Scarica il documento per vederlo tutto.
Scarica il documento per vederlo tutto.
Scarica il documento per vederlo tutto.
Scarica il documento per vederlo tutto.
Scarica il documento per vederlo tutto.
Scarica il documento per vederlo tutto.
Scarica il documento per vederlo tutto.
Scarica il documento per vederlo tutto.
Scarica il documento per vederlo tutto.
vuoi
o PayPal
tutte le volte che vuoi
CANCER IMMUNOTHERAPY
It includes compounds capable of reactivating immune system’s response, in order to
make it able again to recognize tumor cells
DOMANDA D’ESAME: Which are the features of immunologic (immunogenic) cell
death?
Immunogenic Cell Death (ICD) triggers an immune response by releasing danger
signals and tumor antigens that activate the immune system. Unlike other forms of cell
death, such as apoptosis or necrosis, which can be immunologically silent or even
suppressive, ICD is highly pro-inflammatory and enhances anti-tumor immunity.
This is done through the release of DAMP molecules (Damage-Associated Molecular
Patterns).
Key DAMPs include: →
- Calreticulin (CRT) A protein exposed on the cell surface that acts as an “eat-
me” signal for dendritic cells and macrophages. →
- ATP and HMGB1 (High-Mobility Group Box 1) They trigger the activation
of Toll-like receptors (TLR4) on dendritic cells, enhancing antigen presentation.
→
- Heat shock proteins (HSPs) They are translocated in stress conditions on the
plasma membrane, where they exhibit immunostimulatory effect, based on their
interaction with APC’ surface receptors CD91 and CD40; they also facilitate
cross presentation of tumour cells’ antigens on MHC class I molecule, leading
to the CD8+ T cell response.
DNA-damaging agents can
enhance the efficacy of immune
checkpoint inhibitors like anti-
PD1/PDL1 or anti-CTLA4,
enabling a sustained anti-tumor
immune response.
Immunogenic cell death bridges
both immunotherapy and
mutation-inducing therapies, as
therapies that induce DNA
damage can enhance immune
responses against cancer.
• Targeting cancer cells Cytokines
By combining a compound able to induce immunogenic cell death with cytokines,
which activate the immune system against the tumor, we can influence the early stages
of tumor-immune cell interactions (elimination).
In the lab, we have access to a wide range of cytokines, which they can be infused into
patients to stimulate the immune system, promoting T cell priming.
Furthermore, we can adapt specific cytokines or metabolites to target and enhance
specific stages of immune response. For instance, we can influence timing, recruitment
of immune cells like NK cells, or even checkpoint inhibitors (CPI). However, this
approach isn’t always the ideal, as tumors can take advantage of immune respone, for
example using VEGF to start migrating. Thus, while reactivating the immune response
with cytokines can be beneficial, it also enhances the probability to facilitate tumor
growth and proliferation. Cancer vaccines
Vaccines are potentially the most powerful tool we have, as they are proven to work.
Usually, vaccines are used for prevention rather than treatment, but research is
progressing in this area, and the development of peptidomics is a growing field in
cancer research.
Peptidomics involves identifying tumor antigens presented on MHC or HLA
molecules. This helps determine if specific peptides are associated with each cancer
type. By identifying and administering these antigens using personalized strategies, we
can enhance the immune response.
Adoptive cell transfer
It is one of the most classical applications, which includes isolating and expanding
immune cells ex vivo: we can take blood samples from patients, then isolate the
immune cells of interest (such as T cells), stimulate them with specific antigens (often
in combination with APCs), and re-infuse them into the deriving-patients to maximize
the anti-cancer response.
This can be done in different ways:
1) TIL therapy.
Isolate directly the tumor-infiltrating lymphocytes (TILs), which can be collected via
biopsy, then stimulated with specific antigens such as interleukin-2 (IL-2), which helps
support their growth and activation, and re-infused into the patient.
These kinds of ex-vivo approaches have shown significant results in certain cancers
like melanoma. So, even though they have the limitation of requiring time (typically 2
to 3 weeks), they allow the expansion of a robust immune cell population.
During this process, additional analyses, like exome sequencing, can identify tumor-
specific antigens.
Synthetic antigens can then be used to prime APCs, which, in combination with TILs,
create a potent immune response.
However, these protocols are complex, time-consuming, and costly. Indeed,
manipulating cells ex vivo requires specialized facilities, permissions, and skilled
personnel, often limiting such activities to specific labs or companies. Anyway, they
hold significant promise for advancing cancer immunotherapy.
TCR/CART therapy
By genetically engineering T cells to express chimeric antigen receptors (CARs), we
can target tumor antigens more effectively.
We also have the option to induce a cytokine response directly within the tumor, which
can help stimulate other immune cells and enhance the overall immunotherapeutic
effect.
There are different generations of CAR-T cells based on the activation pathways and
signalling mechanisms. The first generation provides limited activation, while the
second generation incorporates additional signalling domains that enhance cell
activation.
The protocol is the same:
‐ T cells are collected.
‐ They are manipulated, typically using viral vectors to express the CAR receptor
targeting the tumor.
‐ Then expanded in vitro and infused back into the patient.
CAR-NK cells are emerging as a promising alternative to CART, combining immune
checkpoint inhibitors with other immune approaches for enhanced results.
Tumor-specific mAbs
Antibodies play a critical role in cancer therapy, as they serve as an effective tool to
deliver targeted therapies against specific cells.
Immune checkpoint inhibitors are part of this group.
One of these inhibitory molecules is CTLA-4 (cytotoxic T-lymphocyte-associated
protein 4), which binds to the CD28’s ligands, CD80 and CD86 (B7).
When a T cell needs to be deactivated, it upregulates CTLA-4 expression; so, the shared
ligand B7, preferentially binds to CTLA-4 instead of CD28, resulting in a negative
signal that inhibits T cell activation. This mechanism ensures that the immune response
is appropriately regulated and prevents excessive immune activation.
Immune checkpoint inhibitors are designed to block the immunosuppressive phenotype
induced by tumor cells. Specifically, they use monoclonal antibodies to block negative
immune receptors, thereby preventing the deactivation of T cells. This strategy is
especially relevant in the tumor microenvironment, where not only tumor cells but also
other immune cells, such as macrophages, contribute to T cell suppression.
Thus, immune checkpoint inhibitors allow us to manipulate both positive and negative
co-stimulatory molecules.
While T cell activation is critical for eliminating pathogens like viruses or bacteria, it
is equally important to switch off this response at the appropriate time. Without proper
regulation, immune cells could potentially destroy all our cells, leading to a spread
damage.
This is particularly evident in systemic autoimmune diseases, where immune cells
slowly destroy various tissues throughout the body. However, in acute autoimmune
conditions, such as type 1 diabetes, the destruction of specific cells can occur rapidly,
sometimes within months. This destruction can result in conditions like hyperglycemia.
Immune checkpoint inhibitors have shown great results, especially in preclinical
models: early use of anti-PD-1 or anti-CTLA-4 antibodies resulted in the first examples
of tumor-free mice. Even after one year of treatment, these mice remained tumor-free,
demonstrating the durability of the response. This approach, which proved effective in
preclinical models, was successful also for human treatments, leading to positive
results in many patients.
So, it’s crucial to stratify patients based on tumor mutational burden (TMB): for
instance, melanoma and mismatch repair (MMR)-deficient colorectal cancer have high
mutational burdens and are responsive to these therapies, because they present many
neoantigens, which play a critical role in achieving a good therapeutic outcome.
On the contrary, tumors classified as cold, meaning that they lack sufficient
neoantigens, typically show poor responses to these treatments.
One important side effect of immune checkpoint inhibitors is immune system
hyperstimulation, which can lead to autoimmune diseases and inflammation. For
example, recent reports have linked these therapies to conditions such as diabetes
(classified as type 3 diabetes), arthritis, and even rare cases of leukemia.
Bispecific Abs
They bridge tumor cells and immune system, such as T cells or NK cells, to stimulate
cytotoxic effects against the tumor: by manipulating the antibody’s regions, we can
design molecules that enhance the interaction between immune cells and tumor
antigens, thereby promoting the formation of a cytolytic synapse. This facilitates the
activation of T cells or NK cells to destroy tumor cells.
These molecules are engineered antibodies that can bind two different targets
simultaneously.
Compared to more complex therapies like CAR-T cells, bispecific antibodies offer a
simpler and more accessible approach. They can be further modified by conjugating
them with other molecules to enhance their specificity and effectiveness. For example,
these antibodies can link T cells to tumor antigens or even target other immune cells to
amplify the immune response. Oncolytic viruses
These viruses can be genetically engineered to specifically target tumor cells: by
modifying their structure, we can direct these viruses to attack tumor-specific receptors
on the cell membrane.
Additionally, oncolytic viruses can be designed to deliver compounds that stimulate
immune cells or induce ICD in tumor cells. This process releases tumor antigens,
triggering an immune response and restoring immune surveillance.
Oncolytic viruses can also be combined with DAMPs and pathogen-associated
molecular patterns (PAMPs) in subsequent phases of treatment. This combination
amplifies the immune response, creating a multi-faceted attack to the tumor. While this
approach is highly specific and shows great potential, it involves complex protocols
and is still under investigation.
MODELS IN CANCER RESEARCH
Different models are used in cancer
research:
a. In vitro models → cell lines (usually)
b. In vivo models → usually rodents, but
also C.elegans, Drosophila or
Zebrafish
c. Ex vivo model → when
experimentation is done in or on
tissues obtained from an organism and maintained under optimum conditions,
mimicking the natural condition.
When choosing a model system, we should consider:
→
- Cost in vivo models are more expensive than in vitro model
→
- Complexity for example, to study the microenvironment (important in tumor
progression) we cannot use a sin