Skip to product information
The Minimum You Need to Know to Understand Immunotherapy
The Minimum You Need to Know to Understand Immunotherapy
Description
Book Introduction
It contains the history of immunotherapy from a frame perspective, the immune system understood from a perspective of balance, the mechanisms of immunotherapy, the achievements of immunotherapy and the issues raised in the process of application, an introduction and analysis of the latest research trends, and the 'minimum things you need to know to understand immunotherapy' surrounding all of this.
  • You can preview some of the book's contents.
    Preview

index
Acknowledgements 002

I.
Prologue: Trend 013: Immunotherapy


II.
Start 021

The Age of Swords and Poison 023 / A Slightly Reckless Beginning 028 / Ignorance, Conservatism, and Failure 033

III.
The Minimum You Need to Know About Immunotherapy 037

Innate and Adaptive Immunity 039 / Immune Memory 043 / Dendritic Cells 044 / Cancer-Immune Cycle 046

IV.
Immunotherapy using toxic lymphocytes 051

Toxic lymphocytes 053 / Hematopoietic stem cell transplantation 054 / Tissue typing 062 / T-cell receptors and antigen specificity 065 / Use of in vitro cultured T cells / Immuno-oncology 070 / TCR-T cell therapy 073 / CAR-T cell therapy 075 / Current status and prospects of T-cell therapy 083 / NK cell therapy 094 / γδ T cell therapy 102

V.
Immune Checkpoint Inhibitors 1: Visible Results 109

Direction and Balance 111 / CD28 and CTLA-4: The Accelerator and Brake of T Cells 113 / Rational Judgment and a Shift in Thinking 116 / Yervoy 118 / Opdivo 124 / Keytruda 129 / Progress and Limitations 136

VI.
Immune Checkpoint Inhibitors 2: Incomplete Mechanisms 139

Anti-CTLA-4: Eliminating Regulatory T Cells? 143 / Antibody Fc 144 / Are Multiple Anti-PD-1 and PD-L1 Antibodies Equally Effective? 148 / PD-1 and PD-L1 Antibody Therapeutics: Overcoming Adaptive Resistance? 152 / Clinicians in Conflation 155

VII.
Biomarker 159

PD-L1 161 / Tumor-infiltrating T cells 163 / Tumor mutational burden 165 / Blood 170 / Microbiota 171

VIII.
Combination therapy 1: Activation of the innate immune system 175
Blind(?) combination therapy 177 / Immunogenic cell death 182 / Dendritic cells in tumors 186 / CD47: immune checkpoint of macrophages 188 / Neoantigens 192 / Cancer vaccines 197 / Oncolytic viruses 205

IX.
Combination Therapy 2: Overcoming Intratumoral Immunosuppression 209

Chemokines 211 / Normalization of tumor blood vessels 213 / Extracellular matrix of tumor tissue 217 / Immunosuppression in tumors 218 / Immunosuppressive cells in tumors 219 / Surface area similarity between regulatory T cells and T cells 222 / Metabolic activity in tumors 225 / Positive costimulatory agents 227 / Bispecific antibodies 231 / Cytokines 235 / NKG2A: immune checkpoint for NK cells and T cells 236

X.
Epilogue 239


Reference 243

Into the book
It is clear that efforts prior to 2010 laid the foundation for immunotherapy for cancer.
However, the process is largely remembered as a dark history.
Although outstanding scholars did their best to research, there was a problem with the frame.
First of all, there was a problem with the frame of immunity itself.
Until at least the 1990s, immunity was thought to be a system for responding to invasion by foreign substances such as viruses and pathogens.
That is, 'self vs. non-self'.
It is a frame of ‘non-self’.
Within this frame, cancer cells are the result of cells in the body becoming transformed.
The premise was that cancer cells were 'self' and had nothing to do with immunity.
Although cancer is a vicious lump that needs to be eliminated, I couldn't grasp the concept that 'it can be controlled by the immune system.'
The limitations of the concept led to limitations of the research.
The conservative frame also played a part.
In the early 2000s, clinical trials of immuno-oncology drug candidates, such as immune checkpoint inhibitors and cancer vaccines, were conducted primarily by small-scale bio-ventures and biotech companies.
This is because the interest of large pharmaceutical companies at the time, which were at the forefront of new drug development, was in targeted anticancer drugs.

Before the advent of targeted anticancer drugs, the mechanism of anticancer drugs was to kill cancer cells with toxic chemicals.
The drugs were developed primarily using toxic chemicals that responded to the rapid growth characteristics of cancer cells, but this method caused significant side effects as it also caused significant damage to normal cells.
However, targeted anticancer drugs target specific molecular characteristics of cancer cells.
Therefore, it is possible to selectively eliminate only cancer cells while minimizing side effects on normal cells.
Gleevec, which opened the era of targeted anticancer drugs with FDA approval in 2001, was successful as a treatment for chronic myeloid leukemia (CML).
However, although targeted anticancer drugs were also born from innovative science, their superior efficacy has again led to conservatism.
Everyone was only looking at research on targeted anticancer drugs, and immunotherapy was pushed out of the spotlight.
--- p.33~35

In the 1950s, there were quite a few patients whose blood cells died due to radiation exposure from nuclear power plant accidents or excessive exposure to toxic anticancer drugs or radiation during cancer treatment.
Dr. Edward Donnall Thomas (1920-2012) confirmed that bone marrow contains cells capable of producing blood cells and studied methods for treating patients using bone marrow transplants.
It was later discovered that hematopoietic stem cells produced in the bone marrow play a major role in the regeneration of blood cells such as white blood cells, red blood cells, and platelets.
Hematopoietic stem cells are also called hematopoietic stem cells because they have the ability to differentiate into various cells and to replicate themselves.
Thomas initially studied hematopoietic stem cell transplantation from a regenerative medicine perspective.
The idea was to transplant healthy hematopoietic stem cells to regenerate the bone marrow, which was damaged by radiation exposure and causing the blood cells to die.
Thomas saved the lives of many patients through hematopoietic stem cell transplantation, and was awarded the 1990 Nobel Prize in Physiology or Medicine in recognition of his contributions.
Hematopoietic stem cell transplantation is considered the origin of not only regenerative medicine but also immunotherapy.
It was thought that hematopoietic stem cells were transplanted, but in reality, various immune cells in the bone marrow were transplanted together.
Controlling the function of transplanted immune cells has become a key factor in successful hematopoietic stem cell therapy, and the technology and know-how developed in the process have since become key elements in the development of cancer treatment technologies using immune cells.
So the Fred Hutchinson Cancer Research Center in Seattle, where Thomas spent most of his research time, also played a key role in the development of immuno-oncology cell therapy technologies.
--- p.54~56

The important things in the immune system are direction and balance.
When we learn that immunity is what can fight against a powerful enemy like cancer, it is common to wonder, "How powerful is the power of immunity?"
It's important that our immune system is strong enough to overcome cancer, but more importantly, when it's out of balance and out of control, it can attack the wrong parts of our body with equal force.

When your immune system is suppressed, you become more susceptible to illness when exposed to infections and even cancer.
However, excessive activation of the immune system is also a problem.
This is because it can cause autoimmune diseases, allergies, and immune hypersensitivity such as asthma, in which the body's immune cells attack the body.
Immune checkpoints are what put the brakes on when the immune system becomes overly activated, losing direction and balance.

But clever(?) cancer uses the immune checkpoint for its own benefit.
It is to avoid an attack by putting the brakes on the immune system.
Immune checkpoint inhibitors are anticancer drugs that enhance the immune anticancer response by preventing the immune system from braking.
There are two places in the cancer-immune cycle where immune checkpoint inhibitors act.
The process of activating T cells in the lymph nodes (3) and the process of sending a signal to the cancer cells not to attack them, preventing the immune cells from killing the cancer cells (7).
--- p.111~113

All the biomarkers discussed so far were measured using tumor tissue.
Tumor tissue is rich in information, but difficult to obtain.
If the patient had surgery, they could have obtained a fairly large tumor tissue.
However, if tumor tissue is obtained for biomarker testing, only a minimal amount of tissue is extracted.
However, since tumors are not uniform across different parts, the question arises as to how much information the extracted tumor tissue can represent about the entire tumor.
For this reason, efforts to develop blood-based biomarkers are active.
Blood, unlike tumor tissue, can be obtained at any time.
This is a good way to look at changes over time.
First, we can imagine a way to utilize various immune cells in the blood.
You may also be able to see the number of cells of a particular type, the ratio of two types of cells, etc.
Meanwhile, by comparing the patient's blood before and after receiving immunotherapy, it may be possible to see changes in immune cells due to treatment.
Biomarker development is currently underway, utilizing the differences in blood immune cells between responders and non-responders to immunotherapy.
Next, there is a method of analyzing small amounts of circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and exosomes in the blood using liquid biopsy.
The only problem is that the quantity is too small.
For example, there is about 1 CTC in the blood per million immune cells.
It is technically difficult to extract and analyze such a small amount of material.
Nevertheless, technological advancements have made it possible to test for TMB and MSI using liquid biopsies.
The efficacy in clinical trials is also in the verification stage.
--- p.170~171

Several phagocytes engulf dead tumor cells.
The subsequent immune response may also vary depending on which phagocytes are involved.
There are several types of macrophages and dendritic cells within tumor tissue.
Most of these enter the tumor tissue through the bloodstream.
They interact with various cells in tumor tissue and differentiate into cells with independent functions.
Among the various types of macrophages, some trigger an immune response against tumors, while others induce immunosuppression against tumors.
The origin, classification, and function of various intratumoral macrophages are currently being studied.
Here, we will briefly look at cDC1 (conventional Dendritic cell 1), which is receiving the most attention in the field of immuno-oncology drug development using dendritic cells.
cDC1 expresses the CD141 (CD103 in mice) receptor along with the transcription factor Batf3 (Basic leucine zipper ATF-Like transcription factor 3).
According to the research team led by Dr. Matthew Krummel of California State University, cDC1 carries tumor tissue information and moves to the lymph nodes to activate naive T cells (Cancer Cell, 2016) and also reactivates effector T cells in tumor tissue to promote cancer cell death (Cancer Cell, 2014). The fact that cDC1, which expresses textbook dendritic cell functions, is given such importance can be said to reveal an abnormal aspect of tumor tissue.
Although the number of cDC1 is usually low in solid tumors, it plays an important role in the prognosis of cancer patients and the response to immunotherapy.
So what factors regulate the number of cDC1s within tumors? --- p.186~187

Tumor tissues contain various immunosuppressive cells, including tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs), myeloid-derived suppressor cells (MDSCs), and regulatory T cells.
Among these, TAM and CAF are cells that exist in normal tissues but have been transformed by tumors.
While macrophages engulf and destroy tumor cells, TAMs promote tumor development and suppress immunity.
Macrophages can be polarized into M1, which activates the immune response, and M2, which suppresses the immune response and promotes tissue regeneration.
When a tissue is infected, M1 must first be activated to eliminate the pathogen through a strong immune response.
During this process, tissues and normal cells are also damaged.
Therefore, in the later stages of the inflammatory response, M2 alleviates inflammation and regenerates tissue.
In this way, the activation and suppression of immunity must be appropriately regulated according to the situation.
In 1986, Dr. Harold Dvorak of Harvard Medical School published an article in the New England Journal of Medicine comparing tumors to a "wound that does not heal."
Wounds that do not heal cause chronic inflammation.
Chronic inflammation exhausts T cells.
Macrophages are polarized to M2 and suppress immunity.
Fibroblasts, which produce ECM to repair wounds in normal tissues, become CAFs in tumor tissues and continuously produce ECM to treat non-healing wounds, thereby inducing fibrosis.
Fibrosis prevents T cells from reaching tumor tissue.
In addition, CAFs secrete several immunosuppressive substances, thereby interfering with antitumor immunity.
Looking at the wounds that do not heal, many of the immunosuppressive properties of tumors can be explained as follows.
--- p.219~221

Publisher's Review
An engineering professor studying immunity and cancer

The American Association for Cancer Research (AACR) is a global cancer society that has been active for over 100 years.
Tens of thousands of people, including doctors, researchers, and pharmaceutical industry professionals fighting cancer around the world, are members.
They gather once a year at an annual conference to present and discuss their research, and the booklet listing the presentations and seminars held over the course of about a week is well over 200 pages long.
When four surgeons, five pathologists, and two biochemists founded the AACR in 1907, did these 11 founding members ever imagine the society would grow to such prominence a century later? Or even that a complete cure for cancer would still elude them?
When life expectancy was not long, cancer was a disease that people got unlucky with.
But now, cancer has changed its status to a disease that people encounter at least once in their lifetime.
When your status changes, your treatment also changes.
Scientists are rushing forward with cutting-edge science to develop cancer treatments.
And the representative protagonist born in that process is ‘immunotherapy’.

In 2011, the U.S. FDA approved Yervoy (ingredient name: Ipilimimab), an immune checkpoint inhibitor, as a cancer treatment.
Four years later, Opdivo (Nivolumab), an immune checkpoint inhibitor with a similar function, also received FDA approval.
Opdivo is prescribed to patients with melanoma, non-small cell lung cancer, kidney cancer, bladder cancer, etc., and the number of cancers for which it can be prescribed continues to increase.
As of 2018, Opdivo was sold for approximately $6.7 billion and Yervoy for approximately $1.3 billion.
Money doesn't tell the whole story, but it's clear that immunotherapy is one of the hottest areas in the science of developing cancer treatments.
This book, "The Minimum You Need to Know to Understand Immunotherapy" (Scientist's Writing 2), provides a comprehensive map of the history, concept, current status, and future prospects of this hot topic of immunotherapy.
The person who drew the map is neither a doctor nor a life science or biotechnology expert.
I majored in materials engineering and am currently a professor of engineering teaching materials engineering at a university.


Performance at different angles
Insight without preconceptions


Dojun Sang is well-known among researchers studying immunotherapy for his consistent participation in academic conferences, large and small, held around the world on the topics of 'immunity' and 'cancer', such as the Immunotherapy of Cancer Conference and the Natural Killer Cell Symposium.
Since he is an engineering professor, you might misunderstand that he has nothing to do with immunity or cancer.
However, he has been working on integrating immunology and engineering since he received his doctorate in 2002 and until now in 2019.
He describes himself as an engineering professor specializing in immunology and is also the co-author of Microfluidics in Cell Biology Part A: Microfluidics for Multicellular Systems (Academic Press, 2018), published in the United States.
Perhaps it's more unusual to wonder about the context when an engineering professor studies immunology and writes a book on immunotherapy.
If learning, studying, and research are processes of finding answers, then what matters is what the questions are and how the answers are found.
This book is a work by a researcher who is an engineering professor, who seeks answers to questions about immunology and immuno-oncology, and seeks to share with the public the knowledge he has gained.
Insights from different angles and unbiased perspectives are actually good conditions for focusing on problems, questions, and answers.


To understand immunotherapy
The bare minimum you need to know


As the title suggests, the contents of the book are 'the minimum things you need to know to understand immunotherapy.'
First, we begin by outlining the history of immunotherapy.
But this is not a long-winded timeline.
Instead, we will focus on the development of immune-oncology drugs that are evolving around the change in the ‘frame.’
Knowing about the existence of immunity and attempting to treat cancer with immunity has been around for over 100 years.
But this was a non-mainstream trend.
It has only been about 20 to 30 years since actual immunotherapy has begun to attract public attention.
The immune system is a mechanism that protects the body from foreign substances, called 'self vs.
According to the 'non-self frame', cancer is not a foreign substance and therefore is not under the jurisdiction of the immune system.
Long-standing self vs.
The non-self frame ended up preventing people from showing interest in immunotherapy.
This is because it is no different from hindering the development of immuno-oncology drugs until the efforts of a few researchers and the results of chance come out.
Do Jun-sang presents the 'fear of frames' as the first of the minimum things to know to understand immunotherapy.
Even a single frame error could misdirect the progress made in immunotherapy to date.

Immunity is still an area that requires more research.
Even life science majors find it difficult to understand the complex mechanisms of the immune system that have been discovered to date.
So, in order to not deviate from the standard of 'the minimum things you need to know to understand immunotherapy,' this book guides you to the minimum immunity you need to know to understand immunotherapy.

Next, we introduce the ideas, challenges, failures, and processes of overcoming these challenges and moving on to the next stage of immunotherapy that have been attempted so far.
In this process, the cancer treatment principles of currently prescribed immunotherapy drugs such as Yervoy, Opdivo, Keytruda, and Tecentriq are naturally explained.
This also tries to adhere to the standard of 'the minimum things you need to know to understand immunotherapy'.
As interest in immunotherapy grows, attempts to explain it are increasing.
However, because it is too difficult, there is a tendency to replace the fundamental mechanisms with inappropriate metaphors, or to fill it with terms and data that are difficult even for experts to understand in the hope of explaining everything.
This book attempts to find a place somewhere in between, precise but appropriate.

The achievements and limitations of immunotherapy cannot be left out of 'The Minimum Things You Need to Know to Understand Immunotherapy.'
And the achievements and limitations are organized from the perspective of ‘direction and balance.’
If immunotherapy is going to exploit the immune system, we need to look at what the immune system is trying to achieve and where it might fail.
The logic that you need to use equally virulent methods to fight a tough opponent like cancer may not be suitable for immunotherapy.
The beauty of the immune system is that it goes straight to where the problem is and restores the balance that has been disrupted.
From this perspective, we can interpret the performance and limitations of immunotherapy drugs currently being prescribed or studied.
The analysis is that where there is success, there is a well-defined direction and balance, and where limitations appear, there is confusion about direction and balance.

Finally, we consider the future prospects of immunotherapy.
It also presents ideas on how to manage biomarkers to enable more precise prescribing to more types of cancer and more patients, and to increase efficiency and effectiveness in the research and development of immuno-oncology drugs.
We also do not forget to analyze and provide suggestions on the phenomenon of clinical trials focusing on combination immunotherapy drugs, which sometimes end up being discontinued due to a lack of subjects to receive the drugs.
GOODS SPECIFICS
- Publication date: December 13, 2019
- Page count, weight, size: 260 pages | 113*188*20mm
- ISBN13: 9791196079369
- ISBN10: 1196079366

You may also like

카테고리