Immunotherapy 14/03/23 Flashcards

1
Q

What is immunotherapy?

A

Immunotherapy is defined as harnessing the patient’s own immune system to fight disease.

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

What are immuno-oncology therapies?

A

Immuno-oncology therapies represent a significant advance in cancer treatment beyond traditional treatment.

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

How do immunotherapies work?

A

Immunotherapies engage and recruit the immune system to combat disease by inducing an antibody generation response against a specific antigen (foreign entity) typically present on the cell surface of a virus or cancer cell. This is very similar to a traditional vaccine approach seen for a whole host of diseases throughout history. These cell surface antigen targets typically evade the surveillance mechanisms of the immune system and thus persist and manifest disease within the host. As a result of the body’s inability to recognize and destroy these antigen targets via antibody generation and subsequent eradication, these antigens must be presented to the immune system exogenously. An option for the immune-tolerant host outside of traditional medical intervention is immunotherapy via exogenous delivery. Immunotherapies are directed at a specific antigen or cluster of antigens that compose the unique signature of a virus or cancer cell that is dissimilar to its host thus recognizing self from non-self.

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

Where has there been success with immunotherapies?

A

There are lots of different types of immunotherapy, but for all types, its success requires the tumour load being reduced first by surgery, irradiation, or chemotherapy for 2 reasons:

-Immune system cannot cope with large tumours
-If had lots of antigen shedding from the tumour it would stimulate the regulatory T-cells, and thus stop an immune response against tumour
-After debulking the tumour, smaller amounts of tumour are left as the main target for immunotherapy

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

What are humanised monoclonal antibodies?

A

Humanized monoclonal antibodies that target cell surface components of tumour cells are increasingly being used to treat cancer been approved for therapeutic use.

Antibodies reacting with antigens on the surface of tumour cells protect the host by complement mediated opsonization and lysis and also through recruitment of macrophage and NK ADCC. The FcR cells not only act as cytotoxic effectors but cause crosslinking of antibody coated cells which leads to apoptosis or exit from the cell cycle, and makes the cells sensitized to irradiation and DNA damaging chemotherapy.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What are immunoconjugates?

A

A lot of interest has been made into therapeutic immunoconjugates which consist of a tumour targeting antibody linked with a toxic effector component such as radioisotope, toxin or small drug molecule, or prodrug (antibody enzyme prodrug treatment-ADEPT-so drug gets activated at the site of the tumour-so highest concentrations delivered there).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What are some of the approved humanised monoclonal antibodies?

A

More than 20 humanized or fully human monoclonal antibodies and immunotoxins were approved by the US Food and Drug Administration by the end of 2019 for use in cancer. All of these antibodies listed target a cell surface component except the one targeting Vascular endothelial growth factor (VEGF), this neutralizes VEGF to prevent angiogenesis which is required for tumours to grow

Examples include:
-MAb approved for HER2 in breast cancer
-CD20 in lymphoma
-EGF in colorectal cancer

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

How do the humanised monoclonal antibodies work?

A

Therapeutic antibodies are specific for a protein on the surface of the tumor cells, such as the binding of rituximab to CD20. The antibodies coat a tumor cell with their Fc regions pointing away from the cell. These can then engage the FcγRIII receptors on an NK cell. Signals from the receptors activate the NK cell to kill the tumor cell. This is an example of ADCC (antibody mediated cell-mediated cytotoxicity). The monoclonal antibody bindstothetumorcellantigenwithitstwoFabarmsandtoFcγRIIIontheNKcellwith itsFcregion,thuscreatingastrongadhesionbetweenthetwocells. The activatingsignalsgeneratedbyFcγRIIIareaugmentedbysignalscomingfromNKG2 andotherNK-cellreceptorstoactivatethecytotoxicmachineryoftheNK cell.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Why are mouse monoclonal antibodies not used?

A

Monoclonal antibodies were originally produced using mouse hybridomas. Specificity of mouse monoclonal antibodies makes them very useful for treating a broad range of clinical conditions. Problem though, when mouse monoclonal antibodies are introduced into humans they are recognised as foreign, and evoke an immune response known as the HAMA response. Human anti mouse antibody, which quickly clears the mouse monoclonal antibodies from the bloodstream and lowers the therapeutic efficiency of subsequent administration.
It does not just reduce the efficiency of the treatment it can also cause a host of other complications including allergic reactions, or the accumulation of mouse and human ab complexes in organs such as the kidneys which can cause life threatening problems.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

How are humanised monoclonal antibodies made?

A

One way to avoid these undesirable mouse reactions would be to use human monoclonal antibodies, produced by human hybridoma cell lines but this is not possible due to technical problems. So alternative methods of antibody engineering using recombinant DNA technology have been developed to overcome these difficulties. It is now possible to create ‘man made‘ abs that have less non-human protein in them.

Two main ways of making these:

-Design and construct genes so that you clone the promoter, leader and variable region from a mouse ab gene, and constant region exons come from a human ab gene. An ab produced this way using recombinant DNA technology is a mouse human chimera, and is called a chimeric ab, this type of ab is partially humanised. Its antigen specificity, which is determined by the variable region, is encoded by the mouse, and its isotype, which is determined by the constant regions, is derived from the human DNA. Because the constant regions are encoded by human DNA, the abs have fewer mouse antigenic determinants, and are far less immunogenic when administered to humans than mouse monoclonal antibodies. Of the 20 antibodies that have been approved for use in humans to treat various diseases, approximately half are chimeric. One such chimeric ab that is used for treatment is rituximab which targets CD20 of the B cells in non-Hodgkin’s lymphoma. When the chimeric antibodies were introduced, a human anti-chimeric antibody response (HACA) was observed.
-However because the entire mouse VH and VL regions are retained in chimeric abs, they contain significant amounts of mouse ig sequence, which can cause HACA (human anti chimera antibody) responses. It is now possible to engineer abs in which all of the sequence is human except the CDRs. This is known as CDR grafting-involves the substitution of non-human CDR domains from an mouse ab into the most closely related human ab sequence available, so that only the CDR domains are non-human. This is a fully humanized ab. These humanized abs retain the biological effector functions of human antibody and are more effective than mouse abs in triggering complement activation and fc receptor mediated processes such as phagocytosis in humans. They are also less immunogenic in human than mouse-human chimeric abs.

However even when graft CDR regions into the human framework region, you see and anti-variable region response. It is thought that only 20-30% of the residues in the CDRs are crucial for antigen binding called the specificity determining residues (SDRs). Minimally immunogenic molecules are generated by grafting the SDRs onto a human framework. Veneering is another approach, based on the premise that only the surface residues contribute to the immunogenicity of the variable regions-so it changes only exposed residues that differ from human amino acid sequence, while those residues which are buried within molecule are not modified. In both the chimeric and humanised abs the specificity was determined from the mouse (clone the sequence from the mouse hybridoma lines which can be readily generated), whereas the isotype of the ab is governed by cloning human gene sequences corresponding to constant segments. Because of these approaches the abs contain fewer mouse antigenic determinants, so are less immunogenic in humans. Another advantage to humanising abs is that mouse abs have very short half-life of a few hours, compared with the 3 week half lives of their human or humanized counterparts.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Process of production of full human Ig in mice by antibody engineering?

A

Chimerization and humanization of abs are labour intensive procedures, involving sequence analysis, engineering approaches, analysis and optimization of binding affinity, and evaluation of immunogenicity for each ab. So technological developments have been undertaken so that fully human antibodies are produced by engineering the Ig loci, rather than components within the Ig molecule.
Technology involves removal of the Ig H and L chain loci of mice which creates knock out mice.

Embryonic stem (ES) cells were collected from these KO mice from the blastocyst. Human artificial chromosome containing the Ig H (1.5 Mb) and lamda loci (1 mb) were transfected into ES cells.
These modified ES cells were put back into a blastocyst and transplanted into surrogate mothers. The B cells of the offspring produced human abs in response to antigen.
The transgenic mice produced completely human abs in response to antigenic challenge, in cells that you can make hybridomas from (because they’re produced in mice B cells). So can produce human moabs of any specificity by this method.
This approach particular useful for producing large quantities of human abs, for example, if replace cattle loci with human ig koci, larger quantities of human Moabs produced because contain ~60 litres of blood.

-Panitumab (colorectal Ca) and Zanolimumab (lymphoma) have been produced using this approach. Panitumab has been approved for tx of metastatic colorectal cancer, and zanolimumab is currently in phase III trials for t cell lymphoma.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What has changed in melanoma treatment since monoclonal antibodies?

A

Melanomaaccountsfor75%ofthedeathscausedbyskincancer.In2018,therewere61,000deathsfrommelanoma,and288,000newcasesofthediseasewerediagnose worldwide. At that time, 75% of patients with advanced melanoma survived less than 1 year so great need to develop more effective treatments, hence a lot of research done using immunotherapies for this disease. Since 2011, more than 17,000 patients have been treated with ipilumumab (binds to CTLA4), the 5 year survival rate of melanoma has increased from 8% to 18%.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What are the immune checkpoint inhibitors for antibodies?

A

Designed to overcome blockages to T cell activity mediated by immune checkpoints.
Work by reactivating TILs (tumour infiltrating lymphocytes), particularly CD8+ cytotoxic T cells. CTLA-4 Checkpoint Inhibitors: Anti-CTLA-4 mAbs are designed to augment T cell activation by blocking inhibitor receptors such as CTLA-45.
Checkpoint inhibitors against PD-1 and its ligand PD-L1 release PD-1 pathway-mediated inhibition of T cell activation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What was Allison’s research?

A

Allison’s discoveries built on the work of French immunologists from the 1980s who were studying T cells, components of the immune system that attack cells that the body recognizes as foreign. They identified a key receptor on the surface of T cells that they called cytotoxic T-lymphocyte antigen 4, or CTLA-4. Allison and others found that the receptor puts the brakes on T cells, preventing them from launching full out immune attacks. Other groups hoped to use the receptor to help treat autoimmune disease in which the immune system’s brakes aren’t strong enough. But Allison had a different idea. Cancer develops when the body’s immune system fails to attack tumour cells, even though they are growing out of control; Allison wondered whether blocking the blocker—the CTLA-4 molecule—would set the immune system free to destroy cancer. This was a new concept, to target the body’s system of immunosuppression as a tool to help defeat tumours. In 1996, Allison published a paper in Science showing that antibodies against CTLA-4 erased tumours in mice.

Pharmaceutical companies initially shied away from cancer immunotherapy, wary of possible side effects and also of an approach so different from the standard treatments of surgery, radiation, or chemotherapy. So the job of getting anti–CTLA-4 into people fell to a small biotechnology company, Medarex, in Princeton, New Jersey. It acquired rights to the antibody in 1999 and were the first to use it as a drug.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What was Honjo’s research?

A

Honjo, meanwhile, in the early 1990s discovered a molecule expressed in dying T cells, which he called programmed death 1, or PD-1; he recognized PD-1 as another brake on T cells. Initially, “I didn’t realize there was a connection to cancer,” Honjo said at today’s press conference. Later, however, he and others performed key experiments showing that the molecule could be a target in cancer therapies. The first clinical trials using PD-1 were even more dramatic than those with CTLA-4. Several patients with metastatic cancer were apparently cured. And side effects seemed milder than those observed with CTLA-4 therapies.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What is KEYTRUDA treatment in non small cell lung cancer?

A

KEYTRUDA FDA approved in May 2017 as the first cancer treatment for any solid tumour with a specific genetic feature. First time the FDA approved a cancer treatment based on a common biomarker rather than tumour location.
FDA approval of Keytruda (pembrolizumab) for the treatment of adult and paediatric patients with unresectable or metastatic solid tumours with a biomarker referred to as microsatellite instability-high (MSI-H) or mismatch repair deficient (dMMR).

KEYTRUDA (Pembrolizumab by Merck), PD-1 (programmed cell death protein 1) inhibitor, blocks PD-1/PD-L1 pathway resulted in 3 year survival of metastatic melanoma patients 40%, whereas previously survival was measured in months. KEYTRUDA approved as first line treatment for advanced non small cell lung cancer (NSCLC) whose tumours express PD-L1 at >50%, and other cancers. PD-LI is useful biomarker for effectiveness of PD-1 inhibitors such as KEYTRUDA or nivolumab as 1st line treatment in advanced NSCLC.

17
Q

What cancers can KEYTRUDA treat?

A

-Melanoma
-Non small cell lung cancer
-Head and neck squamous cell cancer
-Urothelial cancer (bladder/urinary tract cancer)
-Colona or rectal cancer
-Oesophageal carcinomas
-Cervical cancer
-Renal call carcinoma
-Advanced endometrial carcinoma
-Cutaneous squamous cell carcinoma

18
Q

What are monoclonal antibodies as immune agonists?

A

These mainly antibody based drugs target specific cell surface proteins on T cells, causing stimulation of T cell activity. CD27 (on T cell) interacts with CD70 on antigen presenting cell to deliver “second signal”. Varlilumab can substitute CD70 by delivering costimulatory signal to CD27 positive T cells with engaged TCRs. Preclinical studies have demonstrated costimulatory activity of Varlilumab and decreased Treg in tumours. Varlilumab may also have direct benefits in CD27 positive tumours (some B and T cell lymphomas).

19
Q

What are modified monoclonal antibodies?

A

CD30 is found on the surface of anaplastic large cell lymphoma tumour cells (this is a T cell lymphoma) or Hodgkin’s lymphoma consequently CD30 is a target for therapeutic antibodies. Antibodies are used to target toxins to tumour cells. The anti-CD30 antibody brentuximab is coupled to the cytotoxic drug auristatin by a cathepsin cleavable linker. This conjugate is called brentuximab–vedotin. After the antibody has attached the conjugate to the surface of the tumor cell, the conjugate is internalized into endosomes, where cathepsin cleaves the linker and releases the auristatin. This potent anti-mitotic drug passes to the nucleus, where it binds to microtubules and prevents them from forming a mitotic spindle. This prevents mitosis and induces the tumor cell to die by apoptosis.

20
Q

What are bispecific T cell engager antibodies?

A

Bispecific T cell engager (BiTE®) antibodies are designed to bridge cancer cells to CTLs. The BiTE® antibody construct utilizes the binding properties of the variable domains of two monoclonal antibodies. One domain is designed to target an antigen on the surface of a cancer cell whereas the other is designed to engage CD3 on the surface of a T cell. With these two different domains, BiTE® antibodies aim to engage the endogenous cytotoxic potential of CTLs, bypassing MHC/antigen-dependent activation of T cells.

21
Q

What is adoptive T cell transfer?

A

Adoptive T cell transfer involves generating large numbers of T cells, outside the body (ex vivo). These isolated T cells can be genetically engineered so that they produce cytokines such as IL-2 which will boost their activity. The T cells are expanded by in vitro stimulation with antigen presenting cells and are then put back into the patient. By growing these cells in vitro, responses can be generated which are not found in vivo due to tumour derived inhibitory factors or the presence of T regulatory cells.

-10^11 cells
-IL-2 in vivo or Th cells improves CTL survival & function
-75% of circulating T cells with anti-tumour activity
-Response rates 40-50% in melanoma
-Transferred cells remain for up to 4 months
-Careful selection of T cell for tumour antigen to prevent autoimmunity

22
Q

How do T cells improve response to melanoma?

A

The tumour is surgically removed from the patient. Tumour fragments are separately cultured in vitro with IL-2 to activate the T cells infiltrating the tumour. The lymphocytes expand in number and kill the melanoma cells. Individual T-cell cultures are tested for the presence of melanoma-specific T cells. The cultures containing tumour-specific T cells are activated to proliferate, generating up to 1010 tumour-specific effector T cells, which are frozen in aliquots. For 5 to 8 days the patient is conditioned with cyclophosphamide and fludarabine, immunosuppressive drugs that deplete lymphocytes from the circulation. An aliquot of tumour-specific T cells is then infused into the patient.

23
Q

What is CAR T-cell therapy?

A

CAR (chimeric antigen receptors) are genetically engineered protein constructs that can be incorporated into a patient’s own cytotoxic T cells with the goal of helping them recognize and fight cancer cells. This technology creates a new T cell receptor that binds to antigens found on tumour cells and activates the T cell in response to that binding. CAR T cell therapy is provided by removing or harvesting T cells from a patient with cancer, transfecting the cells with CAR genes that are directed against the patient’s tumour type, expanding the modified T cell population, and reinfusing the cells back into the patient.

24
Q

How are B cells killed by T cells expressing CAR?

A

The target antigen on the B-cell tumour cells is CD19. The antigen-binding site of the CAR polypeptide comprises the VH and VL regions from a high-affinity human anti-CD19 monoclonal antibody. These are linked to a transmembrane region and a cytoplasmic tail containing three signaling motifs derived from three activating receptors: CD28, the ζ chain of the T-cell receptor complex, and CD137. The patient’s T cells are activated in vitro and transfected with a retroviral vector carrying the DNA construct encoding the CAR. The transfected cells are then infused into the patient’s circulation.

25
Q

What is the course of CAR T immunotherapy?

A

A CD8 CAR T cell interacting with the CD19 antigen of a B-tumour cell and killing the tumour cell. The in vivo time course of the immune response of a patient’s CAR T cells to the B-cell tumour. IL-6, IFN-γ, and IL-15 are inflammatory cytokines and markers of inflammation. GM-CSF is a homeostatic cytokine. IL-10 is an anti-inflammatory cytokine. Granzyme B is a cytotoxic effector molecule produced by CD8 T cells. IL-8 and MIP-1α are chemokines. CRP, C-reactive protein.

26
Q

What are dendritic cell vaccines?

A

Dendritic cells are a type of antigen presenting cell. Dendritic cells take up antigen and present this antigen on MHC class II to T cells thus inducing an adaptive immune response. Tumour cells are normal cells that due to mutations divide unceasingly. They resemble normal cells and possess normal cell molecules thus they are not killed by the immune response. Tumour cells also make factors that down regulate the immune response. Some tumour cells do express antigens that can initiate an immune response. Peripheral Blood mononuclear cells (Monocytes, NK cells and lymphocytes) are isolated from the blood of a patient. Dendritic cell progenitors are negatively selected using magnetic beads (CD3, CD11b, CD16). These cells are cultured in the presence of the growth factors GM-CSF and TNF for 9 days.
Tumour cells from the patient or tumour specific antigens are added to the cells and they are re-infused back into the patient. The antigen pulsed dendritic cells will now stimulate an adaptive immune response. There are many clinical and research studies currently being undertaken using DC cancer vaccines. For example, 7/17 patients with late stage kidney cancer developed tumour specific IR after DC administration. Four of these patients went into complete remission. Normally less than 10% survive after conventional therapy.

Dendritic cell therapy involves injecting isolated dendritic cells loaded with tumour lysates or tumour antigens or peptides derived from them. Considerable success has been achieved in animal models and increasingly with human patients. The large numbers of dendritic cells needed are obtained by expanding the CD34 positive precursors in bone marrow by culture with GM-CSF, IL-4 and TNF, and sometimes stem cell factor. A mixture of recombinant proteins to load the DC will be beneficial, as it will stimulate more CTLs and better able to deal with any new tumour mutations. Procedure is costly-but so is conventional. One question that has been raised is why does administration of small numbers of antigen pulsed dendritic cells induce specific T cells and tumour regression in patients in whom both the antigen and dendritic cells are already plentiful? This is thought to be because DCs in or near malignant tissues may be defective due to vascular endothelium growth factor (VEGF) or IL-10 secretion by the tumour which may arrest DC maturation and produce tolerogenic immature DCs.

27
Q

What happens with adoptive transfer of dendritic cells loading in vitro with tumour antigens?

A

Monocytes are isolated from a blood sample obtained from a patient with prostate cancer and cultured with sipuleucel-T, a fusion protein of a prostate cancer antigen (prostatic acid phosphatase; PAP) and GM-CSF, a monocyte growth factor. The monocytes differentiate into dendritic cells that present peptides derived from PAP on MHC class II. The dendritic cells are harvested and infused back into the patient’s circulation.

28
Q

What are hepatitis B vaccines?

A

Vaccination to prevent against virus associated cancer has been enormously successful. One such example is the hep b subunit vaccine, which protects against the development of liver cancer. A chronic infection with hepatitis B virus increases the risk of developing liver cancer about 200-fold. Approximately 20% of long-term hep B carriers eventually develop liver cancer. Hep b is one of the most infectious of all viruses transfer of a fraction of a drop of a blood is sufficient to transfer the virus from one human to another. A subunit vaccine that protects against Hep B viruses have been given in the USA since 1982 to healthcare professionals who routinely come into contact with blood and blood products, and to children. This means that if infection with hep B does occur, the immune system is prepared and can quickly eradicate the virus, effectively preventing hep B associated liver cancer.

29
Q

What are HPV vaccines?

A

Human Papilloma Virus (HPV) caused 250,000 deaths from cervical cancer worldwide per year prior to HPV vaccination. Around 12 types of HPV, but HPV-16 and HPV-18 responsible for 70% of all cases. Merck trial vaccinated 500 women 16-23. 5 years later 0 had precancerous lesions, whereas 6 of unvaccinated controls had. July 2018 Advisory Committee stated that HPV vaccine should be given to boys.
HPV vaccination (Gardasil: 6, 11, 16 and 18 in UK), to prevent cervical cancer, anal cancer, genital cancers, and cancers of the head and neck.

30
Q

What is the immunotherapy of cancer?

A

Stimulation of cell-mediated immunity:
Vaccination with viral antigens from oncogenic viruses-eg EBV (Epstein-Barr virus) to target Burkitt’s lymphoma in development.

Immunization with whole tumour cells:
Advantage - Tumour antigen does not need to be identified. Disadv: tumours weakly immunogenic. Human clinical trials have been unsuccessful. Transfection with B7 in animal models improved response. Similarly, BCG plus irradiated melanoma cells had improved response-26% of vaccinated were alive at 5 years, whereas only 6% of those who had best available treatment.

31
Q

How do cytokines function as cancer immunotherapeutic?

A

Antigen independent cytokine therapy use cytokines such as IL-2, interferon and TNF to boost the immune system non-specifically (ie not in an antigen dependent manner).

Interleukin treatment:
-High doses of IL-2 have been given to patients with metastatic melanoma or kidney cancer, and at least partial tumour regression was observed in 15-20% of patients, with some patients displaying complete regression. This is thought to be due to stimulation of pre-existing T cells or due to NK activation.
-Despite the introduction of newer immunotherapies, such as checkpoint inhibitors, they haven’t managed to produce the durable long term responses achieved with Proleukin in metastic melanoma and metastatic renal cell carcinoma
-Complete response rates were 7% in mRCC patients and 6% in mM patients
-Partial regression in 15-20% of mRCC and mM patients

32
Q

What are the response rates for interleukin treatment?

A

IFNα and IFNβ response rates: 10-15% in renal carcinoma, 20% Kaposi’s sarcoma, 40% in lymphomas, 80-90% hairy cell leukaemia. Mechanisms: antiproliferative, activate NK and macrophage, increase MHC class I expression, antiviral.
Colony stimulating factors e.g. GM-CSF* to induce tumour cell differentiation and suppress tumour growth-worked in mice, trialled in humans.

33
Q

How are natural killer cell used for cancer therapy?

A

NK cells are important in tumour surveillance and killing-therefore in vivo expansion and transfer of large numbers of activated NK cells may be beneficial, just like what we saw with Adoptive t cell transfer-however this has not yet been tested. Trials have shown that daily administration of low dose IL-2 following high dose chemotherapy did expand NK cell numbers, however the cells were not maximally toxic. NK cells from related haploidentical donors have been given to acute myeloid leuk patients with a poor prognosis-the idea was to achieve a partial mismatch between the donor NKs and the recipient that may provoke NK activation and greater tumour kill as a result-a very positive response was obtained with 5 of 19 patients going into complete remission.

34
Q

What is vaccination against neovascularistion?

A

Solid tumours cannot grow to any appreciable size without a blood supply so they stimulate the production of new blood vessels by secreting angiogenic factors such as VEGF (vascular endothelial growth factor) that stimulate endothelial cell proliferation. So one treatment rationale is that if you target the antigens expressed in the blood vessels, you should deprive the tumour of oxygen and nutrients and cause tumour regression. Using antibodies to either VEGF or its receptor has not proven useful in humans due to delivery of insufficient antibodies. An alternative strategy involves breaking immune tolerance to VEGF-RD positive endothelial cells by pulsing in vitro generated dendritic cells with soluble VEGF-R2, followed by transferring these cells back into the animal. A major advantage of this approach is that the tumour endothelium is genetically stable as it is non transformed tissue, whereas the tumour is genetically unstable so this makes it unlikely that mutant cells will arise that have lost VEGF-R2 expression.

Treatment of leukaemia:
Leukaemia’s are treated by bone marrow transplantation (BMT) and this is classed as a type of immunotherapy as it involves removal of a patients bone marrow, and replacement of these cells. There are 2 types of BMT:
Autologous-cytoablative treatment using radiotherapy and chemotherapy destroy bone marrow stem cells, and leukemic cells are removed by antibodies, or hemopoietic stem cell isolated by using antibodies to the Cd34 marker.

35
Q

What is the treatment of leukaemia - suicide gene therapy?

A

Allogeneic BMT involves transplantation of bone marrow from a reasonably compatible MHC donor and this results in an important graft versus leukemia effect. Unfortunately the graft also launches an attack on the recipient this is known as graft versus host disease which is a complication of this procedure. However if you removed the T cells to prevent the graft versus host, you would also lose the beneficial graft versus leukaemia effect.

One possible alternative - Stem cells from the allogeneic bone marrow, which has had its T cells removed, are given the donor T cells tha have been transfected with herpes simplex virus thymidine kinase. The t cells provide factors that facilitate engraftment, defence against viral infection and the graft versus leuk effect at a time when the patient has a low tumour burden (to get rid of any residual disease). With time, graft versus host disease develops, however this can be stopped by giving the patient gangliclovir. Gangiclovir is converted by the thymidine kinase into a nucleoside analog which is toxic for dividing cells.

36
Q

What is the best description of sipuleucel-T?

A

A fusion protein of GM-CSF and prostatic acid phosphate.

37
Q

What are the characteristics of Hodingkins, anaplastic large cell lymphoma, Burkitts lymphoma?

A

Hodingkins:
-Owl eyes
-PAX-5 missing
-Reed-sternberg cells
-MYC proto oncogene and ch8

ALCL:
-T cell lymphoma
-Horseshoe
-ALK gene on ch 2 and NPM on ch 5
-CD30 expression

Burkitts lymphoma:
-Germinal B cell
-Starry sky
-MYC protooncogene ch 8