People and Illness Flashcards
What are the main clinical features which define ADHD?
- Inattention
Lacks of persistence in activities requiring concentration
Selected attention - distraction
Sustained attention - problem-solving - Impulsivity
Inappropriate/defective filtering of information
Poor awareness of risk makes individuals accident-prone
Social disinhibition, excessive talking
Poor peer relationships, aggression, emotional dysregulation - Hyperactivity
Psychomotor agitation
Restless, fidgety, disorganised, ill-regulated
Discuss the influence of genetics on the development of ADHD
Candidate ADHD susceptibility genes: those involved in reward pathways, dopamine regulation
Associated learning disorders: Fragile X, Klinefelter & Williams syndromes
Genetic-environment interactions - genes can increase or reduce the impact of the environment on the development of ADHD
Which characteristics might be present in an fMRI of an individual with ADHD?
Reduced frontoparietal volume
Right dorsolateral prefrontal lobe reduced
Smaller basal ganglia
Smaller cerebellar vermis
Attentional systems involved: anterior fronto-striatal networks
Posterior parieto-cerebellar circuits
Which co-morbidities is ADHD associated with?
- Mood disorders
- Tic disorders e.g. Tourette syndrome
- Insomnia
- Anxiety
- Learning disorders
- Behavioural difficulties: ODD/CD (oppositional defiant disorder/conduct disorder)
- Social communication difficulties
Describe the process of development of attention in children
- 0-12 months: fleeting; focus very briefly, quickly distracted
- 1-2 years: rigid; can attend to task of their own choosing
- 2-3 years: single-channelled; can only focus on one thing at a time
- 3-4 years: focus; attention is single-channelled but child can control this
- 4-5 years: dual-channelled; can do task & listen to someone at same time
- 5+ years: integrated; mature attention control
Briefly outline the process of assessment of ADHD
- No specific diagnostic test, so assessment includes:
- Direct observations in >1 setting: ADHD must be pervasive
- Structured questionnaires given to reliable informants
- Psychoeducational assessment
- Identifying co-morbid mental health problems
- Developmental history
Which structured questionnaires are commonly used in the assessment of ADHD?
- SNAP-IV Teacher and Parent Rating Scale
- Conners Scale for ADHD Assessment
What type of information would be relevant when taking a history for assessment of ADHD?
- Past history
Pregnancy/delivery (any alcohol/illicit drugs?)
Patterns of feeding, sleeping, play
Activity levels, impulsivity, inattention - Medical history
Head injury
Hearing problems
“Tics”/funny turns/seizures - Family history
Learning difficulties in the family
Stressors in the family
Child’s care history
Discuss the use of behavioural parent training in the management of ADHD
- Encourage consistency in managing less desirable behaviour
- Do not personalise behaviour problems
- Routines, countdowns, reminders
- Planned ignoring, time-outs, quiet time
- Positively reinforce appropriate behaviour
- Clear rules with consequences
Discuss the use of behavioural training in schools to manage ADHD
Management of the environment
- Provide a calm environment, reducing background noise
- Avoid too many distracting stimuli when wanting a child to concentrate
- Initially avoid situations that require quiet, still behaviour for long periods of time
- Maintain structure & supervision longer than you think necessary
Behavioural management
- Do not give instructions without first gaining the child’s attention
- Give clear, direct, short instructions & provide visual aids if needed
- Ask child to repeat back instructions
- Improve concentration skills with activities the child enjoys
- Plan ahead for problem situations
- Model good listening skills
Outline the mechanism of action of methylphenidate, including different preparations & dosages
Methylphenidate aka ritalin, concerta, equasym, medikinet
Blocks dopamine and noradrenaline reuptake by blocking transporter: increases NA and DA
Immediate release tablets: 5mg 3-4x a day
Modified release/sustained release preparations available, 8-12h duration
Usually give long-acting in the morning and short-acting in late afternoon
Drug holidays required
Outline the mechanism of action of dexamphetamine, including different preparations & dosages
Dexamphetamine acts to release dopamine stored in presynaptic vesicles, whilst blocking reuptake via transporter, increasing dopamine
Dexedrine 5mg: immediate release; max dose 20-40mg
Elvanse: lisdexamphetamine dimesylate, sustained release, 13h duration
- Increases available NA and DA
- Max dose 70mg
- Pro-drug, converted to dexamphetamine via first-pass metabolism
- Less susceptible to abuse
Outline the mechanism of action of atomoxetine
Noradrenaline reuptake inhibitor: enhances noradrenaline transmission in prefrontal cortex
Takes time to have an effect, approx 6 weeks, but no drug holidays required
Effective for co-morbid anxiety, depression
Outline the mechanism of action of guanfacine, including dosage and preparations
Selective central alpha 2a adrenergic receptor agonist
Stops the effect of noradrenaline at the synapse
Acts on prefrontal cortex, not nucleus accumbens, reducing the potential for abuse
Helpful with sleep & appetite issues; significant improvement within 3 weeks
Oral prolonged release tablet formulation
Basal metabolic rate is higher in children than adults, so dosage relative to body weight is higher/more frequent
Discuss the side-effects of psychostimulants and ways to manage them
- Anorexia, nausea, weight loss, growth concerns
- Sleep difficulties
- Dizziness, headache, abdominal pain
- Involuntary movements or tics
- Dysphoria, agitation
- Tachycardia, hypertension
- Syncope suspected to have cardiac origin
Stop medication & seek specialist advice
Discuss the side-effects of atomoxetine
- Nausea/vomiting
- Excessive tiredness
- Insomnia
- Abdominal pain, appetite suppression, weight loss, constipation
- Headaches
- Mood swings/rage, suicidal ideation
- Hepatic impairment (monitor LFTs)
- Tachycardia, hypertension
Discuss the side-effects of guanfacine and clonidine
Guanfacine
- Sedation
- Dizziness
- Hypotension: monitor BP and heart rate
- Precaution when combined with pscyhostimulants, reports of sudden death
Clonidine
- Paradoxical hypertension
- Paradoxical sleep disturbance
How would you manage side-effects like loss of appetite and insomnia?
Loss of appetite
- Caloric augmentation
- Administer medication with food
- Drug holidays
- Monitor height and weight
- Dietetic advice
Insomnia
- Administer medication earlier in the afternoon or reduce evening dose
- Sleep hygiene advice
- Melatonin
Discuss how ADHD medication monitoring is carried out
- Height and weight: on a growth chart every 6 months
- Heart rate and BP: repeat at every dose adjustment and every 6 months, ECG if treatment affects QT interval
- Complete history Concomitant medicines Past, present medical history Psychiatric disorders Family history of sudden cardiac and unexplained death
- Contraindications
Depression, anorexia, suicidal tendencies, psychosis
Pre-existing cardiovascular disorders
Discuss the legal status of psychostimulant medications
- MPH and dex are schedule 2 controlled drugs (prescription only medicines)
- BNF (British National Formulary)
Prescription and requirements: form and strength of preparation
Total quantity in words & figures
Dose to be administered
28 day validity - signed & dated by prescriber
Signed on collection + proof of identity
30 day supply - Good practice: pharmacist may supply more if requested but prescriber must state why
After titration and dose stabilisation, prescribing and monitoring of ADHD medication is carried out under shared care protocol arrangements with primary care
Discuss the features which can influence the prognosis of ADHD going into adulthood
Prognosis depends on co-morbidity
- Organic disorders
- Psychiatric disorders e.g. ODD/CD
- Learning difficulties
- Which symptoms predominate & environment in which they predominate
Factors associated with persistence into adulthood
- Progressive reduction in cerebellar & hippocampal volumes
- Maternal depression, marital discord, negative parent-child interactions
- Family socioeconomic disadvantage
- Familial ADHD
Discuss the influence of arousal systems on the pathogenesis of ADHD
- Deficiency in arousal mechanisms, defective inhibitory response
- Neurons in prefrontal cortex are out of tune and can’t distinguish between important signals & background noise
- Can’t focus as all the signals are the same - easily distracted
Hypoarousal: low firing of NA and DA neurons
Need to improve the “signal to noise” ratio; increase the drive of the arousal network to improve efficiency of information processing
Hyperarousal: increased phasic firing of NA and DA neurons
Excess NA and DA stimulate additional receptors and cause the signal-to-noise detection to deteriorate
Need to downregulate neuronal activity to return to normal phasic firing
Discuss the anatomical changes in the cortical grey matter which take place during the development of the adolescent brain
- Matures from back to front; increases and then decreases in volume as white matter increases
- Maximum density of cortex is achieved in sensorimotor cortex first, while prefrontal cortex is last (superior temporal gyrus is last to change)
- Massive synaptic proliferation in prefrontal area (early adolescence) followed by plateau phase
- Then, reduction and reorganisation via neural pruning
Discuss the anatomical changes in the subcortical grey matter which take place during the development of the adolescent brain
Basal ganglia (involved in movement, higher order cognitive & emotional functioning) matures after limbic system
Limbic system is involved in emotional regulation, reward processing, appetite and pleasure seeking
Discuss the anatomical changes in the white matter which take place during the development of the adolescent brain
White matter tracts between the prefrontal cortex and subcortical areas develop in a steady non-linear way
White matter increases in all lobes simultaneously
The changes reflect ongoing myelination and increasing axonal diameter
Discuss the hormonal changes which affect adolescent brain development
Girls’ and boys’ grey matter changes in the same sequence, but girls’ grey matter peaks a year before
Corresponds to pubertal maturity, not age
Brain development and puberty are interrelated
Differential sensitivity to testosterone between boys and girls in limbic structures
> Greater risk of anxiety & depression in girls
Discuss the behavioural changes observed during adolescence
- Novelty seeking
Renders adolescents more susceptible to harm (boys especially) - Impulsivity
Impulse control improves as prefrontal cortex and basal ganglia mature - Risk-taking e.g. drug-taking, unprotected sex
Drive to try something new as subcortical structure development is outrunning that of prefrontal cortex - Reward seeking
Nucleus accumbens and amygdala have increased activity in response to rewards; increases at onset of puberty, peaking at 15 - Social behaviour: more sociable, form complex social relationships
More sensitive to peer acceptance & rejection
Skills in empathy, theory of mind, facial processing… develop, corresponding to grey & white matter changes in medial prefrontal cortex & temporoparietal regions
Describe changes in neurotransmitters during adolescence
- Not all neurotransmitters develop at the same pace; serotonin systems develop fully earlier than noradrenaline and dopamine
- Dopamine
Sensitivity peaks during adolescence; dopamine receptors in striatum and prefrontal cortex increase in adolescence & then decline
Neurocircuitry of reward seeking is determined by dopamine signals received by nucleus accumbens and basal ganglia - Oxytocin
Receptors proliferate in limbic areas
Adolescents show heightened response to emotional stimuli
List the different components of a mental state exam
- Appearance and behaviour
- Speech
- Mood and affect
- Thought
- Perception
- Cognition
- Insight and judgement
What are the different features which should observed during the “appearance and behaviour” part of a mental state exam?
- Age
- Body mass
- Personal care and hygiene
- Distinguishing features
- Signs of disease
- Autonomic arousal
- Personal objects
- Psychomotor behaviour
- Abnormal movements e.g. parkinsonism, tremors, dyskinesia
- Level of cooperation and engagement
List the effects of alcohol on different body systems
- GI: acute gastritis, chronic pancreatitis, carcinoma of the oesophagus/large bowel, diarrhoea & malabsorption
- Cardiovascular: hypertension, arrhythmias (atrial fibrillation), cardiomyopathy, CHD
- CNS: Wernicke-Korsakoff syndrome, alcoholic dementia, cerebellar atrophy
- Liver: steatosis, hepatitis, cirrhosis, hepatocellular cancer
- Muscle: acute and chronic myopathy
How do lymphatic capillaries absorb excess fluid from interstitial tissues?
Heart pumps blood along arteries under hydrostatic pressure
Blood pressure causes fluid to move out of the arterial end + move back in the venous end via osmotic pressure
However excess fluid and associated proteins remain in interstitial tissues > absorbed by lymphatic capillaries
Describe the structure of lymphatic capillaries
- Very thin blind-ended tubes
- Lined by a single layer of overlapping endothelial cells
- Minimal basement membrane, no surrounding tissue
- Special junctions: loosely attached so fluid passes between
- Many vesicles of fluid & protein in cytoplasm (uptake of fluid via pinocytosis)
- Fine anchoring filaments outside the cell stop collapse
- 2 valves to prevent backflow of lymph
Describe the histology of the lymph node
- Dense, collagenous outer capsule (extends into the interior via trabeculae)
- Meshwork skeleton of reticular fibres
- Densely packed cortex
> Outer cortex: dominated by B cells in lymphatic nodules when active
> Paracortex: transition zone dominated by T cells - Loosely arranged medulla
> Contains sinuses within which lymph flows > subcapsular sinus > cortical sinus > medullary sinus > efferent vessels - Medullary cords dominated by B cells (plasma cells)
Describe the return of lymph to the venous system
- Lymph of lower body returns to thoracic duct (left venous angle where internal jugular meets left subclavian)
Thoracic duct located in posterior mediastinum (T5)
- Cisterna chyli: most inferior part of thoracic duct
- Lymph of right head, neck & upper limb: right lymphatic duct (union of internal jugular and right subclavian)
Name the primary lymphoid organs and describe their function
- Red bone marrow:
> Production & maturation of B lymphocytes & T cell precursors
> Also granulocytic precursors, megakaryocytes & erythroblasts - Thymus:
> Maturation of T cells - leave thymus as CD4+ “helper” or CD8+ cytotoxic T cells - Proliferation of clones
- Development of immunological self-tolerance
> selection of auto-reactive cells for removal (apoptosis) to prevent autoimmunity; export of repertoire of T cells for life
Which structures are located in the corticomedullary junction?
Post-capillary venules analogues to endothelial venules in lymph nodes in the thymus
What are Hassall’s corpuscles?
Characteristic regions of degenerating epithelial-reticular cells in the thymus
Which hormones are produced in the thymus?
Thymulin
Thymopoietin
Thymosins for T cell maturation
Describe the blood supply to the thymus
Inferior thyroid arteries and internal thoracic artery
Describe the development of the thymus
Develops from the endoderm of the 3rd pharyngeal pouch
Most active in childhood - increases in size until the first year of life
Remains the same size until 60, then decreases
In adulthood, involution occurs: fatty infiltration and lymphocyte depletion
Describe the structure of the lymph node
- Small kidney bean-shaped aggregates of lymphoid tissue with fibrous capsule
- Site of adaptive immune response activation; function is the filtration of lymph
- Lymph node is compartmentalised
> Germinal centre: B lymphocytes
> Paracortical area: T lymphocytes
> Primary lymphoid follicle: B cells
> Medullary cords: macrophages & plasma cells
> Medullary sinus: drains lymph to efferent lymphatic vessels
Which cells form the boundary between the sinuses and the dense lymphoid tissue of the cortex and medullary cords of the lymph node?
Littoral cells
Lis the locations of major lymph nodes
- Cervical
- Axillary
- Lumbar
- Pelvic
- Inguinal
Large germinal centres develop light and dark poles - describe the differences between them
Light pole: towards capsule & source of antigen
> More active
> Greater content of tingible body macrophages
Dark pole: towards medulla
> Less active
Where are immature lymphocytes located in the lymph node?
Mantle zone / outer corona
Describe the location and function of stellate macrophages
Stellate macrophages line sinuses in the lymph node & are present on reticular fibres
> Stellate macrophages phagocytose foreign bodies & present antigens to lymphocytes
Describe the changes in the lymph node when an immune response is initiated
- Humoral/antibody response (B cells)
> Growth of lymphoid nodules/follicles in outer cortex - become secondary follicles
> Enlargement of medullary cords (become populated with plasma cells) - Cell-mediated response (T cells)
> Division and enlargement of discrete lymphocytes in paracortical area
> High endothelial venules with cuboidal epithelium are specialised for the exit of lymphocytes
Which cells make up the filtration mechanism in the spleen?
Stave cells
Describe the blood supply to the spleen
- Splenic artery branches into central arteries surrounded by T cell (periarterial lymphoid sheath)
- Further branching at right angles into penicillar arteries (supply red pulp)
> 3 regions
- Pulp arteriole
- Sheathed arteriole: surrounded by macrophages
Terminal arteriole capillary (deliver blood to splenic cords)
Describe mucosa-associated lymphoid tissues
Lymphoid nodules underlying mucous membranes (GI, respiratory, urinary & reproductive tracts)
Diffusely scattered lymphocytes in the lamina propria - mainly T cells
Specialised overlying M cells (APCs) in microfolds
Peyer’s patches of the bowel predominantly produce IgA antibodies
List the 9 characteristics of cancer cells
- Self-sufficiency
- Insensitivity to growth-inhibitory signals
- Evasion of apoptosis
- Limitless replication potential
- Sustained angiogenesis
- Tissue invasion & metastasis
- Avoiding immune destruction
- Genome instability & mutations
- Deregulating cellular energetics
List the steps involved in the metastatic cascade
- Invasion
- Intravasation
- Transport
- Extravasation
- Colonisation
- Angiogenesis
Compare the properties of epithelial and mesenchymal cells
Epithelial cells:
- Cohesive interactions among cells
- 3 membrane domains: apical, lateral and basal
- Presence of tight junctions
- Polarised distribution of cell components
- Lack of mobility
Mesenchymal cells:
- Loose/no interactions between cells
- No clear apical/basolateral membranes
- No cell-cell junctions
- No apicobasal polarised distribution of organelles
- Motile cells w/ invasive properties
Describe the epithelial-to-mesenchymal transition (EMT)
- Epithelial cells lose typical characteristics & become migratory
- In epithelial cells, actin is arranged cortically
- Cells undergoing EMT rearrange their actin cytoskeleton to form thick strands (stress fibres)
> Actin strands help movement
Describe the function of E-cadherin
- Transmembrane protein which forms part of the adherens junction
- E-cadherin binds w/ itself on adjacent epithelial cells via calcium-dependent homotypic binding
- Mediate cell-cell adhesion via their extracellular domain & connect to actin cytoskeleton by associating with alpha and beta catenin via their cytosolic domain
Discuss the effects of aberrant E-cadherin expression in human tumours
- E-cadherin becomes N-cadherin
> N-cadherin is expressed in mesenchymal cells and promotes invasion and motility - Loss/induction of E-cadherin can result from
> Upregulating mutations in transcriptional repressors e.g. snail
> Silencing of CDH1 promoter by methylation
> Mutations in beta catenin
Describe the role of altered integrin expression in the formation of tumours
- Integrins form part of the cell-substratum junction
- Composed of 2 non-covalently associated transmembrane glycoprotein subunits alpha and beta
> Capable of bidirectional signalling - Found in basal epithelial cells (hemidesmosomes) and focal adhesions of migrating cells
- Connect to actin stress fibres and altered expression promotes cell motility and invasion
> Support oncogenic GFR signalling
Extravasation and survival of CTCs
Colonisation of metastatic sites
Describe the role of hepatocyte growth factor in metastasis
HGF or scatter factor can induce epithelial cells to dissociate and scatter
- HGF is a mitogen (growth factor) and a motogen (motility factor) produced by stromal cells in the tumour microenvironment
- Binds to c-met, a RTK on tumour epithelial cells
> Activation of c-met increases tyrosine phosphorylation of beta-catenin
> Results in disrupted E-cadherin-mediated cell-cell junctions
Increased proteolytic activity is necessary for invasion; which proteases are expressed in tumours?
- Serine proteases: urokinase plasminogen activator (uPA)
> Cleaves plasminogen to plasmin, which activates MMPs & degrades ECM - Matrix metalloproteinases (MMPs)
> Require calcium/zinc to degrade matrix components
> MMP-2 degrades type IV collagen - Cysteine proteases
> Cathepsin K collagenolytic activity results in matrix degradation
Describe different modes of tumour spread
- Lymphatic spread
> Common in carcinomas, e.g. breast cancer travels to axillary nodes > thoracic duct > systemic circulation - Haematogenous spread
> Common in sarcomas - Transcoelomic spread
> Across peritoneal cavity
> Incidence higher with tumours arising from peritoneal cavity e.g. ovarian
Describe intravasation and transport
Intravasation
> Attachment > degrade BM > diapedesis > new blood vessels are leaky facilitating entry
Transport
> Most tumour cells do not survive the shear stress of blood flow, immune detection & anoikis (cells initiate a form of apoptosis when detached from substratum)
Describe the process of extravasation
> Slow rolling by forming attachment with adhesion molecules on endothelial lining
> Tumour cells express high levels of ligand for selectins
> Cells roll until further interactions with cell surface molecules arrest the cell & facilitate diapedesis (integrins, cell adhesion molecules and junction adhesion molecules)
Describe the role of selectins in tumour formation
- Selectins are carbohydrate-binding transmembrane molecules
> P-selectin (platelets & endothelial cells)
E-selectin: endothelial cells, ligand is a carbohydrate on tumour cells: sialyl-Lewisx antigen, associated w/ metastasis and poor prognosis
- L-selectin: leukocytes
Faciliate tumour cell dissemination and formation of tumour microenvironment
Describe the process of angiogenesis
As tumour cells become hypoxic, they produce a transcription factor - hypoxia inducible factor (HIF)
- Transcribes genes which promote angiogenesis such as VEGF
- Endothelial cells proliferate to produce sprouts
Neovasculature is leakier and facilitates extravasation
What is meant by the “angiogenic switch”?
- Initiation of blood vessel formation is induced when angiogenic activators dominate over angiogenic inhibitors
- Angiogenic inhibitors downregulated in cancer
> Thrombospondin: antagonises VEGF
> Angiostatin and endostatin inhibit epithelial cell migration
> Tumstatin inhibits endothelial cell proliferation
List the cells of the tumour microenvironment
- Cancer-associated fibroblasts (CAFs)
- Endothelial cells
- Pericytes (perivascular stromal cells)
- Immune cells
> Mast cells
> Tumour-associated macrophages
> Neutrophils - Mesenchymal stem cells
Describe the function of cancer-associated fibroblasts (CAFs)
Secrete MMPs, cytokines, IL-8 and VEGF
Describe the function of pericytes in the tumour microenvironment
- Low pericyte coverage of vasculature - leaky vessel structure
- Facilitates tumour cell invasion/extravasation
Describe the function of tumour-associated macrophages
Can polarise to
M1 - tumoricidal
M2 - promote tumour growth > Chronic inflammation > Immune suppression > Angiogenesis > Invasion/metastasis: CSF-1/EGF paracrine loop; EGF acts on tumour cells to promote invasive & proliferative capabilities
> Produce growth factors & MMPs to promote angiogenesis, cell invasion & intravasation
Explain the interactions between programmed death 1/programmed death ligand-1 (PD-1/PDL-1)
- T cells are specifically activated by the T cell receptor
- Binds to tumour antigen expressed on MHC on tumour cell
- “On switch” for cytotoxic T cell response
- PD-1 expressed on CD8+ T cell: off switch
- PDL-1 expressed on tumour cell interacts with PD-1 to dampen immune response
Describe the MAPK cascade
Mitogen-associated protein kinase cascade
- Signalling molecule e.g. growth factor binds to receptor tyrosine kinase
- Relay protein Grb/Sos activates ras
- Ras activates Raf kinase
- Raf kinase phosphorylates & activates MEJ
- MEK activates ERK
- ERK goes to nucleus & alters gene transcription to produce response
Describe the pathophysiology and treatment of Chronic Myeloid Leukaemia (CML)
- Philadelphia chromosome: reciprocal translocation between chromosomes 9 and 22
- Forms a lethal recombinant gene: Bcr-Abl (strong kinase)
- Tyrosine kinase is permanently switched on leading to rapid uncontrolled progression through cell cycle in myeloblasts
Treatment: tyrosine kinase inhibitors (TKIs) e.g. imatinib
> Bing to ATP-binding site in tyrosine kinase preventing functioning
> Major improvement in survival but drug resistance can arise
Describe the use of target therapies for the treatment of non-small cell lung cancer
- Non-small cell lung cancer with mutations in anaplastic lymphoma kinase: crizotinib
- Non-small cell lung cancer with EGFR mutations: erlotinib
- Both highly selective
Differentiate between predictive and prognostic markers
Predictive markers
> Predicts which patient will benefit from specific treatment, helping to choose a drug
> Basis of precision medicine e.g. Raf mutations in melanoma
Prognostic markers
> Inform about outcome regardless of treatment
> Helps in choosing which patients to treat, but not how to treat them
> E.g. circulating tumour cells in breast cancer
Oestrogen receptors are prognostic + predictive (high ER expression gives positive prognosis, ER+ve tumours benefit from tamoxifen)
Describe the mechanism of action of PARP inhibitors
E.g. olaparib, treats ovarian, breast & prostate cancer
- Targets PARP, which is involved in single-strand repair
- If single-strand break is not repaired, a double-strand break results
- Double strand breaks repaired via homologous recombination
> BRCA involved in double strand repair
Aberration in BRCA means double strand breaks are not repaired, resulting in cancer cell death
Describe the role of the innate immune response in eliminating nascent cancers
- Innate lymphocytes - NK cells, NK T cells, gamma delta T cells - recognise aberrant cancer cells
- NK cells recognise a change in MHC I expression, bind & release cytotoxic factors to kill cells
- Bring other cells to the area like dendritic cells and macrophages
- Release interferon gamma: initiates cytokine cascade
- Trafficked to local lymph nodes or spleen generating adaptive immune response
Describe the process of immunoediting
Immunoediting produces low antigenicity tumour cells by selection; pressure from immune system + genomic instability enables evasion of immune response
- Elimination: removal of tumour by immune system
- Equilibirum: tumour does not growth further due to constant attack by immune system; latent
- Escape: tumour constantly generates new clones & variants
> Eventually heterogeneous tumour evades immune system
> Grows uncontrollably becoming clinical cancer
Describe the mechanism of action of checkpoint inhibitors (non-cellular therapy)
E.g. ipilimumab
Targets “off switch” on T cells: CTLA-4 (co-stimulatory for T cells), which interacts with B7 to suppress T cell proliferation
> T cells continue to be aggressive
Associated with adverse events like skin mucosa & gut problems, but they can be managed with steroids
Includes PD-1/PDL-1: nivolumab, pembrolizumab
Give examples of non-specific therapies (non-cellular therapy)
- Using toll-like receptors e.g. imiquimod (TLR-8 agonist) mimics concentrated viral assault on skin, producing a localised inflammatory response > kills cancer cells
- IL-2 immunotherapy
> High toxicity & narrow therapeutic window but can be effective
Describe vaccination strategies (non-cellular cancer therapy)
- Require that the tumour-associated antigen is only expressed in that tumour; brings strong immune response specifically towards tumour
- Peptide therapy: single or multiple peptides (wide antigen panel)
Describe monoclonal antibody therapies
- Administration of monoclonal antibodies which target either tumour-specific or over-expressed antigens
Mechanisms:
- Apoptosis induction
- Complement-mediated cytotoxicity (CDC)
- Antibody-dependent cell cytotoxicity (ADCC) - opsonisation
- Conjugated to toxin/isotope
Discuss the role of haematopoietic stem cells in cancer treatment
- Bone marrow donation, mobilise stem cells into peripheral blood using GCSF
- Destroy immune system via total body irradiation/chemotherapy
- Re-infuse HSC into clean system
- Innate IR recovers first (primarily RBCs & platelets)
- Potentially curative
List cellular immunotherapies used in cancer treatment
- Haematopoietic stem cells
- CAR (chimeric antigen receptor) T cells
- NK cells
- Gamma delta T cells
- Tumour-infiltrating T cells
- Dendritic cell vaccines