top of page

A Dynamic Duo: How CRISPR is revolutionizing CAR T Cell therapy

Leilani Pradis

Source: Christoph Burgstedt, 2017

CAR T-Cell Therapy

The immune system fights thousands of diseases every day by locating antigens,

which are proteins that elicit an immune response. These could be viruses, harmful

bacteria, and even cancer cells. However, some immune cells do not possess the right

receptors to attach to the cancer cells, preventing the immune system from demolishing

them [1].


Officially approved by the FDA in 2017, a team of researchers in the Perelman

School of Medicine at the University of Pennsylvania produced the first treatment that

harnesses the power of the immune system in defeating cancerous cells, called CAR

T-cell therapy.


CAR (chimeric antigen receptor) T-cell therapy is a type of treatment that utilizes

a patient's own immune system to fight cancer. Specifically, an individual’s T-lymphocyte

cells, which are a type of white blood cell, are manipulated in a way that causes them to bind to and destroy cancer cells. The T cells are collected and taken to a laboratory,

where a chimeric antigen receptor gene is inserted into them. These immune cells are

placed back into the patient, where they will bind to the specific antigen of the cancer

cell and hopefully kill them [2].


Currently, this immunotherapy is primarily used to battle hematologic (blood)

cancers and has had promising results. Due to the recent discovery and approval to use

CAR T-cell therapy, there are very few patients to study the long-term effects. However,

it is worth mentioning that two patients participated in a clinical trial years ago (achieving

remission in 2010), with the results being keeping their chronic lymphocytic leukemia in

remission for more than a decade [3]. This feat has further raised hopes about the

effectiveness of the treatment.


Nonetheless, there are still many limitations. Shortly, I will go over some major

limitations that can be addressed by CRISPR/Cas9 technology.


CRISPR/Cas9

CRISPR/Cas 9 is a breakthrough gene-editing tool that could be the key to

relieving many genetic diseases. CRISPR is an acronym for “clustered regularly

interspaced palindromic repeats”, which are repeating sequences in prokaryotic DNA

that serve a purpose in the organism's defense system. Cas9 stands for “CRISPR-associated protein 9”, which is an endonuclease that will make a cut on both strands in the target DNA. Briefly, this is how it works: there is a guide RNA that directs Cas9 to the target location on the DNA that will be cut. This cut allows the DNA to then be manipulated, commonly through what is called homology-directed repair. This type of repair uses a similar piece of DNA that includes any desired edits and is incorporated into the original DNA [4]. Two women made significant contributions to developing this mechanism, Jennifer Doudna and Emmanuelle Charpentier, for which they were awarded the 2020 Nobel Prize in Chemistry.


CRISPR/Cas9 is exciting because there are many things it can do, with one of its

applications to be used in a general lab setting, where it can efficiently create

animal/cell models that can be used for research like never before. Another use is

through plant science, where genetic modifications have increased bacterial resistance and even increased yield and nutrition in crops. In animal science, the tool has been

used to increase health elements in products produced by animals, such as milk, and in

research regarding pig xenotransplantation. Lastly and most relevant to this article is its

use in human science and medicine. The pair can help treat HIV, sickle cell anemia, and

even certain cancers directly [5].


Despite its vast potential, the genetic editing technology is not free from ethical

issues and debates surrounding its use. There is a great concern about the “disruption

of ecological balance” when pertaining to the use on animals, plants, and bacteria [5]. It

becomes more complicated with humans, with points bringing up legislation, safety, and

eugenics, to name a few.


How CRISPR/Cas9 can optimize CAR T-Cell Therapy

One of the major limitations of CAR T-cell therapy is cell exhaustion. This is a

state where the T-cell is no longer working efficiently, which in this case is caused by

persistent antigen stimulation. Specifically, the T-cell will not fight the cancer cells as

well (as it will have decreased cytotoxicity- that is a lower ability to be ‘toxic’ towards the

cancer cells) and tend to express proteins such as PD-1 (programmed cell death protein

1), amongst other inhibitory receptors [6]. Utilizing CRISPR/Cas9 to disrupt PD-1 has

been shown to be a possible key to reducing T-cell exhaustion. I have found two articles

where this method has been applied to two different cancers: breast cancer and

glioblastoma. In triple-negative breast cancer, there is a protein called mesothelin that is

overexpressed, which is the target for CAR T-cells. When researchers used

CRISPR/Cas9 to disrupt PD-1, they found that there was a significant increase in

cytotoxicity and cytokine production, less relapse, and greater control of the tumor [7].

With glioblastoma, a similar method was used and resulted in a decrease in t-cell

exhaustion [6]. With that said, using this gene editing tool can prove useful for

overcoming inhibiting mechanisms in CAR T-cells, making treatment more effective,

especially in solid tumors. Further, highly specific CAR T-cell therapy can take time to

develop, within that time allowing the cancer to progress. These methods have shown

promise in creating a more universal treatment, eliminating the impediment previously

stated. Cytokine secretion is an important aspect of effectual treatment using CAR

T-cells. However, one big issue with the therapy is complications such as cytokine

release syndrome, caused by the increase in cytokine excretion from immune cells.

Most cases are mild, but it can be life-threatening. Researchers have been able to

modify genes that control cytokine secretion in CAR T-cells to take out harmful/toxic

secretion while amplifying the ones that increase tumor-fighting abilities [8]. That is, it

has been found that certain cytokines are more beneficial in attacking cancer cells

(IL-18, IL-15 & IL-23) and increasing persistence [6]. These are possible avenues for

CRISPR/Cas9 technology to target to improve the immunotherapy.


Overall, CAR T-Cell Therapy is a novel approach to targeting cancer cells

through the immune system and continues to develop rapidly. Using other breakthrough

biotechnology tools such as CRISPR/Cas9 has been shown to make this treatment

even more effective, and new research resumes to address limitations.








220 views0 comments

Comments


bottom of page