CBIO 8: Cancer Therapy Flashcards

You may prefer our related Brainscape-certified 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 are the five pillars of cancer treatment?

A
  • surgery
  • radiotherapy
  • chemotherapy
  • immunotherapy
  • targeted therapy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What is the purpose of cancer treatment?

A
  • prolong survival time
  • improve quality of life
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What are the three key goals of cancer treatment?

A
  • cure
  • control
  • palliative care

The aim is always to eliminate cancer cells and reduce the chance of recurrence but in cases where cancer cannot be cured, palliative treatment is important.

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

What type of treatment is most commonly used in the UK?

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

Give an example of the treatment strategy for breast cancer

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

Define neoadjuvant therapy

A
  • treatment given to shrink the tumour before the primary treaatment
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Define adjuvant therapy

A
  • treatment given in addition to the primary treatment
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Define cancer stage

A
  • describes the size of tumour and how far it has spread from the original site
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Define cancer grade

A
  • described the appearance of tumour compared to original normal cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Describe complete remission

A
  • treatment has eliminated cancer as measured by medical tests
  • does not mean a cure
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Define anti-angiogenic

A
  • drugs to stop tumours growing their own blood vessels
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What are the different functions of surgery in cancer treatment?

A
  • cancer prevention
  • diagnosis
  • staging
  • primary treatment
  • debulking
  • relieving symptoms or side effects
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Describes the aims of surgery in cancer treatment

A
  • Surgery is often used with curative intent.
  • It is usually the first line of treatment for many patients that have localised solid tumours, or tumours caught at an early stage of development.
  • However it is not suitable if the cancer is systemic such as in leukaemia (haematological cancer) or lymphatic cancer.
  • It is also less suitable for metastasised cancer, or in case the tumour is near a risky or delicate area, such as a major blood vessel.
  • The aim of surgery is to remove the tumour mass or the whole host organ, sometimes including the surrounding lymphatic system.
  • A margin of healthy tissue is also removed to reduce the chance of recurrence.
  • Surgery alone can be used as cancer treatment, but it’s often used in combination with other modalities.
  • For example, surgery can be performed before chemotherapy or radiotherapy as an adjuvant treatment, or radiation can be given to reduce the tumour size and then surgery can be performed to remove it (neoadjuvant treatment).
  • Surgery can also be performed to control symptoms or extend life/improve quality of life, for example to remove the tumour even though it is not a cure.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Describe the different types of surgery in cancer treatment

A
  • debulking:
  • Refers to surgery that removes as much of the tumour as possible but not all of it.
  • This improves the chances of successful chemo- or radiotherapy, for example in the case of advanced cancer of the ovary.
  • laparoscopic surgery:
  • This type of surgery is less invasive as it is carried out through smaller incisions.
  • Also referred to as ‘keyhole’ surgery, laparoscopic surgery uses a specialised instrument called a laparoscope.
  • A laparoscope is a small tube with a light source and a camera, which relays images of the inside of the abdomen or pelvis to a television screen for the surgeons to monitor their progress.
  • radical surgery:
  • To lower the chance of recurrence, radical surgery will remove all nearby tissue including lymph nodes, muscles and nerves, for example radical mastectomy is the removal of the breast and surrounding tissue.
  • see image
  • preventative (prophylactic) surgery:
  • Surgery to remove non-cancerous areas of tissue in patients who are genetically at a high risk of developing particular cancers.
  • Two examples are patients with a family history of breast cancer (BRCA1 and BRCA2) and familial adenomatous polyposis (a condition where large numbers of polyps form, mainly in the epithelium of the large intestine).
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What are the main risks associated with cancer surgery?

A
  • Bleeding
  • Blood clots
  • Damage to nearby tissues, nerves
  • Adverse reactions to drugs used in surgery
  • Damage to other organs
  • Pain
  • Infections
  • Slow recovery of other body functions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What is radiotherapy?

A
  • Radiotherapy uses ionising radiation (IR), normally delivered by a linear accelerator, to cause DNA damage and kill malignant cells.
  • Radiotherapy can be curative in many cases where the tumour is localised and it can be used instead of surgery to remove a tumour in cases where the patient is too weak to undergo surgery.
  • Radiation is delivered to match the 3D shape of the tumour so that intense radiation is delivered to tumour cells with less exposure to the surrounding healthy tissue.
  • Not all cancer can be treated with radiotherapy - some cancers are radio-resistant, such as renal cell carcinoma.
  • Metastatic cancer is also less suitable for radiotherapy treatment due to its systemic nature.
  • Radiotherapy is usually given as doses over days, allowing time in between treatments for the normal cells that have been exposed to IR to repair and recover (reduce side effects).
  • It can also be adjuvant or neoadjuvant with other cancer therapies.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Describe the types of radiotherapy used in cancer treatment

A
  • 3DCRT (three-dimensional conformal radiotherapy):
  • This is the most commonly used radiotherapy
  • It uses computer programmes to analyse and design radiation beams to match the shape of the tumour.
  • IMRT (intensity-modulated radiation therapy):
  • Uses linear accelerators to deliver precise radiation doses to the 3D shape of the tumour.
  • The 3D shape of the tumour is divided into segments and during treatment different intensities of radiation are delivered to each segment via computer-controlled modulation of multiple radiation beams.
  • IGRT (image guided radiotherapy):
  • Is used to treat tumours in areas that move, such as the lungs or close to critical organs.
  • Therefore frequent imaging is taken before and during radiotherapy to assist precise delivery of irradiation to the tumour.
  • SBRT (stereotactic body radiation therapy):
  • Uses imaging and computer programmes to precisely deliver a higher dose (higher than other types of radiotherapy) of radiation.
  • It is usually given in a single or small number of treatments, where delivery is accurate to within one to two millimetres.
  • It is also used as an alternative to open surgery for removal of small to moderately-sized cancers.
  • brachytherapy:
  • A radioisotope source in a sealed container such as a capsule is placed in or near the tumour.
  • see image
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

How does radiotherapy work to kill cancer cells at the molecular level?

A
  • Ionising radiation consists of particles, X-rays, or gamma rays with sufficient energy to cause ionisation.
  • This means that during an interaction with an atom, it can remove tightly bound electrons from the orbit of the atom, causing the atom to become charged or ionised.
  • During radiotherapy, using photons, protons or particle radiation, the ionising radiation can damage the DNA directly.
  • It can also have indirect effects such as affecting water molecules, which become free radicals that can themselves damage the DNA (see diagram below).
  • Although cells have mechanisms in place to repair DNA damage, these are often significantly disrupted in cancer cells.
  • As a consequence, DNA damage is more likely to trigger cell death signals in cancer cells.
  • Also, the sheer amount of DNA damage induced can overwhelm the repair machinery.
  • Whilst IR causes several types of DNA damage including single-stranded DNA breaks, it is the double stranded DNA breaks that are responsible for inducing cell death.
  • IR induces several cell death mechanisms
  • apoptosis and mitotic catastrophe are the main mechanisms, but necrosis, IR-induced senescence followed by apoptosis, and IR-induced autophagy followed by apoptosis are also triggered by IR.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Define mitotic catastrophe

A
  • a type of cell death that occurs during mitosis
  • results from premature or inappropriate entry of cells into mitosis
  • can be caused by chemical or physical stresses
  • it is unrelated to programmed cell death or apoptosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Define senescence

A
  • cells cease to divide and grow
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Define autophagy

A
  • A natural process to self-degrade cellular components, important for balancing energy during critical times such as nutrient stress.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What happens to healthy cells adjacent to cancer cells exposed to IR?

A
  • Healthy cells adjacent to cancer cells or in the path of radiation are exposed to some IR.
  • These normal cells proliferate more slowly and therefore have time to repair DNA damage before replication.
  • They can also repair themselves at a faster rate and retain their normal function more effectively than cancer cells.
  • However, even though healthy tissue is better at recovering from IR it can nevertheless be damaged resulting in short or long term side-effects.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What are some general side effects of radiotherapy?

A
  • Sore, red skin
  • Feeling tired most of the time (due to low red blood cells or body is using most energy to repair healthy tissue)
  • Hair loss in the area being treated (IR affects cells that proliferate at a faster rate, such as hair folicles)
  • Feeling sick
  • Loss of appetite
  • A sore mouth
  • Diarrhoea
  • Lymphoedema (swelling of limbs due to damage to the lymphatic system that normally regulates body fluid)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What types of genetic mutations cause cancer?

A
  • Chromosome translocation
  • Gene amplification (copy number variation)
  • Point mutations within promoter or enhancer regions of genes
  • Deletions or insertions
  • Epigenetic alterations to gene expression
  • Germline mutations (can be inherited)
26
Q

What is chemotherapy?

How does it work?

A
  • Chemotherapy is the use of cytotoxic drugs to kill cancer cells.
  • It is a systemic treatment, meaning the drugs travel through the blood stream and affect cells all over the body.
  • This form of treatment is administered intravenously or by mouth and is given in cycles, with a recovery period of months.
  • Chemotherapy targets dividing cells at various stages of the cell cycle.
  • As it targets rapidly dividing cells, normal rapidly dividing cells such as those in the gastrointestinal tract can also be affected, and this leads to some of the nasty side effects seen with chemo.
27
Q

What are the normal cells most likely damaged by chemo?

A
  • Cells in the mouth, digestive tract, and reproductive system
  • Blood-forming cells in the bone marrow
  • Hair follicles
28
Q

Is chemotherapy given alone or in combination with other treatments?

Give some examples

A
  • Chemotherapy can be given alone or as an adjuvant or neoadjuvant treatment and the patient is usually given several drugs in combination.
  • Occasionally it is also combined with other medicines, such as steroids or biological therapies.
29
Q

Describe the chemotherapy: alkylating agents

A
  • Nitrogen mustards were the first alkylating agent used medically and also the first chemotherapy drug.
  • Alkylating agents add alkyl (CNH2N+1) groups to guanine residues in DNA which causes cross-linking of DNA strands thus preventing DNA from uncoiling at replication.
  • The replication arrest activated by checkpoint pathways triggers apoptosis.
  • Pseudo-alkylating agents add platinum to guanine residues in DNA, which activate the checkpoint and trigger apoptosis.
  • Most of the alkylating agents are also carcinogenic as they encourage mispairing during DNA repair.
  • They are toxic to normal cells, particularly cells that divide frequently, such as those in the gastrointestinal tract, bone marrow, and in the testicles and ovaries, which can cause loss of fertility.
30
Q

What are the side effects of alkylating agents?

A
  • hair loss (not carboplatin)
  • nephrotoxicity (toxicity in the kidneys)
  • neurotoxicity
  • ototoxicity (platinums) (toxic to the ear)
  • nausea
  • vomiting
  • diarrhoea
  • immunosuppression
  • tiredness
31
Q

Describe the chemotherapy: antimetabolites

A
  • Antimetabolites are drugs that interfere with enzymes that are necessary for DNA synthesis.
  • They are a group of drugs that masquerade as purine (adenine and guanine) or pyrimidine (thymine/uracil and cytosine) residues, leading to the inhibition of DNA replication (DNA-DNA) and transcription (DNA–RNA).
  • In addition, antimetabolites can be folate antagonists which inhibit the dihydrofolate reductase required to make folic acid.
  • Folic acid is an important building block for all nucleic acids especially thymine.
  • As a consequence, antimetabolites induce cell death during S phase of the cell cycle, when DNA is synthesised.
32
Q

What are the side effects of antimetabolites?

A
  • Fatigue
  • Hair loss (alopecia)
  • Bone marrow suppression causing anaemia, abnormally low white blood cells and low platelet count
  • Increased risk of neutropenic sepsis (a potentially fatal inflammatory response to infection) or bleeding
  • Nausea and vomiting (dehydration)
  • Mucositis and diarrhoea
  • Palmar-plantar erythrodysesthesia (PPE) (reddening, swelling, numbness and skin sloughing or peeling on palms of the hands and soles of the feet)
33
Q

Describe the chemotherapy: anti-microtubule agents

A
  • Antimicrotubule agents are originally derived from natural sources.
  • They inhibit cell proliferation by binding to microtubules and suppressing microtubule dynamics during the particularly vulnerable mitotic stage of the cell cycle.
  • Vinca alkaloids and taxanes are the two main groups of anti-microtubules.
  • They work by inhibiting assembly (vinca alkaloids) or disassembly (taxanes such as Paclitaxel) of mitotic microtubules.
  • This causes dividing cells to undergo mitotic arrest which activates cell death.
  • Antimicrotubules are also antiangiogenic which means they prevent tumour vasculature forming and have vascular-disrupting effects.
34
Q

What are the side effects of anti-microtubule agents?

A
  • Nerve damage: peripheral neuropathy, autonomic neuropathy
  • Hair loss
  • Nausea
  • Vomiting
  • Bone marrow suppression (also called myelosuppression, decrease in the production of blood cells in bone marrow)
  • Arthralgia (pain in joints)
  • Allergy
35
Q

Describe the chemotherapy: topoisomerase inhibitors

A
  • Topoisomerases are enzymes that regulate the unwinding of DNA.
  • They are required to prevent DNA torsional strain during DNA replication and transcription.
  • They induce temporary single-strand (topoisomerase I) or double-stranded (topoisomerase II) breaks in the phosphodiester backbone of DNA.
  • Topoisomerase I cuts one of the DNA strands, relaxes the strand and then re-ligates the strand.
  • Topoisomerase II cuts a double-stranded break in one DNA segment, to allow another DNA segment to pass through, and re-ligates the first DNA segment.
  • Topoisomerase inhibitors are drugs that bind to the enzyme and prevent the re-ligation, which means DNA remains cut.
  • There are DNA repair mechanisms in place within cells for double-stranded breaks, such as the Non-Homologous End Joining pathway (NHEJ).
  • However, chemotherapeutic topoisomerase inhibitors will cause significant single- and double-stranded DNA breaks that will overwhelm the repair mechanisms and therefore trigger apoptosis of the cancer cells.
36
Q

What are the side effects of topoisomerase inhibitors?

A
  • (Irinotecan): Acute cholinergic type syndrome – diarrhoea, abdominal cramps and diaphoresis (sweating).
    • Therefore given with atropine, which is a medication to counteract the side effects (by blocking the aberrant nerve impulses).
  • Hair loss
  • Nausea, vomiting
  • Fatigue
  • Bone marrow suppression
37
Q

Describe the chemotherapy: cytotoxic antibiotics

A
  • Cytotoxic antibiotics are a group of naturally-occurring drugs with antibiotic activity that are also effective at killing cancer cells.
  • Although they have varying mechanisms of action, they generally interrupt cell division.
  • The two main groups are Anthracyclins and Bleomycins.
  • Anthracyclins include Doxorubicin and Daunorubicin from the bacterium Streptomyces peucetius.
  • The main mechanisms of action for this treatment include intercalating DNA that prevents transcription and DNA synthesis, inhibiting topoisomerase II, producing free radicals that cause DNA damage and damaging cell membranes.
  • For example, Bleomycin is isolated from Streptomyces verticillus and its mechanism of action is to produce free radicals that cause DNA damage.
38
Q

What are the side effects of cytotoxic antibiotics?

A
  • Cardiac toxicity (arrhythmias, heart failure) – probably due to damage induced by free radicals
  • Alopecia
  • Neutropaenia
  • Nausea and Vomiting
  • Fatigue
  • Skin changes
  • Red urine (doxorubicin is nicknamed “the red devil”)
39
Q

What is a significant risk with chemotherapy?

A
  • cells that are not killed by chemotherapy are at risk of mutation.
  • These mutations may result in the cancer cells developing resistance to this type of treatment.
40
Q

What are some mechanisms by which tumour cells might develop resistance to chemotherapy drugs?

A
  • Increased detoxification (or clearing) of the drug
  • Inhibition/loss of apoptosis
  • Increased DNA repair of damage caused by chemo
41
Q

What are the four main types of targeted therapy?

A
  • Cancer growth blockers
  • Monoclonal antibodies (MAOs)
  • Anti-angiogenics
  • PARP inhibitors (PARPi)
42
Q

What are cancer growth blockers?

A
  • they are a group of drugs that target the growth signals of cancer cells
43
Q

Describe the types of cancer growth blockers

A
  • tyrosine kinase inhibitors:
  • Tyrosine kinases are enzymes that when activated, transfer a phosphate group from ATP to a protein involved in the signal transduction cascade.
  • As these enzymes help to send growth signals in cells, blocking them with drugs stops the cell growing and dividing.
  • Gleevec is an example of tyrosine kinase inhibitor.
  • proteasome inhibitors:
  • Drugs to inhibit the proteasome machinery that normally breaks down unwanted cellular proteins.
  • mTOR inhibitors:
  • Within a cell mTOR signalling is activated by growth factors such as insulin and it increases cell size and proliferation by producing new proteins and controlling cytoskeletal reorganisation.
  • mTOR inhibitors are drugs that inhibit the protein kinase mTOR.
  • PI3K inhibitors:
  • Drugs that target PI3K family of enzymes.
  • They are involved in many cellular processes including cell growth, proliferation, differentiation, motility, survival and intracellular trafficking PI3K signaling is often altered to in cancer.
  • Histone deacetylase inhibitors:
  • Histone deacetylase are a group of enzymes that control the coiling and uncoiling of DNA around histones required for gene expression and transcription.
  • Hedgehog pathway blockers:
  • The hedgehog pathway is an embryonic signaling pathway involved in proper differentiation of cells.
  • This signalling pathway is activated in the development of some cancer types, including brain and skin cancer.
44
Q

Briefly describe the tyrosine kinase inhibitor: Gleevec

A
  • Gleevec was the first “targeted therapy” (also known as Glivec and Imatinib).
  • In 1973, the human geneticist Dr Janet Rowley became the first scientist to identify a chromosomal translocation as the cause of cancer in patients with chronic myeloid leukaemia.
  • The chromosomal translocation was found to create its own unique fusion protein called Bcr-Abl, an enzyme which drives over-production of white blood cells (see diagram below).
  • This is an example of “Oncogene Addiction” – a uniquely hyperactive oncogene driving a tumour (Achilles’ heel).
  • In 1996 Buchdunger et al published data on a drug that could specifically target Bcr-Abl (and not affect other proteins).
  • Fantastic clinical results (90% complete response rates in patients with CML) heralded the departure from “conventional cytotoxics” into a new era of “targeted therapies”.
  • In 2017, a 10 year follow-up study reported that the overall survival rate was 83.3%.
45
Q

How does Gleevic inhibit Bcr-Abl enzyme activity?

A
  • The 9;22 chromosome translocation is named the Philadelphia chromosome.
  • The translocation creates the fusion protein Bcr-Abl which is a membrane-associated protein with ‘always on’ tyrosine kinase activity.
  • It speeds up cell division through the regulation of cell cycle proteins.
  • Gleevec works by binding close to the ATP binding site of Bcr-Abl, preventing the enzyme binding to ATP and inhibiting the enzyme activity.
  • Gleevec is quite selective for Bcr-Abl, though it does also inhibit other targets such as c-Kit and the PDGF-Receptor.
  • C-Kit is mutated in gastrointestinal stromal tumours (GIST) and Gleevec is used in therapy, however resistance to Gleevec unfortunately occurs (75% at 2.5 years).
46
Q

Describe receptor tyrosine kinase signalling pathways

A
  • Receptor tyrosine kinases are a large family of tyrosine kinases that include EGFR (epidermal growth factor receptor).
  • On the membrane, external growth factors (such as EGF) bind to and activate the Receptor Tyrosine Kinases (RTK).
  • Inside the cell, the tyrosine kinase component of RTK transduce the signal via components of the PI3 kinase (left) and MAP kinase (right) pathways to affect cellular processes such as the promotion of proliferation.
  • As you can see most oncoproteins in carcinomas are components of the RTK signalling pathways and therefore targeting RTK signalling is a good idea.
47
Q

Describe the targeted therapy: monoclonal antibodies

What are their limitations?

A
  • Monoclonal antibodies (mAbs) are directed against antigens located on the surface of tumour cells (TAAs/TSAs, see CBIO7).
  • A limitation in using monoclonal antibodies is that, as antibodies themselves are proteins, they can sometimes cause something like an allergic reaction.
48
Q

What are the types of monoclonal antibodies used in cancer treatment?

A
  • Naked monoclonal antibodies: these are antibodies that work by themselves.
  • There is no other therapeutic module attached to them. These are the most common type of mAbs used to treat cancer.
  • They can, for instance, trigger Antibody-Dependent Cell-mediated Cytotoxicity (ADCC), or simply block cancer-associated growth factor receptors.
  • Conjugated monoclonal antibodies:
  • these are antibodies joined to a chemotherapy drug or to a radioactive particle.
  • The mAb is used as a homing device to take one of these substances directly to the cancer cells.
  • Bispecific monoclonal antibodies:
  • these are made up of parts of 2 different mAbs, meaning they can attach to 2 different proteins at the same time.
  • By binding to both proteins, this drug brings the cancer cells and immune cells together, which is thought to cause the immune system to attack the cancer cells.
49
Q

Describe the targeted therapy: anti-angiogenic therapy

What are the two types?

A
  • When a tumour has reached 1 to 2 mm across it requires its own blood supply.
  • Therefore in order to survive, tumour cells produce and release pro-angiogenic factors such as VEGF, that act on endothelial cells found lining the blood vessels.
  • This encourages the vessels to grow towards, and supply blood to, the tumour
  • Drugs that block blood vessel growth factors are called anti-angiogenics.
  • There are two types of anti-angiogenics:
    1. Drugs that block blood vessel growth factor, for example by preventing VEGF from binding to the blood vessel cell receptors
    2. Drugs that block signalling within the cell, these are the tyrosine kinase inhibitors
50
Q

What is synthetic lethality?

A
  • Synthetic lethality is a simple genetic concept to help identify new cancer treatment targets.
  • The concept originated in genetic studies of model organisms such as fruit flies.
  • The idea is that loss of cellular viability occurs when the interaction between two genes is disrupted simultaneously but cells survive when either gene alone is disrupted.
  • This is ideal for cancer treatment as cancer cells usually harbor genetically altered genes that are not present in healthy cells.
  • Since the synthetic lethality concept was proposed 20 years ago there is only one successful story that has been translated into clinical therapy.
  • This is the use of PARP inhibitors (PARPi) in patients with BRCA1/BRCA2 deleted or mutated cancer.
51
Q

How do PARP inhibitors work?

A
  • Poly (ADP-ribose) polymerases (PARPs) constitute a large family of 18 proteins.
  • PARP1 and PARP2 are enzymes, activated by DNA damage, and facilitate DNA repair of single-strand breaks (SSBs) and base excision repair (BER).
  • Suppression of PARP leads to stalling of replication forks due to the accumulation of unrepaired SSBs.
  • The stalled replication forks degrade into highly cytotoxic double strand breaks (DSBs) which are normally repaired by homologous recombination (HR), considered to be the most precise DSB repair mechanism.
  • BRCA-mutated cells (either germline or somatic) are incapable of HR, therefore PARP inhibition results in genomic instability and cell death.
52
Q

How effective are the PARP inhibitors?

A
  • Olaparib (Lynparza)
  • In phase 1 of a clinical trial for Olaparib, including patients with germline BRCA1 or BRCA2 mutations, 63% of the patients demonstrated a clinical benefit.
  • Myelosuppression (bone marrow activity is decreased) and central nervous system side effects were observed in some patients, but overall, side effects were less severe than those seen with chemotherapy.
  • Phase 2 trials involving patients with germline BRCA mutations of breast, ovarian, pancreatic, or prostate cancers confirmed that Olaparib offered a clinical benefit.
  • Recently a clinical trial found it to be useful in patients with BRCA-mutated (HER2)-negative metastatic breast cancer who had been previously been treated with chemotherapy in the neoadjuvant, adjuvant or metastatic setting.
53
Q

How are the PARP inhibitors clinically useful?

A
  • In 2014 both the European Medicines Agency (EMA) and U.S. Food and Drug Administration (FDA) approved Olaparib as a treatment for patients with advanced ovarian cancer.
  • In addition EMA approved Olaparib for fallopian tube, or primary peritoneal cancers.
  • More recently, in 2018 (FDA) and 2019 (EMA), Olaparib was approved to treat germline BRCA-mutated, HER2-negative metastatic breast cancer.
  • FDA approval for germline BRCA-mutated metastatic pancreatic cancer has since followed and the list of indications that benefit from Olaparib treatment continues to grow.
54
Q

What are the limitations of PARP inhibitors?

A
  • The PARP inhibitors were initially hypothesised to be effective in breast cancer treatment as about 10% of breast cancers are hereditary due to BRCA 1/2 mutations.
  • However in a clinical trial for patients with Triple negative breast cancer (TNBC), the responses to Olaparib were somewhat mixed; some patients showed some disease stabilisation but no sustained responses were recorded.
  • In addition, resistance to PARP inhibitors can occur.
  • The mechanism of resistance is not well understood but involves the restoration of homologous recombination (HR) repair in tumour cells.
55
Q

What is the future for PARP inhibitors?

A
  • combination therapies:
  • Combining PARPi with targeted agents such as ATR inhibitor. ATR (ataxia–telangiectasia and Rad3 related) is a protein involved in cell cycle arrest that allows time for DNA repair.
  • Combination of PARPi with immunotherapies such as antibodies.
  • Stratified trials – there is some evidence that PARP inhibitors are more effective in tumours with mutations in the DNA damage repair pathways, and these responses may be “swamped” in large trials unless patients are selected on the basis of having mutations that predispose them to benefit.
  • This is an example of stratified, or personalised, medicine.
56
Q

What are some new and emerging therapies?

A
  • PROTACS
  • DNA cages
  • Cannabis
  • Gut microbiome and cancer treatment
  • ZIKA virus oncolytic therapy
  • cryotherapy
57
Q

Describe the emerging cancer therapy: PROTACs

A
  • PROTAC: PROteolysis TArgeting Chimera
  • Targets cellular proteins for degradation by co-opting the ubiquitin-proteasome system

-> chemical knockdown approach

  • Improved selectivity compared to classic inhibitors
  • Higher sensitivity to drug resistant targets
  • PROTACs have been successfully developed for >50 cancer relevant proteins
  • Clinical trials ongoing
58
Q

Describe the emerging cancer therapy: DNA cages

A
  • Nanostructures modified with lipid-like molecules – DNA origami
  • Lipids can act like sticky patches that come together and create a “handshake” inside the DNA cube, creating a core that can hold cargo such as drug molecules
  • A DNA cube can carry drug cargo to a diseased cell environment.
  • This triggers the release of the drug when it encounters particular DNA sequences

• Can also be used for in vivo imaging

59
Q

Describe the emerging cancer therapy: Cannabis

A
  • The cannabis plant produces substances known as cannabinoids, which have been explored for their therapeutic potential
  • Two main cannabinoids have been identified:
  • Delta-9-tetrahydrocannabidiol (THC)
  • Cannabidiol (CBD)
  • Cannabinoids bind to receptors in the human body and, in turn, modulate the endocannabinoid system

Cannabinoids have been shown to:

  • Induce apoptosis and inhibit proliferation of cancer cells
  • Stop the development of new blood vessels
  • Modulate metastatic potential
60
Q

Describe the emerging cancer therapy: Gut microbiome

A
  • Increasing evidence that gut microbiome influences both susceptibility to certain cancers as well as response to therapy
  • This influence has most convincingly been shown in immunotherapy
  • Therapeutic strategies to target the gut microbiome include lifestyle changes, administration of probiotics and fecalmicrobiota transplant
61
Q

Describe the emerging cancer therapy: zika virus for oncolytic therapy

A
  • Zika virus is usually harmless in adults, but can target the fast-growing stem cells in the brain of a developing baby. This causes the brain to not grow properly, known as microcephaly
  • Cancerous brain tumours contain very similar stem cells, which are thought to be the source of tumour recurrence
  • The aim of the development of Zika virus oncolytic therapy is to totarget and destroy glioblastoma cells in brains, while leaving healthy brain cells untouched
  • If successful, this could become a much-needed new option for people with these hard-to-treat brain cancer
  • Immunity will be a limiting factor in areas were Zika virus is endemic
62
Q

Describe the emerging cancer therapy: Cryotherapy

A
  • Cryotherapy is a treatment that uses extreme cold to destroy cancer cells

Some facts about cryotherapy:

  • Cryotherapy can be used to treat different types of cancer and precancerous conditions
  • Cryotherapy is a local treatment
  • After cryotherapy the body’s immune system gets rid of the dead tissue
  • For cryotherapy of cancers inside the body, a small probe (cryoprobe) is inserted next to or inside the tumour that is attached to a supply of liquid nitrogen
  • Depending on the treatment area, cryotherapy can take a few minutes to a couple of hours
  • Cryotherapy can be administered through the skin (percutaneously) or through a scope