IMI 10: Cancer Immunotherapy Flashcards

1
Q

Observe the learning outcomes of this session

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

What is immunotherapy?

A
  • Immunotherapy is a unique approach aimed at defeating cancer.
  • It is designed to instruct the body’s own immune system to kill off the patient’s own cancer cells in the same way it does with other foreign invaders, such as bacteria.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Describe the different antigens of cancer cells

A
  1. Self-antigens:
    - all nucleated cells in our body present self antigens or self-peptide on MHC Class I
    - the immune system recognises these peptides as self and does not mount a response (tolerance)
  2. Tumour specific antigen (TSA)
    - mutated genes can be transcribed and translated and the resulting altered protein is a tumour-specific antigen (TSA)
    - they are present only in cancer cells but not normal
    - the immune system will mount a response against TSA because these antigens are new to the immune system
  3. Tumour-associated antigens (TAA):
    - cancer cells can increase the expression of some proteins that make them thrive
    - e.g. some growth factors
    - the immune system is tolerance to these proteins because they are also present in normal cells
    - however, some TAAs are expressed at a very low level in normal cells and when expressed in higher levels within cancer cells, they cna trigger an immune response
    - they can also be used to develop some immune therapies
  4. Embryonic antigens (TAA):
    - some proteins are expressed during early embryonic development and are switched off in adult life
    - some of these proteins are expressed before the immune system fully develops and acquires self-tolerance
    - cancer cells have the ability to activate the transcription and translation of embryonic genes that make them thrive
    - once these are active, the immune system encounters them for the first time and is able to mount a response
    - embryonic antigens are considered tumour-associated antigens because at a stage of development they were expressed in normal cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Describe immune surveillance, equilibrium and escape

A
  1. Immune surveillance:
    - the immune response cells of the innate and adaptive immune system are able to identify cancerous cells and eliminate them
    - these include dendritic cells, inflammatory macrophages, T helper cells, CTLs and NK cells
    - the immune response to cancer cells is very similar to that of virus infected cells
  2. Equilibrium:
    - if cancer cell are not completely eradicated during the tumour elimination phase, another transitory phase kicks in, called the equilibrium phase
    - when the immune system and the developing tumour are in a balanced state
    - tumour cells can remain dormant or keep on acquiring genome modifications
    - this can result in the alteration of tumour-specific antigens which may no longer be recognised by the immune system
    - there is also equilibrium between tumour-promoting cytokines (e.g. IL-10, IL_23) and anti-tumour cytokines (e.g. IL_12, IFN-gamma)
  3. Escape
    - see future flashcard
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Describe the escape stage of the immune response in cancer

A
  1. Escape: cancer cells subverting the immune response
    - Mutations:
    - cancer cells mutate continuously and a new set of cells of the adaptive immunity are able to recognise the new mutated antigens need to be activated
    - this prolonged stimulation can lead to immune cell exhaustion
    - Low MHC molecule expression:
    - viruses can repress the expression of MHC molecules
    - in cancer caused by oncoviruses, loss of MHC is not uncommon
    - cancer with a high rate of mutation may have aberrant MHC proteins
    - lack of MHC will halt the recruitment and activations of T cells
    - in this instance, NK cells should be more active, but often activating factors on target cells have also reduced expression
    - Lack of co-stimulatory signals:
    - tumour cells lack co-stimulatory signals
    - complete activation of cytotoxic T cells requires support from helper T cells and professional antigen-presenting cells in antigen cross-presentation
    - if such support is missing, T cells cannot be activated
    - Immunosuppressive environment:
    - cancer cells induce the production of anti-inflammatory cytokines such as IL_10 and TGF-beta
    - these dampen the immune repsonse inducing helper T cells to acquire a regulatory phenotype expressing CTLA-4
    - CTLA-4 compete with CD28 for CD80/86
    - this further leads to T cell anergy rather than activation
    - similarly, inflammatory macrophages can acquire an alternatively activated (or M2) phenotype and will become unable of antibody-dependent cellular cytotoxicity (ADCC)
    - moreover, cancer cells can exploit immune checkpoints by expressing PD-1L and directly inducing T cells anergy
    - resistance to apoptosis:
    - one of the main mechanisms of tumour cell eradication consists of apoptosis caused by cytotoxic T lymphocytes and NK cells
    - as part of the tumour/immune system co-evolution, tumour cells activate signalling pathways that lead to resistance to apoptotic signals
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What is tumour immune editing?

A
  • During immune surveillance, the immune system is capable of eliminating cancer cells.
  • However, some cells are able to enter a dormant state which enables them, with time, to evolve strategies to escape and/or weaken the immune response.
  • This phase is called tumour immune editing.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Observe this diagram of the location and steps required for the activation of T cells in response to tumour cells

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

How can immunotherapy improve such response(s) so that the immune system can fight cancer progression more effectively?

A
  • Specific
  • Immunotherapy should recognise specific tumour antigens expressed by cancer cells.
  • The first key step is identifying a tumour antigen that is found primarily on cancer cells and typically not on normal cells.
  • Adaptable
  • Immunotherapy should reinforce the immune system to adapt its attack strategy over time.
  • Tumour cells mutate over time, which may make them resistant to traditional anticancer therapies.
  • When tumour cells are killed, immune cells are exposed to tumour antigens (including the ones that have mutated), which expands and adapts the immune response cascade.
  • Durable
  • Immunotherapy should lead to a prolonged antitumor response because it should stimulate immunologic memory.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What are the types of cancer immunotherapies?

A
  • active:
  • aims to trigger an anti-tumour response from the immune system of the patient (e.g. vaccination)
  • passive:
  • requires the use of biological reagents such as mAbs or antigen-specific adaptive immune cells
  • or a combination
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Give examples of active immunotherapy

A
  • Cytokine therapy: stimulation of the patient’s immune system with cytokines.
  • Cancer vaccines: stimulation of the patient’s immune system with vaccines.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Give examples of passive immunotherapy

A
  • Monoclonal Antibody therapy: therapeutic antibodies are provided to the patient.
  • Cell-based therapy: immune cells or genetically modified immune cells are provided to the patient.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Briefly recap interferons (IFNs)

A
  • Interferons (IFNs) are known for their antiviral activity but they also play other key roles in regulating immune activity. They can be divided into three groups:

Type I (these include 13 IFN-α subtypes and IFN-β)

Type II (IFN-γ)

Type III (IFN-λ subtypes)

  • To date, only interferon alpha has been approved for the treatment of cancer. It can promote B cell proliferation, as well as activate T cells and natural killer (NK) cells.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Recap interleukins

A
  • As their name suggests, interleukins work as intercellular signals between leukocytes, our white blood cells.
  • The first immunotherapeutic agent to treat cancer in humans was interleukin-2 (IL-2). It stimulates T cell proliferation, and is largely produced by CD4+ T cells.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Recap chemokines

A
  • Chemokines induce movement of surrounding cells through a process called chemotaxis.
  • They actually have a double-edged role in tumour formation:
  • they can either decrease tumour growth by recruiting leukocytes to the tumour site, or
  • they can stimulate tumour growth by influencing movement of cancer cells.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Observe this figure and describe it

A
  • Note that the tumour microenvironment contains many different cells and cytokines, some with opposing functions.
  • By altering the cytokine milieu at the tumour site using cytokines such as IL-2 or IL-12 we can potentially convert an immunosuppressive microenvironment to one that promotes and enhances an immune response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Describe the drawbacks and limitations of cytokine therapy

A
  • Side effects: cytokine therapy may cause side effects such as flu-like symptoms, depression, and fatigue.
  • Some of them can even be unpredictable since cytokine biology is interpreted using mouse models, which are not always similar to the humans despite the many extrapolations done from research over the years.
  • Short half-life: even if cytokines are tremendously potent, they do have short half-lives.
  • Therefore, to maintain the required blood concentration for biological activity, cancer patients must receive a large amount of the cytokine preparation.
  • Amount of cytokines: expressing sufficient amounts of cytokines in the appropriate target cells is still an issue and cytokine gene therapy has been explored as a possible approach.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What is the current status of cytokine therapy?

A

To date, to the best of our knowledge, only three cytokines have been approved by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for use in cancer patients (sources www.fda.gov and www.ema.europa.eu/ema/).

  • IL-2 targets cells of the adaptive immune system, such as T cells and B cells, to respond to tumours. IL-2 has been approved for the treatment of some forms of metastatic melanoma and renal cell carcinoma (RCC).
  • IFN-α activates innate immune cells, such as dendritic cells (DCs) and macrophages to help the fight against cancer.
  • IFN-α2a has been licensed for the treatment of hairy cell lymphoma and Philadelphia chromosome-positive [Ph+, t(9;22)] chronic myelogenous leukaemia (CML);
  • IFN-α2b has been approved for the treatment of hairy cell leukaemia, AIDS-related Kaposi’s sarcoma, follicular lymphoma, melanoma, multiple myeloma, genital warts (Condyloma acuminata) and cervical intraepithelial neoplasms.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What are cancer vaccines

A
  • Cancer vaccines are a type of vaccines that are designed to boost your immune system to protect you against tumours such as cervical, prostate and bladder cancers.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What response do you think would be the most important one to elicit with an anti-cancer vaccine?

A
  • cytotoxic T cell response

There will be more potential antigens available for presentation to T cells (and vaccination uses antigens to educate the immune system).

  • The most effective response to killing cancer cells is CD8+ T cells and NK cells, so educating these is the most crucial, although B cell and T helper cell responses are also useful.
  • NK cells do not recognise antigen, so cannot be induced by a vaccination strategy, although antibodies against surface antigens can provoke NK cells to kill.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Look at this figure and describe how cancer vaccines work

A
  • all rely on generating an antigen that is presented on MHC class I molecules by dendritic cells (DCs).
  • This is shown using an mRNA molecule (but DNA can be also used) or tumour antigens (proteins) extracted from the cancer cells.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Would you use a vaccine before or after a patient develops the tumour?

A
  • it depends on what you are trying to achieve:
  • If you want to protect a patient before s/he develops a tumour then you will need a prophylactic vaccine.
  • If you want to help the immune system of the patient after s/he has been diagnosed with cancer, then you will need to administer a therapeutic vaccine.
22
Q

Describe prophylactic vaccines

A

-

  • Prophylactic, or preventative, vaccines provide prior immunity so that the body can build up tumour-specific antibodies, or T cells, without a tumour developing.
  • They are administrated to people with elevated risk of developing a particular type of cancer and who may – or may not – have been diagnosed with premalignant changes in their tissues.
  • If the tumour starts to grow, specific memory cells will be reactivated by the tumour antigen reaching lymph nodes.
  • The most successful prophylactic anti-cancer vaccines are those against viruses that can cause cancer (oncogenic viruses).
  • Vaccines against cancer viruses work by preventing infection, a step long before these viruses might induce a tumour.
  • An example of this would be preventing cervical cancer caused by certain strains of human papilloma virus (HPV).
  • These days in the UK all girls (and now boys) aged 12-18 can receive the HPV vaccine on the NHS, free of charge.
23
Q

Describe therapeutic cancer vaccines

types

A
  • These type of vaccines are administered after a tumour diagnosis and the tumour is already likely interacting with the immune system.
  • they are more commonly used in patients in remission to prevent a potential relapse and the cancer from returning.
  • Therapeutic vaccines can be classified in two groups, namely autologous and allogenic vaccines.
  • Autologous cancer vaccines: they are vaccines made from an individual’s own cancer cells and, therefore, can defined effectively as personalised vaccines. You can revise this concept in the figure:
  • Allogeneic cancer vaccines: they are made from cancer cells taken from a different individual and then grown in the laboratory before being introduced into the cancer patient undergoing the treatment.
24
Q

What are the drawbacks of cancer vaccines

A
  • Hostile tumour microenvironment: cancer cells can suppress the immune system.
  • The use of adjuvants in vaccines has been implemented to try to fix this problem.
  • Tumour escape: tumour cells can go through antigenic variations or lose the expression of antigens and/or antigen-presenting molecules, thereby bypassing the recognition by the immune system.
  • Older or sick people can have a weaker immune system: their bodies may not be able to produce an effective enough immune response after vaccination, which can limit how well the vaccine will work.

Because of these reasons, cancer vaccines may work better for smaller tumours or early-stage cancers.

25
Q

What is the current situation of cancer prophylactic vaccines?

A
  • HPV vaccine: this vaccine prevents infection by most of the oncogenic strains of human papillomavirus (HPV). Chronic infection with HPV can cause the development of some types of cancer.
  • As mentioned before, in the UK, the vaccine is recommended and free of charge for any 12-18 years old girls and boys. HPV vaccines approved by the FDA are for the prevention of:
  • cervical, vaginal, and vulvar cancer
  • anal cancer
  • oral cancer
  • genital warts
  • Hepatitis B vaccine: this vaccine prevents hepatitis B virus (HBV) infection.
  • Chronic infection with HBV can cause cirrhosis and the development of liver cancer.
  • Since the 1980s, it is part of babies’ vaccination schedules in the UK, who receive it at 8, 12 and 16 weeks old. It is also given to individuals deemed at risk (drug users, multiple sexual partners, infants of infected mothers, individuals with kidney problems, etc).
26
Q

What is the current situation of cancer therapeutic vaccines?

A
  • No cancer therapeutic vaccines have been approved by the FDA thus far but there are several Phase 2 and Phase 3 clinical trials in process or completed, and thus their use may yet be a reality soon.
  • The FDA has also approved one autologous vaccine made from immune cells:
  • Sipuleucel-t (Provenge®) is an autologous immune cell prostate cancer vaccine and is thus different from previously described cancer vaccines.
  • This vaccine stimulates the immune system to help it fight prostate cancer cells and clinical trials have suggested it can extend life for men with treatment-resistant metastatic prostate cancer.
27
Q

Describe monoclonal antibodies (mAbs)

A
  • Monoclonal antibodies (mAbs) are designed against antigens presented on the surface of tumour cells.
  • As you can see from this image, distinct types of monoclonal antibodies (mAbs for short) are used in cancer treatment:
  • Naked mAbs, which can trigger, for instance, antibody-dependent cell-mediated cytotoxicity (ADCC), or simply block cancer growth factor receptors
  • Conjugated mAbs with, for example, a toxin, a chemotherapeutic agent or a radioisotope
  • Bispecific mAbs bridge (connect) the target cells with cells of the immune system bringing them closer together:
28
Q

What are naked monoclonal antibodies?

A
  • Naked mAbs are named this way because they are not attached to any radioactive material or drug.
  • They work alone or by activating functions through their Fc binding to Fc receptors.
  • They are the most common type of mAbs used as a therapy to treat and manage cancer.
29
Q

What are the different ways naked mAbs can work?

A
  • They can improve the immune response against cancer cells by attaching to the tumour cells and marking them for the immune system to kill them through antibody-dependent cell cytotoxicity (ADCC), which we also encountered as type II hypersensitivity in IMI9.
  • ADCC leads to cell lysis via effector immune cells such as NK cells, eosinophils, macrophages or neutrophils.
  • They can bind to and inhibit proteins on cancer cells, or on other nearby cells, that are necessary for the cancer cells to grow or spread (e.g. growth factor receptors).
  • They can increase the immune response by targeting immune checkpoints such as anti-PD-1/ anti-PD-L1.
  • Immune checkpoint inhibitors work by targeting and disabling the fail-safe mechanisms that have evolved to prevent unwanted immune responses.
  • These checkpoints ensure that responses only occur when needed and that they are switched off as soon as the issue has been resolved (be it an infection, or injury).
  • As shown in the right-hand figure below, they can bind and block either the receptor (PD-1) on the immune cell or the ligand (PD-L1) on the tumour cell.
30
Q

What is checkpoint regulation?

A
  • Checkpoint regulation refers to the increase of ligands (e.g. PD-L1) for inhibitory receptors on the tumour cell surface.
  • This is now recognised as a really important aspect of cancer biology:
  • by upregulating PD-L1, cancer cells can reduce the ability of CD8+ T and NK cells to kill them as, to overcome the negative signal, the cytotoxic cells will need an even stronger stimulation to induce degranulation.
  • This is a key part of the balancing of activating and inhibitory receptors.
31
Q

What are conjugated mAbs?

A
  • these are antibodies that are attached (‘conjugated’) to a radioactive particle or to a chemotherapy drug
  • they are thus used as a shuttle to deliver these substances more specifically to the cancer cells
  • Radio-labelled antibodies: these are attached to small radioactive particles.
  • For instance, to treat some types of non-Hodgkin lymphoma, an anti-CD20 antibody is used to deliver radioactivity directly to cancerous B cells.
  • CD20 expression is restricted to B cells and it is thought that it can oligomerise on the membrane and form channels for Ca2+.
  • This is why this molecule is believed to play a role in B cell activation, thus making it a good vehicle to be used to deliver radioactivity to these type of cancer cells.
  • Chemo-labelled antibodies: these are attached to chemotherapeutic drugs.
  • The drugs used in this way are often the ones that are too strong to be delivered alone and could have dangerous side effects if not attached to an antibody.
  • For instance, Hodgkin lymphoma can be treated by delivering monomethyl auristatin E (MMAE) to cancerous cells, using an anti-CD30 antibody as a shuttle.
  • CD-30 expression is restricted to activated T, B and NK cells, as well as monocytes, and it is involved in the proliferation of B and T cells, which is why it can be used as a vehicle in this case.
32
Q

What are bispecific monoclonal antibodies

A
  • Bispecific mABs are named in this way because they are designed as an antibody that is composed of two different mAbs, and can thus bind two different proteins (antigens) simultaneously.
  • This allows them to bring together, or cluster, the cancer cells and immune cells, which makes it easier for the immune cells to attack the cancer cells.
  • The image below shows an antibody bringing the tumour cell and T cell together, with a third innate cell such as a macrophage, dendritic cell (DC) or innate lymphoid cell/ NK cell, binding the Fc region of the antibody.
33
Q

What are the drawbacks of monoclonal antibody therapy

A

mAb therapies are not without their risks and they do have a number of drawbacks. For example:

  • Allergic reactions such as hives or itching.
  • Since antibodies themselves are foreign proteins, they can sometimes stimulate allergic reactions once injected into the patient.
  • Production costs. They are very expensive and a huge effort is required to produce them.
  • Tissue penetration.
  • Typically, no more than 20% of the mAbs directed against tumour-specific antigens will interact with the tumour, in murine xenograft models (i.e mouse models transplanted with human cancer cells).
  • Most of the mAb molecules appear to remain in the blood instead.
  • Mode of action. mAbs show diverse modes of actions in vitro, but what happens once injected in patients (i.e. which antibody function or mechanism is most important) is not always clear.
  • A high rate of mutation causes antibodies to be rapidly ineffective.
34
Q

What is the current status of monoclonal antibody therapy?

A
  • Even with these drawbacks, many mAbs have been approved for cancer treatment in the last years and many patients have benefited from their use
35
Q

What are human monoclonal antibodies?

A
  • Human monoclonal antibodies can now be generated without the need to immunise humans, thanks to the development of mainly two techniques:
  • transgenic animals (typically mice or rats)
  • antibody phage display.
  • Transgenic animals are animals that are genetically modified in a way that their own immunoglobulin genes are replaced with the human ones.
  • By doing this, upon immunisation they produce human antibodies based on the new inserted human gene sequences.
  • Even more impressively, the other technology, the antibody phage display, permits to generate human antibodies without immunisation at all!
  • These are derived directly from libraries of human antibody molecules with randomised complementarity determining region (CDR) loops (IMI3), and the entire process is performed in vitro.
36
Q

What are humanised and chimeric antibodies?

A
  • Humanised and chimeric antibodies are derived from non-human immunoglobulins (typically generated in mouse or rat) instead.
  • Those protein sequences are then modified using genetic engineering to make them similar to the homologous human ones.
  • Specifically, chimeric antibodies are produced when the mouse constant domains (Fc) are replaced with that from human;
  • humanised antibodies are instead more similar to the human variant (with as many amino acids as possible converted to the human sequence) despite the non-human origin of its complementarity determining regions (CDRs).
37
Q

What is cell-based therapy?

Give some examples

A
  • Cell-based or cellular therapies rely on immune cells derived straight from the patient which are modified in the laboratory to enhance the recognition and killing of the patient’s own tumour cells.
  • Examples of cell-based therapies include:
  • Tumour infiltrating lymphocytes (TILs) with IL-2;
  • Genetically modified cytotoxic T cells.
38
Q

Describe adoptive T cells transfer (ATC) therapy and how tumour infiltrating lymphocytes (TILs) are utilised

A
  • The name of tumour infiltrating lymphocytes (TILs) derives from the fact that they are found in the tumour, where they migrate to in order to kill it.
  • In adoptive T cell transfer (ATC) therapy, TILs are isolated from tumours and expanded ex vivo (i.e. outside the patient) with a high dose of IL-2, well-known for driving the proliferation of T cells
  • note that despite this function, IL-2 is not classified as a growth factor but rather as a cytokine).
  • Not all TILs will be the same or show the same killing efficiency and thus different cells of that population of lymphocytes are grown.
  • TILs that show the best tumour reactivity are then selected and further expanded to be infused back into the patient, as depicted in the figure.
39
Q

Can you think why prior chemotherapy might be important for the patients before a TIL therapy is used?

A
  • there are also the endogenous lymphocytes which, if not removed (killed!), could compete for growth factors or induce immunosuppression, and decrease the anti-tumour effects of these TILs.
40
Q

What are the two classes of genetically enhanced T-cell therapies?

A
  • chimeric antigen receptor (CAR) therapy
  • genetically modified T cell receptor (TCR) therapy
41
Q

Describe CAR-T cell therapy

A
  • Chimeric antigen receptor therapies utilise receptors that have been engineered to give T cells the new ability to target a specific tumour protein independently of the MHC presentation.
  • T cells are harvested from patients, genetically modified, and then the resulting CAR-T cells are re-infused into patients to fight their tumours.
  • CAR-T cells can be either derived from the same patient’s T cells (autologous) or from another healthy donor (allogenic), and the receptors are called chimeric because they are a mixture of various proteins, as you can see in this figure.
42
Q

Describe modified TCR therapies

A
  • Genetically modified TCR therapies are based on modifying the expression of specific TCR α and β chains, which mediate the recognition of the cancer antigen.
  • This means that the tumour-specific TCR α and β chains must be first identified, then genetically engineered to improve specificity and affinity for a tumour antigen and, finally, transferred into T cells to become tumour antigen-specific T cells.
43
Q

Observe this diagram for an overview of CAR-T and modified TCR cell therapy

A
44
Q

What is the main difference between TCR and CAR-T cells?

A
  • CAR therapies (with an antibody-derived binding motif) directly recognise the tumour antigen on the cell surface (external antigens), just like antibodies do.
  • TCR therapies (with a specific TCR-based antigen-binding specificity) recognise cancer peptides presented on HLA molecules (internal antigens).
  • By now you should recall that all proteins synthesised in a cell (both intracellular and extracellular proteins) are degraded and loaded onto an HLA molecule to be presented on MHC class I.
  • Therefore, TCRs can potentially recognise any molecule from the whole proteome, whereas CARs can only be used to attack proteins on the cell membrane.
  • This naturally gives TCRs a bigger advantage over CARs in terms of possible targets.
  • On the other hand, TCRs are difficult to manipulate since they are composed of α and a β chains, and on top of that require a battery of signalling proteins, which are needed to recruit the components of an immune synapse.
45
Q

What is the immune synapse?

A
  • The immune synapse is the interface between a lymphocyte (T/B cells or NK cells) and an antigen-presenting cell (APC).
  • More specifically, it is the dynamic interaction between the membrane molecules responsible for the activation of the lymphocytes and the membrane proteins presenting the non-self antigens.
46
Q

What are the drawbacks of cell-based therapy?

A
  • Side effects. The two most important side effects occurring in patients treated with CAR T-cell therapy are cytokine release syndrome (CRS) and neurotoxicity.
  • CRS is a life-threatening inflammatory response caused by a massive release of cytokines in the bloodstream by over-activated immune cells.
  • Manufacturing process. It takes a few weeks to prepare CAR T cells.
  • The process starts with the isolation of autologous T cells by a process called apheresis, their genetic modification and expansion, and several other steps before being finally reintroduced into the patient.
  • This makes it very expensive and time-consuming.
  • In vivo efficacy.
  • There are no consistent benefits when comparing adoptive T-cell transfer strategies with in vitro experiments.
  • Unfortunately, in vitro studies with human cells, as well as in vivo studies in mice, do not provide us with accurate information on the side effects of the therapy.
  • It is only by doing clinical trials that we can actually determine the safety of the therapy for patients.
  • Antigen loss.
  • Due to adaptive pressure, or, if you like, the evolutionary adaptation of tumour cells, adoptive T cell transfer may produce an evolutionary pressure for cells to lose the antigen, resulting in tumour escape (i.e. evasion from the therapy).
  • Hostile tumour microenvironment.
  • Cancer cells can suppress the immune system, sometimes recruiting repressive immune cells to support tumour survival.
  • These immunosuppressive processes can prevent the infused T cells from acting, just as they prevent the normal immune response from attacking cancer.
  • To get around this, additional immuno-modulatory treatments are often used alongside cell therapies to help them work better.
  • For instance, blocking PD-L1 on the tumour cell (which we mentioned earlier in the eModule) can prevent PD1-mediated inhibition of the infused T cells and thus facilitate tumour killing.
47
Q

What is the current status of cell-based therapy?

A

Five CAR-T cell therapies have thus far received FDA approval:

  • Tisagenlecleucel (KymriahTM) for the treatment of diffuse large B cell lymphoma and B cell acute lymphoblastic leukaemia (ALL);
  • Axicabtagene ciloleucel (YescartaTM), for the treatment of relapsed/refractory large B cell lymphoma in adult patients.
  • Brexucabtagene autoleucel (TecartusTM), for treatment of mantle cell lymphoma (MCL) or acute lymphoblastic leukemia (AML)
  • Lisocabtagene maraleucel (Breyanzi®), for treatment of large B-cell lymphoma in patients that have had at least 2 previous failed treatments.
  • Idecabtagene vicleucel (Abecma®), for treatment of multiple myeloma (MM) in patients who have received at least four kinds of treatment that have failed.
48
Q

Which cells are therapeutic cancer vaccine therapies normally relying on for generating an immune response?

A
  • dendritic cells
  • The vaccine may be made of material from tumour cells, but it is dendritic cells that we rely on to make or process the antigen for presentation on class I MHC molecules.
49
Q

Which of the following cytokines is used to expand TILs in vitro?

A
  • IL-2
50
Q

Which of the following cell-based therapy is directed against cancer antigens that are NOT expressed on the cell surface?

  • CAR-T
  • TCR
A
  • TCR
  • CAR-T cells have antigen specificity based on an antibody domain, so binds to the cell surface.
  • Recombinant TCRs will bind specifically to antigens derived from those synthesised in the cell.
  • These can be from any cell proteins (intracellular, extracellular or secreted proteins) as the antigens are made while the protein is synthesised.
51
Q

Which monoclonal antibody therapy induces antibody-dependent cell-mediated cytotoxicity?

  • naked mAb
  • conjugated mAb
A
  • naked mAb
  • Cell-mediated responses to antibodies (like ADCC) rely on the cell binding to the Fc region of the antibody in naked antibodies.
  • Conjugated antibodies rely on directly binding and causing cytotoxicity to the cancer cell.
52
Q

Observe this diagram of how tumours evade the immune system

A