status in breast cancer, we have
est way of testing is by immunohistochemistry. In this tech- some of the strictest guidelines (by the College of
nique, biopsy tissue or cells collected by cytology techniques, American Pathologists (CAP) and the American
like fine needle aspiration, are exposed to antibodies that rec- Association of Clinical Oncology (ASCO)) and stan-
ognize the targeted protein (so-called primary antibody). The dard operating procedures, and their mandatory appli-
expression of the target protein is then demonstrated by cation is tightly regulated. Some of the
using a secondary antibody against the immunoglobulin recommendations include short ischemic time (the
used as the primary antibody. A distinct color (usually time between removal of the tissue/stopping of the
brown) is produced because the secondary antibody is bound blood supply and the beginning of 10% formalin fixa-
to a reagent that produces color after a specific chemical tion), the fixation time should be at least 6 h, and no
reaction (so-called detection agent). The final product is a more than 72 h, and slides should not be older than
section of tumor stained (because of the presence of color) 6 weeks, mandatory participation in external profi-
with the targeted protein, which we call immunostain. The ciency testing, among many others [17].

Table 1 Tissue sampling for personalized cancer treatment


Sample procedure Samples and tests Important factors
• Surgical resection • Fresh tissue • Preoperative evaluation and need for anesthesia
• Surgical biopsy • Formalin-fixed, paraffin-­embedded (FFPE) • Choice of sampling method (invasive vs.
• Cytology specimen • Smears non-invasive)
(aspirate or fluid) • Liquid biopsies • Need for intraoperative or intraprocedural sample
• Non-­invasive procedures: • Diagnostic evaluation (histology or cytology) adequacy assessment
  – Blood • Protein expression: • Limitations of the length of the ischemic time
  – Urine   – Immunohistochemistry • Assessment of adequacy before sequencing
• Genetic tests: procedures (is there enough material after
   – Conventional PCR diagnosis and further processing)
  – FISH • Is there a good proportion of tumors within the
   – Next-generation sequencing (NGS) sample
   – RT-qPCR/gene expression panels/ • Avoid using old samples
RNA-sequencing • Need for biostatistics and data analysis of NGS
data
6 H. T. Bittar

The use of immunohistochemistry for the assessment of dye to the complementary and highly specific DNA sequence
prognostic biomarkers has many benefits, including shorter of the target gene. Using fluorescent microscopy, the loca-
turn-around time with results being available as early as tion of the probes on chromosomes can be visualized in the
within 24 h, their more cost-effective and less of a financial individual cancer cell using formalin fixed tissue sections.
burden for patients and the health system (compared to the By consensus, at least 50 well-visualized tumor calls must be
more expensive sequencing studies), and their easier to per- present on the slide [6, 22] to be an adequate sample.
form, more widely available and, in many cases, more Immunohistochemistry, a more cost-effective, faster, and
straightforward to interpret [18]. less technically demanding technique, is gradually replacing
the use of FISH in evaluating ALK rearrangement in lung
carcinomas. Confirmation by FISH studies is still necessary
3.1 Sequencing in unequivocal cases [23].

Sequencing of DNA (determining the exact sequence of


nucleotides/bases) and fluorescent in-situ hybridization stud- 3.3 RNA Gene Expression
ies (FISH) are extensively used to select patients for targeted
therapy and biomarker testing. As mentioned, these tech- The most recent molecular technique being used is the RNA
niques are used to find those specific genetic changes in the gene expression panels. Simply said, this technique uses
cancer cell’s genome. RNA instead of DNA as the input to identify gene altera-
tions. The benefit of this is that RNA detection is more sensi-
• The most effective technique for sequencing the tumor tive than DNA by detecting known and novel fusions/
genome is next-generation sequencing (NGS). While the alterations. It measures expressed genes rather than predicted
specifics of this technique are out of the scope of this gene expression, which aligns better with the true pathologic
chapter, the concept is that the whole genome or targeted drivers of the disease. When done correctly, they have better
parts of it are sequenced in parallel by millions of indi- sample yield and are more cost-effective [24, 25].
vidual smaller fragments of genetic material multiple
times (something referred to as depth of the sequencing).
Using bioinformatics tools, the fragments are pieced 3.4 Liquid Biopsies
together, mapping each fragment to the expected normal
genome, allowing by comparison to find those single Clinicians must also be aware of the usefulness of the so-­
nucleotide mutations or much more significant changes in called “liquid biopsies.” Essentially, the principle behind this
individual genes or whole chromosomes. Tumor sequenc- technique is that tumor cells can shed their genetic material
ing is one of the necessary steps for most personalized into the bloodstream. Thus, we can measure the presence of
cancer treatments and often an indication for biopsy pro- that material (providing diagnosis) and its genetic sequence
cedures, particularly in the recurrence setting. (providing personalized treatment options information)
using a simple blood sample. Essentially, replacing the need
It is important to adequately obtain these tissue samples. for tissue, which might be important in patients with poor
Recent recommendations have suggested at least 1 mm2 of performance status for whom undergoing anesthesia would
formalin—fixed-paraffin-embedded tissue, needing as little be contraindicated. Other benefits of liquid biopsy include
as just five sections that must contain at least 20% of viable early cancer detention (when tumors are too small to be seen
tumor [6, 19]. It is imperative to test the adequacy, which is by imaging studies or to obtain a tissue sample), assessment
done by rapid on-site evaluation by cytologists in procedures of treatment response, and easy monitoring of recurrence
like fine needle aspirates or by frozen section examination and development of treatment resistance. Despite this, tissue
[20, 21]. The overall goal is to ensure enough and proper sampling continues to be the preferred method for diagnosis
material has been collected to avoid unnecessary repeated and molecular testing, and is considered the gold standard to
procedures, which will necessitate further anesthesia. this day [26, 27].

3.2 Fluorescent In-Situ Hybridization (FISH) 4 The Environment, Chemicals,


and Cancer
Another molecular technique used in personalized cancer
treatment is fluorescent in-situ hybridization (FISH). In this It is widely accepted that gene expression is affected by the
technique, large gene/chromosome changes are identified by environment and that different environmental conditions par-
hybridizing (binding of complementary DNA sequences) ticipate as risk factors for many cancers. This is supported by
small fragments of DNA that are tagged with a fluorescent the fact that there are regional differences in cancer types and
Oncogenesis, What Is New? 7

incidences. For example, liver cancer is more prominent in 5 Microbial Oncogenesis


East Asia, which is related to the presence of liver flukes; or
the higher incidence of certain types of cancers associated There are a few oncogenic microorganisms, including
with obesity, alcohol consumption, the Western diet, and Helicobacter pylori, and DNA and RNA viruses. Altogether
developed nations. Some of these exposures include chemi- they are believed to be responsible for approximately 15% of
cal carcinogens, ultraviolet light, tobacco smoke, and micro- cancers worldwide. Some effects on oncogenesis include
organisms (see below), among many others, all of which stimulation of cell proliferation, enhancement of cell sur-
share a common potential DNA damaging effect. It has been vival, and interference with cell cycle regulation. Some nota-
proposed that up to 85% of cancers could be prevented by ble examples include:
modifying these genes-environment interactions [28].
Cigarette smoke contains over 60 chemical carcinogens, • HPV in oropharyngeal and cervical cancer via oncogenic
including nicotine, ammonia, carbon monoxide and diox- proteins that inactivate RB and p53. The recent wide-
ide, tar, formaldehyde, acetone, and cadmium) is associated spread use of HPV vaccines is an extraordinary example
with an increased risk of many types of cancers. While lung of cancer prevention via vaccination [30, 31].
cancer is the most known association, smoking is also • Merkel cell polyomavirus, via viral integration and
linked to cancers of the head and neck, esophagus, pan- expression of the viral LT and ST proteins and thus RB
creas, bladder, and liver, among others. The effect of ciga- inhibition. More recently, immunotherapy has been
rette smoking is mainly at the level of the DNA, where it proven to be an evolving option for the treatment of this
induces distinct signature mutations (e.g., C>A transver- malignant neoplasm [32].
sions), indirect activation of DNA editing, and DNA meth- • Hepatitis B and C virus infection are one of the main etio-
ylation, helping in increasing the risk of acquiring cancer logic factors in hepatocellular carcinoma [33, 34]. There
driver mutations. Moreover, the direct effect of smoking are many ways these viruses can be oncogenic, including
inducing epithelial damage (e.g., precursor lesions of can- the promotion of chronic inflammation, dysregulation of
cer like squamous metaplasia), chronic inflammation, and cytotoxic CD8 T-cells, stimulation of hepatocytes prolif-
inhibiting apoptosis support the survival of cancer cells. eration, and increased reactive oxygen species, all leading
Outside of carcinogenesis, cigarette smoke also induces to DNA damage. There are also direct effects of viral pro-
immunosuppression, disbalances in proteases and antipro- teins (e.g., HBx and HVC core protein) on p53 function
teases, and decreases phagocytic activity, leading to many by activating transcription factors.
other diseases, including chronic obstructive pulmonary • Epstein-Barr Virus (EBV) is a human oncogenic virus
disease (COPD) [29]. that can be lifelong asymptomatic but also associated
Certain occupations and their associated exposures are with many diseases. Some of these are non-neoplastic,
known to have links with cancer. These include asbestos and including Burkitt lymphoma, some Hodgkin lymphomas,
mesothelioma, benzene and acute myeloid leukemia, beryl- nasopharyngeal carcinomas, some gastric carcinomas,
lium and lung cancer, cadmium and prostate cancer, vinyl and rare sarcomas. The exact mechanisms of oncogenesis
chloride and angiosarcoma, radon and lung cancer, among linked to EBV remain a research challenge; at least in
others. part, they are related to dysregulated normal B-cells pro-
There are many chemical carcinogens. Some act directly liferation [35].
and do not need to be metabolized. A typical example is • Helicobacter pylori, best known for its role in the patho-
alkylating chemicals, as they are used in some chemotherapy genesis of gastric peptic ulcers, is also the first bacteria to
regimens that can lead to secondary malignancies. Others be recognized as having a role in oncogenesis. Like the
need to be metabolized. Not surprisingly, some include hepatitis viruses, the bacteria, by creating a chronic
chemicals formed after the combustion of tobacco and fossil inflammatory state in the gastric mucosa, leading to atro-
fuels, those found in aromatic amines (used in the dye and phy and intestinal metaplasia, increases the risk of gastric
rubber industry), and in natural sources like aflatoxins in adenocarcinoma and lymphoma. Also, direct effects of
plants). These agents are mutagenic and directly bind to helicobacter pylori genes have been implicated (e.g.,
DNA leading to oncogenic mutations. A last mention should CagA and VacA genes), which can upregulate growth fac-
be given to the role of ionizing and ultraviolet radiations, tors [36, 37].
which are also mutagenic, including their effect on chromo-
some morphology by breaking the DNA, the excessive for- The overall organ microbiome could also participate in
mation of pyrimidine dimers overwhelming the DNA repair tumorigenesis. It has been recently described that gut and
mechanisms, and point mutations. lung microbiomes can predict response to immune therapies
8 H. T. Bittar

(e.g., anti PD-L1) by inducing immune dysregulation and small cell lung cancer and their prognostic significance according
to clinicopathological factors and diagnostic markers. Int J Mol Sci.
tolerance and directly increasing the risk of malignancy due
2019;20(4):824.
to chronic inflammation [38]. 9. Onoi K, Chihara Y, Uchino J, Shimamoto T, Morimoto Y, Iwasaku
Additional attention is now paid to the tumor ecosystem M, et al. Immune checkpoint inhibitors for lung cancer treatment: a
and microenvironment, a field that was until recently over- review. J Clin Med. 2020;9(5):1362.
10. Diaz-Cano I, Paz-Ares L, Otano I. Adoptive tumor infiltrating lym-
looked. The tumor microenvironment includes endothelial
phocyte transfer as personalized immunotherapy. Int Rev Cell Mol
cells, multiple immune cells, fibroblasts, pericytes, the extra- Biol. 2022;370:163–92.
cellular matrix, and many other organ-specific cells. While 11. Rohaan MW, van den Berg JH, Kvistborg P, Haanen J. Adoptive
this field is quickly evolving, some recent discoveries include transfer of tumor-infiltrating lymphocytes in melanoma: a viable
treatment option. J Immunother Cancer. 2018;6(1):102.
regulating immunotherapy efficiency in different tumor
12. Lahiri A, Maji A, Potdar PD, Singh N, Parikh P, Bisht B, et al.
microenvironments by excluding cytotoxic T-cells or induc- Lung cancer immunotherapy: progress, pitfalls, and promises. Mol
ing immunosuppression [39, 40]. Cancer. 2023;22(1):40.
In summary, tissue sampling and acquisition for per- 13. Hoffmann MS, Hunter BD, Cobb PW, Varela JC, Munoz
J. Overcoming barriers to referral for chimeric antigen receptor
forming molecular and immunohistochemical studies in
T-cell therapy in patients with relapsed/refractory diffuse large
cancer specimens have become a standard of care. B-cell lymphoma. Transplant Cell Ther. 2023;29:440–8.
Surgeons, anesthesiologists, radiologists, and of course, 14. Naito T, Udagawa H, Sato J, Horinouchi H, Murakami S, Goto
pathologists must be familiar with the most current Y, et al. A minimum of 100 tumor cells in a single biopsy sam-
ple is required to assess programmed cell death ligand 1 expres-
requirements of tissue sampling. Particularly, tissue ade-
sion in predicting patient response to Nivolumab treatment in
quacy is an important consideration that can dramatically nonsquamous non-small cell lung carcinoma. J Thorac Oncol.
affect the need for further surgical procedures, which is 2019;14(10):1818–27.
undesirable. Nowadays, anatomic and molecular patholo- 15. Faber E, Grosu H, Sabir S, San Lucas FA, Barkoh BA, Bassett RL,
et al. Adequacy of small biopsy and cytology specimens for com-
gists can perform many tests with very little tissue.
prehensive genomic profiling of patients with non-small-cell lung
Moreover, liquid biopsies are becoming more frequent, cancer to determine eligibility for immune checkpoint inhibitor and
reducing the need for procedures with additional morbid- targeted therapy. J Clin Pathol. 2022;75(9):612–9.
ity and mortality, many of which might be done in poor 16. Kalemkerian GP, Narula N, Kennedy EB, Biermann WA, Donington
J, Leighl NB, et al. Molecular testing guideline for the selection of
surgical candidates.
patients with lung cancer for treatment with targeted tyrosine kinase
inhibitors: American Society of Clinical Oncology endorsement of
the College of American Pathologists/International Association for
References the Study of Lung Cancer/Association for Molecular Pathology
clinical practice guideline update. J Clin Oncol. 2018;36(9):911–9.
17. Mahlow J, Goold EA, Jedrzkiewicz J, Gulbahce HE. What to
1. Kontomanolis EN, Koutras A, Syllaios A, Schizas D, Mastoraki A,
expect from the new ASCO/CAP guideline recommendations for
Garmpis N, et al. Role of oncogenes and tumor-suppressor genes
hormone receptor testing in breast cancer: a national reference
in carcinogenesis: a review. Anticancer Res. 2020;40(11):6009–15.
laboratory experience. Appl Immunohistochem Mol Morphol.
2. Du X, Zhang J, Wang J, Lin X, Ding F. Role of miRNA in lung
2021;29(4):245–50.
cancer-potential biomarkers and therapies. Curr Pharm Des.
18. Mino-Kenudson M. Immunohistochemistry for predictive bio-
2018;23(39):5997–6010.
markers in non-small cell lung cancer. Transl Lung Cancer Res.
3. Holesova Z, Krasnicanova L, Saade R, Pos O, Budis J, Gazdarica J,
2017;6(5):570–87.
et al. Telomere length changes in cancer: insights on carcinogenesis
19. Cho M, Ahn S, Hong M, Bang H, Van Vrancken M, Kim S, et al.
and potential for non-invasive diagnostic strategies. Genes (Basel).
Tissue recommendations for precision cancer therapy using next
2023;14(3):715.
generation sequencing: a comprehensive single cancer center’s
4. Dearden S, Stevens J, Wu YL, Blowers D. Mutation incidence and
experiences. Oncotarget. 2017;8(26):42478–86.
coincidence in non small-cell lung cancer: meta-analyses by ethnic-
20. De Las Casas LE, Hicks DG. Pathologists at the leading edge of
ity and histology (mutMap). Ann Oncol. 2013;24(9):2371–6.
optimizing the tumor tissue journey for diagnostic accuracy and
5. Kohno T, Nakaoku T, Tsuta K, Tsuchihara K, Matsumoto S, Yoh K,
molecular testing. Am J Clin Pathol. 2021;155(6):781–92.
et al. Beyond ALK-RET, ROS1 and other oncogene fusions in lung
21. Rafael OC, Aziz M, Raftopoulos H, Vele OE, Xu W, Sugrue
cancer. Transl Lung Cancer Res. 2015;4(2):156–64.
C. Molecular testing in lung cancer: fine-needle aspiration speci-
6. Lindeman NI, Cagle PT, Aisner DL, Arcila ME, Beasley MB,
men adequacy and test prioritization prior to the CAP/IASLC/
Bernicker EH, et al. Updated molecular testing guideline for the
AMP molecular testing guideline publication. Cancer Cytopathol.
selection of lung cancer patients for treatment with targeted tyro-
2014;122(6):454–8.
sine kinase inhibitors: guideline from the College of American
22. Trejo Bittar HE, Luvison A, Miller C, Dacic S. A comparison
Pathologists, the International Association for the Study of Lung
of ALK gene rearrangement and ALK protein expression in
Cancer, and the Association for Molecular Pathology. Arch Pathol
­primary lung carcinoma and matched metastasis. Histopathology.
Lab Med. 2018;142(3):321–46.
2017;71(2):269–77.
7. Perez-Moreno P, Brambilla E, Thomas R, Soria JC. Squamous cell
23. Lin C, Shi X, Yang S, Zhao J, He Q, Jin Y, et al. Comparison of
carcinoma of the lung: molecular subtypes and therapeutic oppor-
ALK detection by FISH, IHC and NGS to predict benefit from
tunities. Clin Cancer Res. 2012;18(9):2443–51.
crizotinib in advanced non-small-cell lung cancer. Lung Cancer.
8. Pawelczyk K, Piotrowska A, Ciesielska U, Jablonska K, Gletzel-­
2019;131:62–8.
Plucinska N, Grzegrzolka J, et al. Role of PD-L1 expression in non-­
Oncogenesis, What Is New? 9

24. Jennings LJ, Arcila ME, Corless C, Kamel-Reid S, Lubin IM, 33. Peneau C, Imbeaud S, La Bella T, Hirsch TZ, Caruso S, Calderaro
Pfeifer J, et al. Guidelines for validation of next-generation J, et al. Hepatitis B virus integrations promote local and distant
sequencing-based oncology panels: a joint consensus recommen- oncogenic driver alterations in hepatocellular carcinoma. Gut.
dation of the Association for Molecular Pathology and College of 2022;71(3):616–26.
American pathologists. J Mol Diagn. 2017;19(3):341–65. 34. Khatun M, Ray R, Ray RB. Hepatitis C virus associated hepatocel-
25. Barua S, Wang G, Mansukhani M, Hsiao S, Fernandes H. Key con- lular carcinoma. Adv Cancer Res. 2021;149:103–42.
siderations for comprehensive validation of an RNA fusion NGS 35. Yu H, Robertson ES. Epstein-Barr virus history and pathogenesis.
panel. Pract Lab Med. 2020;21:e00173. Viruses. 2023;15(3):714.
26. Rodriguez J, Avila J, Rolfo C, Ruiz-Patino A, Russo A, Ricaurte L, 36. Lee YC, Chiang TH, Chou CK, Tu YK, Liao WC, Wu MS, et al.
et al. When tissue is an issue the liquid biopsy is nonissue: a review. Association between helicobacter pylori eradication and gas-
Oncol Ther. 2021;9(1):89–110. tric cancer incidence: a systematic review and meta-analysis.
27. Wu X, Li J, Gassa A, Buchner D, Alakus H, Dong Q, et al. Gastroenterology. 2016;150(5):1113–24.e5.
Circulating tumor DNA as an emerging liquid biopsy biomarker for 37. Sukri A, Hanafiah A, Mohamad Zin N, Kosai NR. Epidemiology
early diagnosis and therapeutic monitoring in hepatocellular carci- and role of Helicobacter pylori virulence factors in gastric cancer
noma. Int J Biol Sci. 2020;16(9):1551–62. carcinogenesis. APMIS. 2020;128(2):150–61.
28. Wray AJD, Minaker LM. Is cancer prevention influenced by the 38. Kennedy K, Khaddour K, Ramnath N, Weinberg F. The lung micro-
built environment? A multidisciplinary scoping review. Cancer. biome in carcinogenesis and immunotherapy treatment. Cancer J.
2019;125(19):3299–311. 2023;29(2):61–9.
29. Yamaguchi NH. Smoking, immunity, and DNA damage. Transl 39. de Visser KE, Joyce JA. The evolving tumor microenvironment:
Lung Cancer Res. 2019;8(Suppl 1):S3–6. from cancer initiation to metastatic outgrowth. Cancer Cell.
30. Shigeishi H. Association between human papillomavirus and oral 2023;41(3):374–403.
cancer: a literature review. Int J Clin Oncol. 2023;28:982–9. 40. Kirchhammer N, Trefny MP, Auf der Maur P, Laubli H, Zippelius
31. Kjaer SK, Dehlendorff C, Belmonte F, Baandrup L. Real-world A. Combination cancer immunotherapies: emerging treatment
effectiveness of human papillomavirus vaccination against cervical strategies adapted to the tumor microenvironment. Sci Transl Med.
cancer. J Natl Cancer Inst. 2021;113(10):1329–35. 2022;14(670):eabo3605.
32. DeCoste RC, Carter MD, Ly TY, Gruchy JR, Nicolela AP, Pasternak
S. Merkel cell carcinoma: an update. Hum Pathol. 2023;140:39–52.
Epidemiology of Cancer

Hui-Yi Lin and Jong Y. Park

1 Introduction a 28% increase, three-fold higher than the trend of total mor-
tality during the same period (9%) [2, 3]. Cancer mortality
Over 19 million new cancer cases [1–4] lead to almost ten rates are varied in different parts of the world due to genetic
million deaths globally in 2020 [1, 2]. The most common and environmental factors, such as quality of patient care,
cancers worldwide are breast and lung cancers, while pros- availability of medical facilities, and socioeconomic status.
tate cancer is the most common in the male population These factors affect the application of screening, prevention
(Table 1) [2]. The responsible risk factors for cancer inci- strategies, and appropriate treatments for cancers. Recent
dence vary by cancer type, geographical region, and popula- advances in medical technologies can provide better screen-
tion [5]. In addition, recent environmental changes by climate ing tools and, more importantly, better patient care and treat-
changes, industrialization, and lifestyle, such as diet patterns, ments. These advances improved the prognosis and survival
are suggested as potential contributing factors for increasing of most cancers. However, patient care and public health sys-
cancer incidence [6]. Increased incidences in different tems are considerably different among countries, and these
regions or populations (such as race or gender) have been differences exist within countries [2]. These systems, socio-
reported for specific cancer types. The incidence rates in men economic, genetic, and environmental factors can be
are higher than those in women in all cancers except thyroid explained different survival rates. Among environmental fac-
cancer. For example, the bladder (2.87), liver (2.35), and tors, diet patterns and physical activity are often associated
esophageal (2.36) cancers showed the highest men/women with a risk for several cancers [7]. Typically, a healthy diet
ratios [2, 3]. and being physically active can lower the risk and improve
In terms of cancer mortality, cancer is the second leading a survival for women with breast cancer. Several factors,
cause of death, accounting for 9.9 million deaths worldwide such as the type of cancer, clinical stage at diagnosis, and
[2]. The trend of cancer mortality in the last decade showed medical care, are the main factors for cancer survival. In
most cases, early diagnosis is a main key for better outcomes
Table 1 Number of new cancer cases in worldwide and survival. The incidence rank of cancer type does not nec-
Cancer sites Cases Percentage essarily overlap with the mortality rank due to different can-
Breast 2,261,419 12.5% cer survival rates. The numbers of deaths from lung, liver,
Lung 2,206,771 12.2% pancreas, and stomach cancers are much higher than others.
Colorectal 1,931,590 10.7% In men, lung, liver, stomach, esophageal, and prostate can-
Prostate 1,414,259 7.8%
cers are the deadliest. In women, breast, lung, and stomach
Stomach 1,089,103 6.0%
cancers are the lethal ones. Colorectal cancer is the second
Liver 905,677 5.0%
leading cause of cancer mortality in both populations [8].
Data from GLOBOCAN 2020 [2–4]

H.-Y. Lin
Biostatistics Program, School of Public Health, Louisiana State
University Health Sciences Center, New Orleans, LA, USA
e-mail: hlin1@lsuhsc.edu
J. Y. Park (*)
Department of Cancer Epidemiology, Moffitt Cancer Center,
Tampa, FL, USA
e-mail: Jong.park@moffitt.org

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 11


J. Huang et al. (eds.), Anesthesia for Oncological Surgery, https://doi.org/10.1007/978-3-031-50977-3_2
12 H.-Y. Lin and J. Y. Park

2 Cancer Incidence and Trend with MRI for prostate cancer provides better sensitivity for
detection, reduces the over-diagnosis, and over-treatment
The World Health Organization (WHO) data from WHO [16]. However, the main obstacles, such as const-­
shows cancers cause the largest economic burden worldwide effectiveness, to apply MRI as a prostate cancer screening
among all human diseases [2, 3]. The most considerable tool need to be cleared.
cancer-­related burden is in the population aged 60 years or Lung cancer is the second most common cancer and the
older. According to the WHO Global Cancer Observatory leading cause of cancer mortality in 2020 [4]. The incidence
data, breast (2.3 million cases), lung (2.21 million cases), rate has decreased since the mid-1980s as the prevalence of
colorectal (1.93 million patients), and prostate (1.41 million smoking declined [17]. However, the incidence of lung can-
patients) cancers are the most common cancers [2, 3]. The cer among women is still rising in many countries. In these
most common cancers in men are lung, prostate, stomach, countries, smoking prevalence in women has either peaked
and liver, while breast, lung, cervical, and colon cancers are recently or continues to rise. Therefore, lung cancer inci-
most frequent among women. In terms of incidence rates, dence in women will most likely increase for at least a few
breast cancer has the highest incidence rate (46.3 per 100,000), decades more [18].
then prostate (29.3 per 100,000), lung (22.5 per 100,000), and Breast cancer incidence has increased since the 1980s due
colorectal (19.2 per 100,000) cancers are followed [4]. to the early detection of asymptomatic cases by mammogra-
Based on the American Cancer Society (ACS) report [8], phy screening. In addition, body mass index (BMI) changes,
the trend of cancer incidence was mainly influenced by envi- rising age for the first births, and declining fertility rate also
ronmental factors and patient care, especially cancer screen- contribute to the increasing trend of breast cancer incidence
ing. The discussion of incidence trends for these common [19].
cancer types is listed below. As for colorectal cancer, its incidence rates are rising in
Prostate cancer cases surged in the 1990s due to the intro- low-income countries, while incidences have declined in
duction of prostate-specific antigen (PSA) screening tests. developed countries since the mid-1990. The introduction of
Most cases identified through this screening test are asymp- screening tests, such as a colonoscopy and fecal occult blood
tomatic and early-stage prostate cancer cases. Therefore, ris- test, in old age groups drives decreased an incidence rate of
ing prostate cancer incidence may lead to the over-diagnosis colorectal cancer. However, incidence rates in the younger
and over-treatment due to these new PSA screening tests. generation are rising, most likely due to environmental fac-
Because of these concerns, there were uncertainty and dis- tors, such as lifestyle, rapid dietary transition, and obesity
agreement on the value of the PSA test [9]. The United States [20].
Preventive Services Task Force (USPSTF) recommended In addition to these four most common cancer types, cer-
new guidelines on PSA tests in 2008 and 2012 [10, 11]. They vical cancer incidence rates have decreased since the 1970s
advised against PSA screening among men older than due to PAP smear screening, which evaluates for cancerous
75 years in 2008. In 2012, the USPSTF recommended or precancerous cells on the cervix. The trend of cervical
against PSA screening for all men, regardless of age [11]. As cancer incidence rates varies by race and age group. Hispanic
a result, prostate cancer incidence was increased from 2013 women in the United States showed the highest incidence of
until now. In 2018, the USPSTF recommended that men cervical cancer compared with other racial groups. Especially
aged 55–69 should discuss the possible benefits and harms of the incidence rates are increased in young Hispanic women
PSA screening with their healthcare provider and make an [21, 22]. Low cervical cancer screening rates and high human
individualized decision about whether to get screened [12, papillomavirus (HPV) infection among Hispanic women can
13]. partially explain rising cervical cancer incidence rates [22].
For the impact of the PSA test on prostate cancer mortal- Perhaps, the most significant impact on reducing cervical
ity, Fenton et al. (2018) reported that PSA screening might cancer incidence is the introduction of vaccines against car-
provide benefits in reducing prostate cancer-specific mortal- cinogenic HPVs, type 16 and 18. These vaccines were
ity. However, this PSA test is notoriously linked to high approved in 2006 by the US Food and Drug Administration
false-positive results, often leading to an unnecessary biopsy [23]. Therefore, incidence rates of cervical cancer in the gen-
and overtreatment. In addition, long-term survival benefits eration who received the HPV vaccine were declined signifi-
among screen-detected prostate cancer were unclear [13]. cantly [24].
Recent studies evaluated magnetic resonance imaging (MRI)
as a new screening tool for prostate cancer [14]. MRI of the
prostate has become an important part of the initial radio- 3 Lifetime Risk for Cancers
graphic evaluation for the diagnosis of prostate cancer.
Recent European Association of Urology (EAU) guidelines The lifetime probability of developing cancer is 41% in men
recommend performing MRI before prostate biopsy in men and 40% in women [21]. The lifetime risk varies significantly
with high risk for prostate cancer [15]. A risk assessment by different cancer types. According to the ACS data, the high-
Epidemiology of Cancer 13

Table 2 Lifetime risk for cancers by gender in the United States Table 3 Five-year relative cancer survival rates of the top 10 common
Cancer sites Gender Percentage cancersa
All sites Male 41% Cancer sites Percentage
Female 40% All sites 68%
Breast Female 13% Prostate 97%
Prostate Male 13% Breast 91%
Lung Male 6.2% Bladder 77%
Female 5.8% Non-Hodgkin lymphoma 74%
Colorectal Male 4.3% Cervix 67%
Female 3.9% Leukemia 66%
a
Modified from ACS data [8] Colorectal 65%
Ovary 50%
Lung 23%
Pancreas 12%
est risks for cancer were found for prostate (13%), lung (6.2%), a
Modified from ACS data 2012–2018 [8]
and colorectal cancer (4.3%) in men, and breast (13%), lung
(5.8%), and colorectal (3.9%) in women (Table 2) [3, 8].
The lifetime risk for overall cancer mortality is 10.6%. leukemia has provided many new innovative treatments,
The highest risks of cancer mortality are from lung (3.19%), such as monoclonal antibodies and immune CAR-T cells.
liver (1.46%), and stomach (1.36%) in men and breast These new treatments significantly improved the prognosis
(1.41%), lung (1.32%), and cervix (0.77%) in women [8]. of chronic myeloid leukemia [27]. For melanoma metastasis,
Generally, the mortality rate of each cancer in men is higher several new immune therapies, and BRAF and MEK inhibi-
than in women, except for thyroid cancer. Especially mortal- tors were introduced [29]. Immune checkpoint inhibitors
ity rate ratios of the bladder (2.87), esophagus (2.36), and induce cancer-cell killing by activated CD8-positive T cells.
liver (2.35) cancers between men and women are among the Immune checkpoint inhibitors, such as anti-CTLA4 and anti-­
highest cancers [8]. PD-­1, changed the methods of patient care in several cancer
types, including melanoma and kidney cancer. Due to these
new therapies, the survival rate of melanoma dramatically
4 Survival Rates for Cancers increased [29]. These immunotherapies also demonstrated
their potential benefits for lung cancer. Due to these treat-
Based on the ACS report for the US population in 2012– ments, 3-year survival for all lung cancer cases increased
2018 [8], the 5-year survival rates of the top 10 common from 22 to 33% over time, corresponding to the timing of
types of cancers are listed in Table 3. The 5-year relative sur- approval of targeted therapy [30]. On the other hand, uterine
vival rate for all cancers was 68% in 2012–2018, while the cancer has not improved its survival rate long time, possibly
5-year survival rate was 49% in the 1970s. Prostate cancer due to the lack of new treatments [31].
showed the best overall prognosis with a 5-year survival rate Survival rates for cancers are also different in different
of 97%. However, lung and pancreas cancers have the worst racial groups. Survival rates in non-Hispanic black group are
5-year survival rates, 23%, and 12%, respectively [8]. usually lower than in non-Hispanic white group for most
Globally, the 5-year survival rates are 70–100% for prostate cancers. Even after adjusting for clinical stage, gender, and
cancer, 80–85% for breast cancer, 50–70% for colorectal age at diagnosis, the survival rates are 33% lower in non-­
cancer, and 10–20% for lung cancer [3]. Hispanic blacks as compared with non-Hispanic whites [32].
Improvement of cancer patients’ survival is affected by For example, survival rates for uterine cancer are different in
multiple factors, including early detection and better treat- different racial groups. African American women are more
ments [25]. For example, the 5-year survival rate for chronic likely to be diagnosed with higher clinical stages and
myeloid leukemia has significantly increased from 17% in this higher clinical stage in diagnosis often leads to lower
1975 to 73% in 2012 [26, 27], although the incidence of survival rates [33]. Therefore, African American women
chronic myeloid leukemia has been growing in the last few have the highest mortality rate in all races for uterine corpus
decades. The potential explanation for the increasing inci- cancer [33]. The recent reclassification of subtypes based on
dence rate can be the reclassification of other leukemia sub- molecular biomarkers from next-generation sequencing was
types, raising awareness, improved diagnostic sensitivity, introduced for targeted therapies. Recent whole-exome
better-reporting systems for new cases, and other risk fac- sequencing studies have identified a role of the HER2/NEU
tors, such as obesity [27]. Further, the 5-year survival rate and driver mutations in the PIK3CA/AKT/mTOR oncogenic
increase of chronic myeloid leukemia was also due to new pathways [34]. These results emphasize the relevance of
innovative treatments, such as stem cell transplantation these novel therapeutic targets for targeted therapy. These
through national health policies and cancer control programs therapies provided a large impact because almost one-half of
[28]. The progress in pre-clinical and clinical research on uterine cancers are candidates for targeted therapies [35].
14 H.-Y. Lin and J. Y. Park

Table 4 Number of deaths by top 5 deadliest cancers based cancer risk stratification [38–40]. Although accuracy
Cancer sites Incidence Percentage of these PRS are different in different cancers, a high poly-
Lung 1,796,144 18.2% genic risk score is correlated with a higher risk of cancer. In
Colorectal 935,173 9.5% prostate cancer, the detection rate of polygenic risk score
Liver 830,180 8.4 ranged from 0.56 to 0.67 [40]. Recent large studies observed
Stomach 768,793 7.8
that the top 10% polygenic risk score group increased risk
Breast 684,996 6.9%
2.7-fold [41, 42].
Data from GLOBOCAN 2020 [2–4]
Smoking is the most well-established risk factor for many
cancers, including lung, laryngeal, oral, esophageal, pan-
5 Cancer Mortality and Trend creas, bladder, kidney, liver, stomach, pancreas, colorectal,
and cervical cancers. Especially cigarette smoking is respon-
Currently, heart disease is the leading cause of death in the sible for approximately 80–90% of lung cancer mortality
world, based on WHO data [4]. The second leading cause of [43]. Heavy smokers showed a 20 times higher risk for lung
death is cancer. In Table 4, a list of cancer deaths was pre- cancer-specific death than never-smokers. Alcohol drinking
sented based on GLOBOCAN 2020 [2, 4]. As expected, the also increases the risk for several cancers, such as oral, laryn-
rank in cancer mortality does not necessarily match with one geal, colorectal, esophageal, liver, and breast cancers. Almost
in cancer incidence because the survival rates are different in 4% of new cancers diagnosed in 2020 worldwide can be
different cancer types. Overall, lung, colorectal, liver, stom- explained by alcohol drinking based on WHO data [4]. Diets,
ach, and breast cancers are the deadliest cancers (Table 4). especially high-fat diets, are associated with an increased
Cancer mortality rates have increased consistently from the risk of colorectal, lung, and prostate cancers. The previous
1930s until the 2000s. One of the main attributable factors is pre-clinical study reported that certain compounds induced
a rapid increase in lung cancer mortality due to the smoking cancers [44]. However, these findings are not consistent with
epidemic. However, smoking reduction has significantly results from human studies. Recent epidemiological studies
declined cancer deaths since 1991 [8]. In addition, lung suggested that high-fat diets have been linked with an
screening with low-dose computed tomography (CT) for increased risk of these cancers [45].
high-risk populations, such as heavy smokers, leads to detect Infections are another risk factor for several cancers.
early-stage lung cancers successfully and lower cancer mor- Approximately 15–20% of cancer incidences in the world
tality [36]. are related to infections, especially carcinogenic viruses
[46], such as Human Papillomavirus, Hepatitis B virus,
Hepatitis C virus, Helicobacter pylori, Human
6 Risk Factors for Cancers Immunodeficiency virus, and Epstein-Barr virus. Human
papillomavirus infection, especially types 16 and 18,
The development of cancers is the result of the combination increases the risk of cervical cancer [47], while Hepatitis B
of genetic and environmental factors. The leading risk factor and C viruses increase the risk of liver cancer [48, 49].
is age, probably due to the accumulation of various risks Helicobacter pylori infection is associated with a high risk of
with age. In addition, DNA repair processes are less efficient stomach cancer [50]. Human immunodeficiency virus
in the elderly. There are other well-established risk factors increases the risk of developing some cancers such as Kaposi
for cancers, such as genetics, smoking, alcohol drinking, sarcoma [51]. Epstein-Barr virus is linked to Hodgkin lym-
high-fat diet, and infections, particularly carcinogenic infec- phoma, Burkitt lymphoma, and nasopharyngeal cancers
tions. Genetic factors contribute to almost one-third of all [46].
cancer cases [37]. However, an individual genetic variant
was not demonstrated as a major factor for cancer risk.
However, the polygenic risk scores provided more reliable 7 Summary
predictions. The polygenic risk scores are usually built based
on the sum of multiple germline genetic variants identified Based on the current trend of cancer epidemiological data,
from genome-wide association studies and can be used to these malignant diseases will be a significant public health
classify people’s risk of diseases based on their genetic pro- issue. This disease will impact clinical and social standards
files. These scores may help to predict lifetime risk, plan for and impose an enormous economic burden. However, recent
better screening, predict prognosis and, more importantly, advances in medical technology provide promising tools for
support personalized medicine [38–40]. Although the pre- better cancer care, screening, and early detection. For exam-
diction of individual cancer risk based on only polygenic risk ple, liquid biopsy, a blood test that detects cancer cells or
scores is not ready for the clinical use, an increasing number tumor DNA that are circulating in the blood, can be used for
of studies support using polygenic risk scores for population-­ more efficient screening, early detection, and monitoring
Epidemiology of Cancer 15

treatment responses. Second, MRI can be used for diagnosis 12. Fenton JJ, Weyrich MS, Durbin S, Liu Y, Bang H, Melnikow
J. Prostate-specific antigen-based screening for prostate cancer:
and monitoring progression. Third, genetic profile contrib-
evidence report and systematic review for the US preventive ser-
utes to a risk classification for utilization in personalized vices task force. JAMA. 2018;319(18):1914–31.
medicine. Fourth, a new generation of treatments, especially 13. U.S. Preventive Services Task Force, Grossman DC, Curry SJ,
immunotherapy, opened a promising path for cancer treat- Owens DK, Bibbins-Domingo K, Caughey AB, et al. Screening for
prostate cancer: US preventive services task force recommendation
ment by enhancing immune function against cancer cells.
statement. JAMA. 2018;319(18):1901–13.
Finally, statistical learning and artificial intelligence 14. Nordstrom T, Discacciati A, Bergman M, Clements M, Aly M,
improved accuracy and efficiency in pathology and imaging Annerstedt M, et al. Prostate cancer screening using a combination
analysis for tumor tissues. of risk-prediction, MRI, and targeted prostate biopsies (STHLM3-­
MRI): a prospective, population-based, randomised, open-label,
Despite recent advances in cancer treatment and screen-
non-inferiority trial. Lancet Oncol. 2021;22(9):1240–9.
ing, there are still several challenges to be overcome for bet- 15. Mottet N, van den Bergh RCN, Briers E, Van den Broeck T,
ter survival and early detection. For example, disparity Cumberbatch MG, De Santis M, et al. EAU-EANM-ESTRO-­
among people with a low socioeconomic status and different ESUR-SIOG guidelines on prostate cancer-2020 update. Part 1:
screening, diagnosis, and local treatment with curative intent. Eur
racial groups is one of the main obstacles. These people are
Urol. 2021;79(2):243–62.
less likely screened and receive less quality cancer cares. In 16. Kasivisvanathan V, Rannikko AS, Borghi M, Panebianco V,
summary, the recent trends in cancer incidence and mortality Mynderse LA, Vaarala MH, et al. MRI-targeted or standard biopsy
rates have increased. These trends may be continued due to for prostate-cancer diagnosis. N Engl J Med. 2018;378(19):1767–77.
17. Jemal A, Ma J, Rosenberg PS, Siegel R, Anderson WF. Increasing
various factors, such as industrialization, air pollution, diet
lung cancer death rates among young women in southern and mid-
change, and longer life span. western states. J Clin Oncol. 2012;30(22):2739–44.
18. Jha P. Avoidable global cancer deaths and total deaths from smok-
ing. Nat Rev Cancer. 2009;9(9):655–64.
19. Giaquinto AN, Sung H, Miller KD, Kramer JL, Newman LA,
References Minihan A, et al. Breast cancer statistics, 2022. CA Cancer J Clin.
2022;72(6):524–41.
1. Ferlay J, Colombet M, Soerjomataram I, Parkin DM, Pineros M, 20. Siegel RL, Torre LA, Soerjomataram I, Hayes RB, Bray F, Weber
Znaor A, et al. Cancer statistics for the year 2020: an overview. Int TK, et al. Global patterns and trends in colorectal cancer incidence
J Cancer. 2021;149:778–89. in young adults. Gut. 2019;68(12):2179–85.
2. Morgan E, Arnold M, Gini A, Lorenzoni V, Cabasag CJ, Laversanne 21. Miller KD, Ortiz AP, Pinheiro PS, Bandi P, Minihan A, Fuchs HE,
M, et al. Global burden of colorectal cancer in 2020 and 2040: et al. Cancer statistics for the US Hispanic/Latino population, 2021.
incidence and mortality estimates from GLOBOCAN. Gut. CA Cancer J Clin. 2021;71(6):466–87.
2023;72(2):338–44. 22. Ortiz AP, Ortiz-Ortiz KJ, Colon-Lopez V, Tortolero-Luna G,
3. Allemani C, Weir HK, Carreira H, Harewood R, Spika D, Wang Torres-Cintron CR, Wu CF, et al. Incidence of cervical cancer in
XS, et al. Global surveillance of cancer survival 1995-2009: analy- Puerto Rico, 2001-2017. JAMA Oncol. 2021;7(3):456–8.
sis of individual data for 25,676,887 patients from 279 population-­ 23. de Martel C, Plummer M, Vignat J, Franceschi S. Worldwide bur-
based registries in 67 countries (CONCORD-2). Lancet. den of cancer attributable to HPV by site, country and HPV type.
2015;385(9972):977–1010. Int J Cancer. 2017;141(4):664–70.
4. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, 24. Mix JM, Van Dyne EA, Saraiya M, Hallowell BD, Thomas
Jemal A, et al. Global cancer statistics 2020: GLOBOCAN esti- CC. Assessing impact of HPV vaccination on cervical cancer
mates of incidence and mortality worldwide for 36 cancers in 185 incidence among women aged 15-29 years in the United States,
countries. CA Cancer J Clin. 2021;71(3):209–49. 1999-2017: an ecologic study. Cancer Epidemiol Biomark Prev.
5. Feola S, Chiaro J, Martins B, Russo S, Fusciello M, Ylosmaki E, 2021;30(1):30–7.
et al. A novel immunopeptidomic-based pipeline for the generation 25. Croswell JM, Ransohoff DF, Kramer BS. Principles of cancer
of personalized oncolytic cancer vaccines. Elife. 2022;11:e71156. screening: lessons from history and study design issues. Semin
6. Eala MAB, Robredo JPG, Dee EC, Lin V, Lagmay A. Climate cri- Oncol. 2010;37(3):202–15.
sis and cancer: perspectives from the hardest hit. Lancet Oncol. 26. Sasaki K, Strom SS, O’Brien S, Jabbour E, Ravandi F, Konopleva
2022;23(3):e92. M, et al. Relative survival in patients with chronic-phase chronic
7. Byrne S, Boyle T, Ahmed M, Lee SH, Benyamin B, Hypponen myeloid leukaemia in the tyrosine-kinase inhibitor era: analysis of
E. Lifestyle, genetic risk and incidence of cancer: a prospective patient data from six prospective clinical trials. Lancet Haematol.
cohort study of 13 cancer types. Int J Epidemiol. 2023;52:817–26. 2015;2(5):e186–93.
8. Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. 27. Yang X, Chen H, Man J, Zhang T, Yin X, He Q, et al. Secular trends
CA Cancer J Clin. 2023;73(1):17–48. in the incidence and survival of all leukemia types in the United
9. Potosky AL, Miller BA, Albertsen PC, Kramer BS. The role of States from 1975 to 2017. J Cancer. 2021;12(8):2326–35.
increasing detection in the rising incidence of prostate cancer. 28. Bailey C, Richardson LC, Allemani C, Bonaventure A, Harewood R,
JAMA. 1995;273(7):548–52. Moore AR, et al. Adult leukemia survival trends in the United States
10. Jemal A, Fedewa SA, Ma J, Siegel R, Lin CC, Brawley O, by subtype: a population-based registry study of 370,994 patients
et al. Prostate cancer incidence and PSA testing patterns in diagnosed during 1995-2009. Cancer. 2018;124(19):3856–67.
relation to USPSTF screening recommendations. JAMA. 29. Carlino MS, Larkin J, Long GV. Immune checkpoint inhibitors in
2015;314(19):2054–61. melanoma. Lancet. 2021;398(10304):1002–14.
11. Moyer VA, U.S. Preventive Services Task Force. Screening for 30. Howlader N, Forjaz G, Mooradian MJ, Meza R, Kong CY, Cronin
prostate cancer: U.S. preventive services task force recommenda- KA, et al. The effect of advances in lung-cancer treatment on popu-
tion statement. Ann Intern Med. 2012;157(2):120–34. lation mortality. N Engl J Med. 2020;383(7):640–9.
16 H.-Y. Lin and J. Y. Park

31. McConechy MK, Talhouk A, Leung S, Chiu D, Yang W, Senz 42. Schumacher FR, Al Olama AA, Berndt SI, Benlloch S, Ahmed
J, et al. Endometrial carcinomas with POLE exonuclease M, Saunders EJ, et al. Association analyses of more than 140,000
domain mutations have a favorable prognosis. Clin Cancer Res. men identify 63 new prostate cancer susceptibility loci. Nat Genet.
2016;22(12):2865–73. 2018;50(7):928–36.
32. Jemal A, Ward EM, Johnson CJ, Cronin KA, Ma J, Ryerson B, 43. Morabia A, Wynder EL. Cigarette smoking and lung cancer cell
et al. Annual report to the nation on the status of cancer, 1975-2014, types. Cancer. 1991;68(9):2074–8.
featuring survival. J Natl Cancer Inst. 2017;109(9):djx030. 44. Yang J, Wei H, Zhou Y, Szeto CH, Li C, Lin Y, et al. High-fat diet
33. Clarke MA, Devesa SS, Hammer A, Wentzensen N. Racial and promotes colorectal tumorigenesis through modulating gut micro-
ethnic differences in hysterectomy-corrected uterine corpus can- biota and metabolites. Gastroenterology. 2022;162(1):135–49 e2.
cer mortality by stage and histologic subtype. JAMA Oncol. 45. Dunneram Y, Greenwood DC, Cade JE. Diet, menopause and the
2022;8(6):895–903. risk of ovarian, endometrial and breast cancer. Proc Nutr Soc.
34. Ding J, Bashashati A, Roth A, Oloumi A, Tse K, Zeng T, et al. 2019;78(3):438–48.
Feature-based classifiers for somatic mutation detection in tumour-­ 46. Gomez K, Schiavoni G, Nam Y, Reynier JB, Khamnei C, Aitken
normal paired sequencing data. Bioinformatics. 2012;28(2):167–75. M, et al. Genomic landscape of virus-associated cancers. medRxiv.
35. Black JD, English DP, Roque DM, Santin AD. Targeted therapy 2023.
in uterine serous carcinoma: an aggressive variant of endometrial 47. Eun BW, Bahar E, Xavier S, Kim H, Borys D. Post-marketing sur-
cancer. Womens Health (Lond). 2014;10(1):45–57. veillance study of the safety of the HPV-16/18 vaccine in Korea
36. de Koning HJ, van der Aalst CM, de Jong PA, Scholten ET, (2017-2021). Hum Vaccin Immunother. 2023;19(1):2184756.
Nackaerts K, Heuvelmans MA, et al. Reduced lung-cancer mortal- 48. Min Y, Wei X, Xia X, Wei Z, Li R, Jin J, et al. Hepatitis B virus
ity with volume CT screening in a randomized trial. N Engl J Med. infection: an insight into the clinical connection and molecular
2020;382(6):503–13. interaction between hepatitis B virus and host extrahepatic cancer
37. Mucci LA, Hjelmborg JB, Harris JR, Czene K, Havelick DJ, risk. Front Immunol. 2023;14:1141956.
Scheike T, et al. Familial risk and heritability of cancer among 49. Fraile-Lopez M, Alvarez-Navascues C, Gonzalez-Dieguez ML,
twins in Nordic countries. JAMA. 2016;315(1):68–76. Cadahia V, Chiminazzo V, Castano A, et al. Predictive models
38. Dai J, Lv J, Zhu M, Wang Y, Qin N, Ma H, et al. Identification of for hepatocellular carcinoma development after sustained viro-
risk loci and a polygenic risk score for lung cancer: a large-scale logical response in advanced hepatitis C. Gastroenterol Hepatol.
prospective cohort study in Chinese populations. Lancet Respir 2023;46:754–63.
Med. 2019;7(10):881–91. 50. Liao O, Ye G, Du Q, Ye J. Gastric microbiota in gastric cancer and
39. Thomas M, Sakoda LC, Hoffmeister M, Rosenthal EA, Lee JK, precancerous stages: mechanisms of carcinogenesis and clinical
van Duijnhoven FJB, et al. Genome-wide modeling of poly- value. Helicobacter. 2023;28:e12964.
genic risk score in colorectal cancer risk. Am J Hum Genet. 51. Mendoza Mori LM, Valenzuela Medina JB, Gotuzzo Herencia JE,
2020;107(3):432–44. Bravo Puccio FG, Mejia Cordero FA, Mohanna Barrenechea S,
40. Oh JJ, Hong SK. Polygenic risk score in prostate cancer. Curr Opin et al. Kaposi’s sarcoma in people living with HIV/aids in a pub-
Urol. 2022;32(5):466–71. lic referral hospital in Peru. Rev Peru Med Exp Salud Publica.
41. Eeles RA, Kote-Jarai Z, Giles GG, Olama AA, Guy M, Jugurnauth 2022;39(3):352–6.
SK, et al. Multiple newly identified loci associated with prostate
cancer susceptibility. Nat Genet. 2008;40(3):316–21.
Immunemodulation and Cancer

Jinhong Liu and Jeffrey Huang

1 Introduction In vivo, tumor antigens are generated endogenously as cyto-


solic or nuclear proteins that are coupled with class I major
The immune system serves as the fundamental defense histocompatibility complex (MHC)-associated peptides.
mechanism of the human body, playing a crucial role in pre- These peptides can then be recognized by CD8+ CTLs that
venting tumorigenesis and managing the pathological pro- are restricted by class I MHC and possess the ability to kill
gression of cancer. Despite constant pressure from the antigen-specific target cells. The process of CTL activation is
immune system and the tumor microenvironment, cancer highly regulated and modulated at both the molecular and
cells can transform into aggressive, abnormal malignant cellular levels, involving multiple stimulatory and inhibitory
cells and develop immunity-suppressing capabilities, even- steps. Failure to properly regulate these positive and negative
tually leading to mutant progression or immune evasion. The balances can result in tumor immune evasion.
immune system is a highly organized and intricate network
of cells that is both dynamic and complex. Immune cells are
motile and migrate to specific organs in a context-specific 2.2 T Cell Activation Require Antigen
manner, where they must come into close proximity with Presenting Cell
other cells to exchange information and function appropri-
ately. These interactions give rise to diverse immune For CD8+ cells to differentiate into CTLs, a healthy individ-
responses in different situations. In cancer, the intra-organ ual requires an antigen-presenting cell (APC) that can present
movement or infiltration of active immune cells into the tumor antigen (a protein peptide associated with class I MHC)
tumor tissue is a decisive factor that determines immune out- to T cells. Tumor antigens or tumor-associated antigens
come [1, 2]. Over the past decade, significant progress has (TAAs) resulting from mutations and epigenetic changes in
been made in cancer immunotherapy, which has successfully APCs induce the expansion of specific CTLs, differentiation
cured certain types of cancer in some patients across several into effector cells, and the generation of memory cells. The
different cancer types. A key factor in this success is under- interaction between the T cell receptor (TCR) and MHC-
standing the mechanisms of immune modulation and effec- associated peptides on APCs (signal 1) determines the quality
tively directing cytotoxic T cell responses against tumors. of T cell activation, which is further influenced by the pres-
ence, type, and strength of signaling through costimulatory
receptors on T cells and APCs (signal 2). The degree and type
2 Tumor Immunity of activation of CD8+ T lymphocytes are strongly influenced
by these factors. As a consequence of interactions and recog-
2.1 Tumor Immunity Require CD8+ T Cell nition of tumor antigen presented by APCs through T cell
Activation antigen receptor, it initiates and activates a series of sequen-
tial process that lead to the generation of both antigen specific
The key immune mechanism that combats cancer is the acti- effector and memory CTLs. These cytotoxic CD8+ T lym-
vation of cytotoxic T lymphocytes (CTLs), which specifi- phocytes provide long-term protection against pathogens and
cally target tumor cells through tumor-specific antigens [3]. carry out immune surveillance to eliminate cancer cells,
therefore, APCs are the most efficient stimulators of naïve T
J. Liu (*) · J. Huang cells into CD8+ CTLs. Many types of cells originating from
Anesthesiology Department, Moffitt Cancer Center, myeloid linage can serve as APCs, including dendritic cells,
Tampa, FL, USA
monocytes/macrophages, and granulocytes [4, 5].
e-mail: Jinhong.Liu@Moffitt.org

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 17


J. Huang et al. (eds.), Anesthesia for Oncological Surgery, https://doi.org/10.1007/978-3-031-50977-3_3
18 J. Liu and J. Huang

3 Antigen Presenting Cells and Cancer tumors through various mechanisms and can impede the effi-
cacy of immunotherapy, chemotherapy, and irradiation. In
3.1 Basic Biology of Dendritic Cells contrast, DCs thus have a unique feature and potential to pro-
cess and transfer tumor antigens to the draining lymph nodes
Dendritic cells (DCs) are vital in activating naïve CD4+ T leading to the activation of T-cell, a crucial step that is abso-
cells and are classified as professional antigen-presenting lutely required for T cell-dependent tumor immunity and
cells (APCs), alongside macrophages, granulocytes, and B response to immunotherapy. Tumor-resident DCs have an
cells. DCs are the essential APCs of the immune system, increasingly important function in regulating the T cell
playing a crucial role in connecting innate and adaptive response within tumors during therapy [5, 6, 9–11]. These
immunity, particularly in stimulating anti-tumor T cells [5– functions establish DCs as a critical component of the antitu-
7]. DCs arise from bone marrow progenitors known as com- mor T cell response and indicate that modulating the biologi-
mon myeloid progenitors (CMPs). CMPs have the ability to cal activity of these cells is a valuable therapeutic strategy
differentiate into monocytes or the common dendritic cell for indirectly promoting a T cell response during therapy.
progenitor, which gives rise to various subpopulations of Among DC suptype, studies in both mice and humans
dendritic cells (DCs), including plasmacytoid DCs (pDCs), have demonstrated that the cDC subset of DCs is primarily
conventional DCs (cDCs), and monocytic-derived DCs. responsible for eliciting an effective immune response
These immature DCs require maturation signals, such as against cancer. The regulation and function of cDCs are gov-
damage or pathogen-associated molecular patterns (DAMPs) erned by CCR7, a leukocyte chemotactic receptor that directs
and inflammatory cytokines, to effectively fulfill their role in the migration of DCs to the lymph node and facilitates the
the immune response. Upon maturation and activation, DCs generation of a systemic CTL-mediated anti-tumor immune
reduce their phagocytic activity, increase expression of MHC response. In mouse tumor models, cDCs that lack CCR7
class II and costimulatory molecules, produce more cyto- expression are unable to attract CD8+ T cells to the tumor
kines, and demonstrate enhanced migration to lymph nodes site and also fail to stimulate T cells within the tumor to dif-
through increased expression of chemokine receptors [5, 8]. ferentiate into effector CTLs capable of eliminating the
DCs exist as resident lymphoid tissue, such as spleen and tumor. Defective expression of CCR7 in cDCs in multiple
lymph nodes were critical for sampling blood and lymph mouse tumor models leads to an inability to attract CD8+ T
born antigens, respectively. This event is relatively important cells to the tumor site, as well as a failure to stimulate T cells
because distributing and presenting antigens is highly within the tumor tissue to differentiate into effector CTLs
context-­specific, depending on the type of antigen and route capable of eliminating the tumors [6]. Notably, it has been
of exposure. In the case of cancer, it is not fully understood shown that the failure of tumors to attract the cDCs inhibits
how endogenous tumor antigen is processed and delivered activation of CD8+ CTLs response to malignant cells [12].
into lymphoid tissue, although naïve T cell activation has However, this defect can be reversed by immunotherapy that
long been known to be mediated by DCs within the draining increases the number of cDCs within the tumor [12], under-
lymph node. Nonetheless, two recent investigations have scoring the critical role of cDCs in regulating CD8+ T cell
revealed that tumor-associated fluorescent proteins are function within tumors. Modulating cDC levels within local
actively conveyed by CD103+ conventional dendritic cells tumors may therefore be a promising therapeutic strategy.
(cDCs) that travel from the tumor to the lymph nodes in a
CCR7-dependent fashion [6, 9]. Notably, this phenomenon
occurs in both implantable and spontaneous tumor models, 3.3 Cross Talk of DC with
circumventing any experimental anomalies that may arise T Cells and NK Cells
when injecting significant amounts of dead or dying cells.
Another level of upregulation of anti-tumor immunity is to
modulate cDC and CD8+ CTL interaction/engagement. The
3.2 DC Plays Pivotal Roles in migration of cDCs to the location of T cells has been shown to
Tumor Immunity be critical to the induction of effective CTL mediated adaptive
immune response [13]. This essential step is dependent on the
Regulation of the function of dendritic cells (DCs) is crucial production of stimulatory cytokines and chemokines by cDCs
in establishing adaptive immunity against cancer, as evi- within the tumor microenvironment. Studies have shown that
denced by both animal models and human cancer patients. In chemokine production by cDCs, such as CXCL9 and
solid tumors, antigen uptake and presentation are mainly car- CXCL10, is essential for enhancing the efficacy of immuno-
ried out by monocytes and DCs. Although monocytes are the therapy. Along with chemokines, immune-related cytokines,
principal phagocytic cells in tumors, they are unable to acti- including IL-12 and IFNγ, are critical for promoting crosstalk
vate T cells in some cases and do not migrate to lymph nodes between CD8+ T cells and cDCs within the tumor microenvi-
[5]. Instead, monocytes often hinder T-cell responses against ronment. Interestingly, cDCs have also been shown to interact
Immunemodulation and Cancer 19

with Natural Killer (NK) cells, which play a crucial role in are critical in bridging innate and adaptive immune responses;
immune surveillance against cancer. Activated NK cells can specifically, TLRs play a critical role in stimulating DC mat-
recruit cDCs to infiltrate the tumor tissue. Analysis of gene uration, antigen uptake and presentation and the differentia-
signatures in human tumors has indicated that the presence of tion of T helper cells such as Th1, Th2 and Th17 and
NK cells is associated with the recruitment and appearance of controlling the suppressive function of regulatory T (Treg)
cDCs within the tumor microenvironment, suggesting that cells. In addition, signals via TLRs represent a potent means
modulation of NK cell ­presence within the tumor could sig- of modulating immune responses to cancer vaccines, a novel
nificantly enhance CTL-mediated immune responses [12, 14]. strategy now being evaluated in clinical trials. Vaccination
The requirement for interaction and cross-talk between cDC against cancer antigens largely relies on the use of DCs [16].
and other types of immune cells, particularly of CD8+ CTLs, As mentioned previously, DCs act as sentinels of the immune
demonstrates that the complexity of the antitumor immune system and play a crucial role in initiating and directing
response within the tumor and indicates that the migration/ immune responses. This is due to their ability to capture, pro-
localization of DCs and lymphocytes within the tumor is a key cess, and present self-tumor antigens to T cells and other
regulator of their function and activities. immune cells, ultimately leading to a potent and cancer-­
specific immune response capable of killing tumors.
However, it is widely acknowledged within the field that
4 Regulation DC by TLR Receptors tumor antigens alone are not strong enough to stimulate
APCs to induce clinically significant anti-tumor immune
4.1 Basic Biology of Toll-like Receptors responses, unless there is the involvement of professional
(TLRs) DCs to some extent [18]. As professional APCs, DCs express
a significant number of TLRs and possess powerful effects
Since activation of DC is central and induction of their on lymphocytes. TLRs can promote DC and immune
in vivo function is critical, therefore targeting DCs may pro- response, because DCs are at the interface of innate and
vide clinical approaches to improve immune responses in adaptive immune responses. However, in most cases, there is
cases where targeting T cells alone is not effective. Over the often lacking activation of DC, as a result of inhibitory sig-
past two decades, a significant advancement in DC activation nals from tumor cells—which may also induce immune tol-
has been the use of exogenous activation signals through erance through T cell deletion or through suppressive Tregs
Toll-like receptors (TLRs). TLRs function as pathogen pat- [19], thus helping tumors escape from immune surveillance.
tern recognition molecules that detect and initiate innate and Over the past decade, numerous efforts have been made to
adaptive immune responses against pathogens and malignant promote DC activation using agonists of the innate immune
cells, acting as a first line of defense against infectious dis- system. For instance, intratumoral injection of TLR agonists,
eases and cancer. Recognition of ligands by TLRs triggers alarmins, defensin peptides, DAMPs, or cytolytic peptides
the secretion of proinflammatory cytokines and promotes can be utilized to improve the in situ antitumor response. As
DC maturation programs for the induction of adaptive a result, many TLR agonists have been investigated in the
immune responses. To date, more than 12 TLRs have been context of cancer treatment and could serve as useful thera-
identified in both humans and animals. TLRs belong to peutic strategies aimed at enhancing DC function to bolster
type-1 integral membrane glycoproteins and are character- the antitumor response [16, 20].
ized by extracellular domains containing a variable number
of leucine-rich repeat motifs and cytoplasmic Toll/interleu-
kin (IL-1R homology domain). TLRs can recognize a highly 4.3 Antitumor Effect of TLR3
conserved sets of molecular structures, so-called pathogen-­
associated molecular patterns (PAMP), and/or damage asso- Among all the TLRs, TLR3 has been broadly studied for
ciated molecular patterns (DAMPs) such as RAGE and more than three decades. TLR3 is mainly expressed in neu-
HMGB, which leads to pathogen antigen uptake, processing, roectodermal and myeloid cells (including DCs). TLR3 of
and presentation to naive T cells in peripheral blood, lymph myeloid cells favorably acts as a receptor for the surface rec-
node, and tissues [15–17]. ognition of viral double-stranded RNA (dsRNA) [21]. The
effect of dsRNA as a TLR3 agonist in oncology has been
accessed by randomized clinical trials since the 1990s. Since
4.2 TLRs Play a Pivotal Role in Bringing then, many TLR3 agonists have been designed to mimic the
Innate and Adaptive Immunity dsRNA and have been studied, indicating that numerous
mechanisms subsidize the efficacy of TLR3 agonists. The
The recognition of PAMPs or DAMPs by TLRs leads to pro- TLR3 agonists, including RGC100, ARNAX, and polyino-
ducing proinflammatory cytokines, chemokines, type I inter- sinic: polycytidylic acid (poly-IC), are currently being exten-
ferons, and antimicrobial peptides 254,281. Therefore, TLRs sively studied. These agonists are used as adjuvants for
Another random document with
no related content on Scribd:
9. The connection of this verse also does not appear. According
to Targum (on verse 13) Jabez = Othniel, the nephew or brother of
Caleb.

And Jabez, etc.] Render, And Jabez came to be honoured


above his brethren, but his mother had called his name Jabez,
saying, Because I bare him with pain. Jabez suggests a
somewhat similar word spelt with the same consonants but in a
different order, and meaning “he giveth pain.” The stylistic similarity
between this verse and many passages in Genesis (e.g. Genesis
xxx.) cannot fail to be noticed, and it is important to observe that a
tradition of this popular character was current in the style of the
patriarchal narratives of Genesis in the Chronicler’s time. Compare
also vii. 23. The man with the ill-omened name staved off ill-fortune
by his prayer which is given in the following verse.

¹⁰And Jabez called on the God of Israel,


saying, Oh that thou wouldest bless me
indeed, and enlarge my border, and that thine
hand might be with me, and that thou
wouldest keep me from evil, that it be not to
my sorrow! And God granted him that which
he requested.
10. that thou wouldest keep me from evil] Literally that thou
wouldest make ... from evil. The Hebrew is therefore strange, though
not indefensible. Perhaps some words have fallen out, and it may
originally have read somewhat as follows, that thou wouldest make
[room (merḥābh) for me, and wouldest redeem (phādīthā) me] from
evil.

that it be not to my sorrow] or so that no pain befell me.

11‒15 for Chelub (verse 11) = Caleb (see on ii. 9, 42).


Sons of Caleb.
¹¹And Chelub the brother of Shuhah begat
Mehir, which was the father of Eshton. ¹²And
Eshton begat Beth-rapha, and Paseah, and
Tehinnah the father of Ir-nahash ¹. These are
the men of Recah.
¹ Or, the city of Nahash.

12. These are the men of Recah] Recah is unknown. Perhaps the
true reading is that of the LXX. (B), the men of Rechab, i.e.
Rechabite families; compare ii. 55, note.

¹³And the sons of Kenaz; Othniel, and


Seraiah: and the sons of Othniel; Hathath.
13. Othniel] Eponym of a Kenizzite clan, closely connected with
the Calebites. Compare note on i. 36, and Judges i. 13 and iii. 9‒11.

¹⁴And Meonothai begat Ophrah: and Seraiah


begat Joab the father of Ge-harashim ¹; for
they were craftsmen.
¹ Or, the valley of craftsmen.

14. Meonothai] perhaps a son of Othniel.

Ge-harashim] or, as margin, the valley of craftsmen. It is


mentioned Nehemiah xi. 35 along with Lod (the Lydda of Acts ix. 32)
and therefore was probably near Lydda.

¹⁵And the sons of Caleb the son of


Jephunneh; Iru, Elah, and Naam: and the
sons of Elah; and Kenaz.
15. Caleb the son of Jephunneh] Compare ii. 42, note.

the sons of Elah; and Kenaz] The sons of Elah and Kenaz seem
to be co-ordinated, as each represented a family descended from
Caleb; but perhaps the text is faulty.

¹⁶And the sons of Jehallelel; Ziph, and


Ziphah, Tiria, and Asarel.
16‒20. The connection of these names with Judah does not
appear. In accordance with the structure of this section (see note on
Carmi, verse 1) we might expect here to find “the sons of Hezron or
Perez”; and, since the relationship between Jehallelel (16) and Ezrah
(17) is not indicated, it has been suggested that we should insert in
verse 16 “And the sons of Perez and Jehallelel and Ezrah.” No
certainty can be felt in the matter.

Ziph] in south Judah, 1 Samuel xxiii. 15. Ziphah, a feminine form


of the same name.

¹⁷And the sons of Ezrah; Jether, and Mered,


and Epher, and Jalon: and she bare Miriam,
and Shammai, and Ishbah the father of
Eshtemoa.
17. and Jalon: and she bare Miriam] As the text stands she has
no antecedent. It has therefore been proposed to transfer from verse
18 the words And these are the sons of Bithiah the daughter of
Pharaoh, which Mered took, and put them after Jalon. Bithiah then
appears as the mother of Miriam, Shammai, and Ishbah, and the
difficulty of the absence of her sons’ names from verse 18
disappears. For father of Eshtemoa see ii. 24 note, and for
Eshtemoa see Joshua xxi. 14.
¹⁸And his wife the Jewess ¹ bare Jered the
father of Gedor, and Heber the father of Soco,
and Jekuthiel the father of Zanoah. And these
are the sons of Bithiah the daughter of
Pharaoh, which Mered took.
¹ Or, Hajehudijah.

18. his wife] the wife of Mered, if the transposition mentioned in


the last note be accepted.

his wife the Jewess] so called in contrast to his Egyptian wife.

Gedor] compare verse 4, where a different person is perhaps by


a different tradition called father of Gedor. Gedor is to be identified
with the ruins of Jedur on the road between Jerusalem and Hebron,
compare verse 39.

Soco ... Zanoah] The Soco (Joshua xv. 48) and Zanoah (Joshua
xv. 56) here mentioned were situated to the south-west of Hebron.
They are to be distinguished from a Soco near the valley of Elah
(Joshua xv. 35, 2 Chronicles xi. 7, xxviii. 18) and Zanoah near Beth-
shemesh (Joshua xv. 34).

these are the sons of Bithiah] See note on verse 17. A “daughter
of Pharaoh” is somewhat strange in such surroundings. For an
interesting emendation and interpretation, see Macalister, Palestine
Exploration Fund, Quarterly Statement, 1905, p. 252.

¹⁹And the sons of the wife of Hodiah, the sister


of Naham, were the father of Keilah the
Garmite, and Eshtemoa the Maacathite. ²⁰And
the sons of Shimon; Amnon, and Rinnah, Ben-
hanan, and Tilon. And the sons of Ishi;
Zoheth, and Ben-zoheth.
19. Keilah] a town of the Shephelah (Joshua xv. 44), the scene of
one of David’s exploits (1 Samuel xxiii. 1‒5).

Eshtemoa the Maacathite] The epithet distinguishes this


Eshtemoa from that of verse 17. The Maacathite may mean the
descendant of Maacah (ii. 48), the concubine of Caleb the brother of
Jerahmeel.

21‒23. These verses purport to give some fragmentary


information of the descendants of Shelah. It must be owned that they
are most obscure, and much more difficult than they appear at first.
In the Palestine Exploration Fund Statement, 1905, pp. 243 ff., 328
ff., R. A. S. Macalister gives a highly ingenious reinterpretation of this
passage in the light of certain jar handles inscribed with names
similar to those mentioned here and in certain other passages of the
Chronicler’s genealogies; and conjectures that throughout the
chapter we have a (mutilated) genealogy of a family of craftsmen,
tracing descent from two individuals, a Jerahmeel and a Caleb. In a
text so obscure it is no objection that the theory involves several
textual emendations, and is too intricate for further treatment here. It
is rejected by Driver, Modern Research, p. 77.

²¹The sons of Shelah the son of Judah; Er


the father of Lecah, and Laadah the father of
Mareshah, and the families of the house of
them that wrought fine linen, of the house of
Ashbea;
21. Shelah] mentioned in ii. 3 as a son of Judah. As there is no
reference to him in iv. 1, perhaps these difficult verses are an
addition. For other references to his descendants, see ix. 5,
Nehemiah xi. 5.
Mareshah] ii. 42; 2 Chronicles xi. 8; Joshua xv. 44 (mentioned
with Keilah). A town in the south of Judah.

the house of Ashbea] Nothing is known of such a family. Render


perhaps Beth-Ashbea, but the place is otherwise unknown.

²²and Jokim, and the men of Cozeba, and


Joash, and Saraph, who had dominion in
Moab, and Jashubi-lehem. And the records ¹
are ancient.
¹ Hebrew words.

22. and Joash, and Saraph, who, etc.] We find no other trace of
these two as rulers of Moab. The Targum fancifully identified them
with Mahlon and Chilion of Ruth i. 2, 4. For a conjecture as to the
cause of their presence in Moab, see Macalister, Palestine
Exploration Fund, Quarterly Statement, 1905, p. 340.

and Jashubi-lehem] render, with Vulgate, who returned to


Bethlehem, a translation which requires only an easy emendation of
the present Hebrew text.

²³These were the potters, and the inhabitants ¹


of Netaim and Gederah: there they dwelt with
the king for his work.
¹ Or, those that dwelt among plantations and hedges.

23. there they dwelt with the king for his work] In the days of the
kingdom the families were clients of the king and did his work;
compare 1 Kings vii. 46. The simplicity of this statement seems to
have been a stumbling-block to the early translators who
paraphrase; LXX., They were strong in his kingdom and dwelt there;
Targum, They made their dwelling there with the Shekinah of the
King of the World for the practice of the Law.

24‒27.
The Genealogy of Simeon.

²⁴The sons of Simeon; Nemuel ¹, and Jamin,


Jarib ², Zerah ³, Shaul: ²⁵Shallum his son,
Mibsam his son, Mishma his son. ²⁶And the
sons of Mishma; Hammuel his son, Zaccur his
son, Shimei his son. ²⁷And Shimei had sixteen
sons and six daughters; but his brethren had
not many children, neither did all their family
multiply, like to the children of Judah.
¹ In Genesis xlvi. 10, Exodus vi. 15, Jemuel.

² In Genesis xlvi. 10, Jachin. ³ Genesis xlvi. 10, Zohar.

24‒27. The sons of Simeon] With unimportant variations in the


names, the five sons of Simeon are mentioned also in Genesis xlvi.
10, Exodus vi. 15, and Numbers xxvi. 12, 13. A sixth son, Ohad, is
given by the list in Genesis. As the mother of one, Shaul, was a
Canaanitess (Genesis xlvi. 10) and Mibsam and Mishma are also
reckoned Ishmaelites (i. 29, 30), the tribe of Simeon contained
marked non-Israelite elements. This is the more natural by reason of
its location in the extreme south of Palestine.

27. six daughters] LXX. three daughters.

28‒33 (= Joshua xix. 2‒8).


The Territory of Simeon.
²⁸And they dwelt at Beer-sheba, and Moladah,
and Hazar-shual;
28. Beer-sheba] at the southern extremity of Palestine, as Dan
was at the northern (1 Samuel iii. 20).

²⁹and at Bilhah, and at Ezem, and at Tolad;


29. Tolad] In Joshua xix. 4 Eltolad.

³⁰and at Bethuel, and at Hormah, and at


Ziklag;
30. Bethuel] compare Genesis xxii. 22. In Joshua xix. 4 Bethul. In
1 Samuel xxx. 27 Bethel (not the famous Bethel near Jerusalem).

Hormah] Numbers xiv. 45, xxi. 3; Judges i. 17.

Ziklag] 1 Samuel xxvii. 6. The site is not certainly identified.

³¹and at Beth-marcaboth, and Hazar-susim,


and at Beth-biri, and at Shaaraim. These were
their cities unto the reign of David.
31. Beth-marcaboth ... Hazar-susim] perhaps royal chariot-cities,
1 Kings ix. 19. The names mean House of chariots and Court of
horses.

Shaaraim] 1 Samuel xvii. 52.

These were their cities unto the reign of David] either a reference
to David’s census; or else it is implied that these cities ceased to
belong to Simeon after David’s day. The clause breaks the
connection of verses 31, 32; and is perhaps a late gloss.
³²And their villages were Etam, and Ain,
Rimmon, and Tochen, and Ashan, five cities:
32. And their villages were Etam] more probably and their
villages (end of verse 31). Etam, etc. (continuing the list of cities as
in verse 31). By villages (Ḥăṣērīm) are meant small hamlets
dependent on larger towns and generally unwalled (Leviticus xxv.
31).

Ain, Rimmon] so also in the parallel passage, Joshua xix. 7; but


certainly only one place is meant, Ain-Rimmon (see the
commentaries on Joshua xv. 42 and xix. 7). The number of the cities
here ought therefore to be reckoned four, not five.

³³and all their villages that were round about


the same cities, unto Baal. These were their
habitations, and they have their genealogy.
33. unto Baal] Baal (“lord”) standing by itself is an unlikely name
for a town. Read Baalath-beer, Ramah of the South (“the mistress of
the well, the high place of the South”), as in Joshua xix. 8.

34‒43.
The Heroes of Simeon and their Exploits.

It seems probable that the exploits mentioned in these verses are


derived from some old, though obscure, tradition, and are therefore
of historical value for the movements of the tribe of Simeon.
Apparently we are to understand that in the time of Hezekiah a band
of the wild semi-nomadic tribe of Simeon made a successful raid
upon a fertile valley near Gerar (a correction for Gedor, see verse
39), a township on the Philistine border, taking by surprise its
peaceful population who were partly Canaanites, partly settlers who
had come originally from a place Maon (see verse 41). Gedor, the
reading of the Hebrew text, was a town just north of Hebron. A raid
by Simeonites on such a town is a startling, but not incredible,
episode in Hezekiah’s time; but see also note on verse 40. Finally
verses 42, 43 record a further assault by Simeonites, this time
against Edomite territory. For full discussion see Hogg in
Encyclopedia Biblia iv. 4527 ff.; also Macalister, Palestine
Exploration Fund Statement, 1905, 335 ff.

³⁴And Meshobab, and Jamlech, and Joshah


the son of Amaziah; ³⁵and Joel, and Jehu the
son of Joshibiah, the son of Seraiah, the son
of Asiel; ³⁶and Elioenai, and Jaakobah, and
Jeshohaiah, and Asaiah, and Adiel, and
Jesimiel, and Benaiah; ³⁷and Ziza the son of
Shiphi, the son of Allon, the son of Jedaiah,
the son of Shimri, the son of Shemaiah;
³⁸these mentioned by name were princes in
their families: and their fathers’ houses
increased greatly.
38. their fathers’ houses] See note on chapter v. 13.

³⁹And they went to the entering in of Gedor,


even unto the east side of the valley, to seek
pasture for their flocks.
39. the entering in of Gedor] Compare verse 18. The Gedor of
Joshua xv. 58 is identified with Jedur, Ijdur (north of Hebron,
Bädeker, Palestine⁵, p. 112). See Macalister, Palestine Exploration
Fund, Quarterly Statement, 1905, 335. LXX. has Gerar (compare
Genesis xx. 1; xxvi. 1), on the Philistine border.

⁴⁰And they found fat pasture and good, and


the land was wide, and quiet, and peaceable;
for they that dwelt there aforetime were of
Ham.
40. they ... of Ham] i.e. Canaanites, who had long been settled in
the district (compare the security felt by the people of Laish, Judges
xviii. 27). Their presence would seem natural in Gerar on the
Philistine border but strange in Gedor near Hebron. If therefore the
reading Gedor be preferred above, there is something to be said for
the suggestion of Macalister (p. 335) that we should here read “of
Menahem” (a change of one letter in Hebrew), some unwarlike
Hebrew family, perhaps potters.

⁴¹And these written by name came in the days


of Hezekiah king of Judah, and smote their
tents, and the Meunim that were found there,
and destroyed ¹ them utterly, unto this day, and
dwelt in their stead: because there was
pasture there for their flocks. ⁴²And some of
them, even of the sons of Simeon, five
hundred men, went to mount Seir, having for
their captains Pelatiah, and Neariah, and
Rephaiah, and Uzziel, the sons of Ishi.
¹ Hebrew devoted them.

41. written by name] Apparently those mentioned in verses 34‒


37; but the names there are of a late character, and have perhaps
been artificially connected with the old tradition of the raiders in
verses 41 ff.

in the days of Hezekiah] The Hebrew is ambiguous, but the


clause should probably be connected with the verb “came,” not with
“written”: the raid, not the record, was made in the days of Hezekiah.
and the Meunim] Here, and in 2 Chronicles xx. 1 (see note), xxvi.
7, the LXX. has Minaeans, an Arabian people who from the 8th or
9th century b.c. or perhaps much earlier exercised great authority in
South Arabia (see Encyclopedia Britannica¹¹ II. 264). The Meunim of
the present passage are to be connected with an Edomitic city or
tribe not far from Petra, south of the Dead Sea; or (so Macalister, p.
336) are simply the people of Maon, a township near Hebron in
Judah. 2 Chronicles xx. 1 (note) and xxvi. 7, Revised Version.

destroyed them utterly] or, as margin, devoted them (compare


Joshua vi. 18, 21, Revised Version). See note 2 Chronicles xx. 23.

⁴³And they smote the remnant of the


Amalekites that escaped, and dwelt there,
unto this day.
43. the remnant of the Amalekites] i.e. the descendants of those
who had escaped the attacks of Saul and David (1 Samuel xiv. 48,
xv. 3 ff.). They had apparently found refuge in some part of the
Edomite territory, for mount Seir is a synonym for the land of Edom.
Chapter V.
1‒10.
The Genealogy of Reuben.

¹And the sons of Reuben the firstborn of


Israel, (for he was the firstborn; but,
forasmuch as he defiled his father’s couch, his
birthright was given unto the sons of Joseph
the son of Israel; and the genealogy is not to
be reckoned after the birthright.
1. he defiled] Genesis xxxv. 22, xlix. 4.

his birthright was given unto the sons of Joseph] Compare


Genesis xlviii. 5, “Ephraim and Manasseh, even as Reuben and
Simeon, shall be mine,” words of Jacob which might be interpreted
to mean that the rights of the firstborn were to pass from Reuben
and Simeon to Ephraim and Manasseh, the sons of Joseph.

the genealogy is not to be reckoned after the birthright] i.e.


though the birthright of Reuben has been given to Joseph, yet the
genealogy of Joseph is not to be given before that of Reuben. Verse
2 intimates that, though Joseph possessed the birthright, Judah had
a primacy as supplying the royal family. In this confusion of claims
the natural order is followed and the genealogy of Reuben is given
first.

²For Judah prevailed above his brethren, and


of him came the prince ¹; but the birthright was
Joseph’s:)
¹ Or, leader.

2. Judah prevailed above his brethren] Compare Genesis xlix. 8


(Jacob to Judah) “Thy father’s children shall bow down before thee.”

the prince] The Hebrew word is nāgīd. The immediate reference


is to David (Saul being virtually ignored by the Chronicler).

³the sons of Reuben the firstborn of Israel;


Hanoch, and Pallu, Hezron, and Carmi.
3. the sons of Reuben] The same four names (with one
unimportant variation in spelling in Authorized Version) appear
Genesis xlvi. 9; Exodus vi. 14.

Hanoch] the correct spelling of the familiar name Enoch; compare


i. 3.

Hezron, and Carmi] also given as sons of Judah: for Hezron,


compare ii. 5 (note); for Carmi ii. 7, iv. 1.

⁴The sons of Joel; Shemaiah his son, Gog his


son, Shimei his son; ⁵Micah his son, Reaiah
his son, Baal his son;
4. sons of Joel] the connection, if any, with the preceding verse is
not known.

⁶Beerah his son, whom Tilgath-pilneser ² king


of Assyria carried away captive: he was prince
of the Reubenites. ⁷And his brethren by their
families, when the genealogy of their
generations was reckoned; the chief, Jeiel,
and Zechariah,
² In 2 Kings xv. 29, xvi. 7, Tiglath-pileser.

6. Beerah ... captive] The deportation of Beerah is recorded only


here, but there is no reason why the tradition should not be
historical. The information is of very slight importance in itself, but
since there is absolutely no motive to make the Chronicler invent the
statement, this is precisely the kind of notice which implies that he
had some sources oral or traditional at his command outside the
canonical material (see Introduction. § 5).

Tilgath-pilneser] called Tiglath-pileser (2 Kings xv. 29), and no


doubt identical with Pul (see below, verse 26).

⁸and Bela the son of Azaz, the son of Shema,


the son of Joel, who dwelt in Aroer, even unto
Nebo and Baal-meon: ⁹and eastward he dwelt
even unto the entering in of the wilderness
from the river Euphrates: because their cattle
were multiplied in the land of Gilead.
8. Aroer] now ‘Ar‘āir, a heap of ruins near the wādy Mojib, i.e. the
Arnon (Joshua xii. 2). According to Joshua xiii. 9‒16 it passed from
Sihon king of the Amorites into the hands of the Reubenites. See
Bädeker, Palestine⁵, p. 153.

Nebo and Baal-meon] A line drawn due north from Aroer (see
last note) passes close first to Ma‘in (which may be Baal-meon) and
then to Jebel Nebā, which evidently preserves the name of Mount
Nebo.

Baal-meon] called more correctly Beth-baal-meon Joshua xiii. 17.


¹⁰And in the days of Saul they made war with
the Hagrites, who fell by their hand: and they
dwelt in their tents throughout all the land east
of Gilead.
10. in the days of Saul] Saul’s victory over the Ammonites (1
Samuel xi.) may have paved the way for the expansion of Israel east
of Jordan, but see also the note on verse 19.

the Hagrites] compare Psalms lxxxiii. 6 (Revised Version margin).


They were an Arab people. See further the note on verse 19.

the land east of Gilead] i.e. the land between Gilead and the
Euphrates (compare verse 9). On Gilead, see note, ii. 22.

11‒17.
The Genealogy and Settlements of Gad.

¹¹And the sons of Gad dwelt over against


them, in the land of Bashan unto Salecah:
11. Bashan] the wide district extending from the Jabbok on the
south to Hermon in the north and from the Sea of Galilee on the west
to the mountains of Hauran on the east (compare verse 23). Remark
that in Numbers xxxii. 33; Deuteronomy iii. 12; Joshua xiii. 25, Gad is
located in Gilead (south of Bashan) where the Chronicler has placed
Reuben.

Salecah] is probably represented at the present day by the ruins


of Salkhad due south of the Jebel Hauran and almost due east of
Boṣra.

¹²Joel the chief, and Shapham the second,


and Janai, and Shaphat in Bashan: ¹³and their
brethren of their fathers’ houses; Michael, and
Meshullam, and Sheba, and Jorai, and Jacan,
and Zia, and Eber, seven. ¹⁴These were the
sons of Abihail the son of Huri, the son of
Jaroah, the son of Gilead, the son of Michael,
the son of Jeshishai, the son of Jahdo, the son
of Buz; ¹⁵Ahi the son of Abdiel, the son of
Guni, chief of their fathers’ houses.
12. and Janai, and Shaphat] LXX. “Janin the scribe”; Targum,
“Janai the judge.”

of their fathers’ houses] This rather awkward phrase means clans


or patriarchal families (πατριαί). Sometimes it is used to denote the
whole tribe, compare Numbers xvii. 17.

¹⁶And they dwelt in Gilead in Bashan, and in


her towns ¹, and in all the suburbs ² of Sharon,
as far as their borders ³.
¹ Hebrew daughters. ² Or, pasture lands

³ Hebrew goings forth.

16. in Gilead in Bashan] a contradictory phrase, since Gilead


means the southern and Bashan the northern part of Israel’s trans-
Jordanic territory. Perhaps in Bashan is here an addition made by
the Chronicler or a later hand to harmonise in Gilead (the territory
usually assigned to Gad—see note on verse 11 above) with verses
11 and 23. The emendation “in Gilead in Jabesh” has been
suggested.

Sharon] some place, unidentified, to the east of Jordan. LXX. (B)


has Sirion. (The well-known Sharon lay in the maritime plain
between Joppa and Caesarea.)
¹⁷All these were reckoned by genealogies in
the days of Jotham king of Judah, and in the
days of Jeroboam king of Israel.
17. reckoned by genealogies ... and in the days of Jeroboam]
“Reckoning by genealogy” is a phrase used only in the writings of the
Chronicler (Chronicles, Ezra, Nehemiah), but the practice probably
resembled what is called in other books “numbering the people”: see
the example in Nehemiah vii. 5‒65. The object however was
different and corresponded with the circumstances of the returned
exiles, who found themselves in the midst of a Gentile population in
Judea. The people were “reckoned by genealogy” not so much to
take a census of them, as to inquire into the purity of their Israelite
descent. The ancient term “numbering” would be a more suitable
description of a transaction belonging to the days of Jotham. For
Jotham see 2 Chronicles xxvii. and for Jeroboam 2 Kings xiv. 23‒29.
The last years of the reign of Jeroboam II probably synchronized
with part of the reign of Jotham.

18‒22.
The War of the Trans-Jordanic Tribes against Arabian
Tribes.

¹⁸The sons of Reuben, and the Gadites,


and the half tribe of Manasseh, of valiant men,
men able to bear buckler and sword, and to
shoot with bow, and skilful in war, were forty
and four thousand seven hundred and
threescore, that were able to go forth to war.
18. skilful in war] compare xii. 8, 21.

forty and four thousand] According to Joshua iv. 13 “about forty


thousand” from these tribes crossed the Jordan with Joshua.
¹⁹And they made war with the Hagrites, with
Jetur, and Naphish, and Nodab.
19. made war with the Hagrites...] Possibly this war described in
verses 18‒22 is only a midrashic variation of the war briefly noted in
verse 10; possibly we have traditions of separate conflicts. That
fighting against Arabian tribes took place in the time of Saul (verse
10) is quite probable, and perhaps there is here a definite
recollection of the fact. Yet conflicts on the eastern borders were no
doubt frequent, and the ancient fights may have been overlaid with
the memories and details of more recent struggles, and especially
the names assigned may be those of later foes. Though the Hagrites
are mentioned in inscriptions of Sennacherib, they seem to have
grown more prominent and powerful in later days (compare Psalms
lxxxiii. 7), and their name in the Chronicler’s time may have been
somewhat loosely and generally applied to the Arabian tribes near
Palestine.

Jetur, and Naphish, and Nodab] Other Bedouin tribes, Jetur,


Naphish, Kedemah are given as sons of Ishmael in i. 31.

²⁰And they were helped against them, and the


Hagrites were delivered into their hand, and all
that were with them: for they cried to God in
the battle, and he was intreated of them;
because they put their trust in him. ²¹And they
took away their cattle; of their camels fifty
thousand, and of sheep two hundred and fifty
thousand, and of asses two thousand, and of
men ¹ an hundred thousand.
¹ Hebrew souls of men.
20. they were helped] with Divine assistance; compare xv. 26.

²²For there fell many slain, because the war


was of God. And they dwelt in their stead until
the captivity.
22. was of God] i.e. was prompted by God; compare 1 Samuel
xv. 2, 3.

23, 24.
The Half Tribe of Manasseh.

²³And the children of the half tribe of


Manasseh dwelt in the land: they increased
from Bashan unto Baal-hermon and Senir and
mount Hermon. ²⁴And these were the heads of
their fathers’ houses; even Epher, and Ishi,
and Eliel, and Azriel, and Jeremiah, and
Hodaviah, and Jahdiel, mighty men of valour,
famous men, heads of their fathers’ houses.
23. For the genealogy of Manasseh, see vii. 14 ff.

Baal-hermon] In Judges iii. 3 a mount Baal-hermon is mentioned.


Here probably a city is meant, possibly Banias, on the eastern slope
of Hermon.

Senir] an Amorite name for Mt Hermon or a peak of the range


(Deuteronomy iii. 9).

25, 26.
The Captivity of the Trans-Jordanic Tribes.

You might also like