Cancer immunotherapy

Cards (38)

  • What is the rationale for targeting cancer through the immune system?
    • Traditional chemotherapy is not specific to cancer cells - rapidly dividing healthy cells are often affected too
    • Targeted therapy via. oncogenes/tumour suppressor genes allows lower doses of chemotherapy but tend to build up resistance
    • In comparison, the immune system is trained to ignore self-cells and avoid autoimmunity - better target
  • What are the problems in targeting cancer through the immune system?
    • Cancer cells are not immunogenic enough to elicit recognition by the immune system
    • Cancer cells cleverly secrete factors which switch off the immune system and allow the tumour to grow
    Need to find a way to reactivate immune cells to target cancer cells
  • How does the immune system affect susceptibility to cancer?
    Increases susceptibility to cancer induced by chemicals and viruses

    Deficiency in cytotoxic and NK T cells (which are largely responsible for killing virally infected cells) = increased sizes of tumour volume
  • How could the immune system be programmed to recognise cancer cells?
    Target the tumour antigens on the surface of cancer cells
    • Works similarly to virally infected cells that express viral antigens
  • What are the key principles of the immune system?
    1. Self-tolerance - during T cell development in the thymus, T-cells are screened prior to maturation to eradicate those which are self-reactive (avoid autoimmunity)
    2. Generate an immune response against foreign (non-self) antigens - body must detect harmful foreign antigens and attract immune cells to attack it
  • What is the problem with targeting tumour-specific antigens (neoantigens)?
    There are few tumour-specific antigens which exist
  • What are tumour-specific antigens know as?
    Neoantigens
  • How are neo-antigens produced?
    Produced as abnormal protein (peptide) as a result of cancer cell mutation
    • Non-self peptides - not produced in normal tissues
    • Generate a targeted immune response when presented by MHC complexes on the cell surface
    • Detected by T-cells which eliminate the cancer cell
  • What is an example of a tumour-associated antigen targeted in breast cancer?
    HER2 receptor
    • Overexpressed in breast cancer
    • Targeted by Herceptin (trastuzumab) antibody
    • However, HER2 receptor exists amongst other non-cancer cells in the body which will also be susceptible to Herceptin
  • In what scenario are neoantigens detected?
    • Detected by T cells
    • Neotigens are released from destroyed cancer cells - most proto-oncogene to oncogene transformations tend to occur intracellularly
    • Need enough neoantigens that can be detected by immune cells to activate immune cells - but struggling to find these
  • What is cancer immunoediting?
    Characterised by changes in the immunogenicity of tumors due to the anti-tumor response of the immune system, resulting in the emergence of immune-resistant variants
    • Phase I = elimination - innate and adaptive immune responses to tumour cells
    • Phase II = equilibrium - tumor cells that have escaped the elimination phase and have a non-immunogenic phenotype are selected for growth
    • Phase III = escape - tumor cells continue to grow and expand in an uncontrolled manner and may eventually lead to malignancies
  • Role of NK cells during phase I of cancer immunoediting
    NK cells
    • Healthy cells express inhibitory ligands against NK cells
    • Reduced expression of these ligand or up-regulation of stress-induced ligands in tumour cells leads to production of IFN-y and direct cytotoxic activity
  • Role of Helper T cells (CD4+) during phase I of cancer immunoediting
    Th1
    • Macrophage activation - tumour cells polarise macrophages to make them tumour promoting - try to manipulate polarisation to favour target the tumour
    • Also suppress Th2 responses and promote cell-mediated cellular cytotoxicity
    Th2
    • Antibody production via. B-cells
    • Also suppress Th1 responses and promote mast cell and eosinophil function
  • Role of Cytotoxic T cells during phase I of cancer immunoediting?
    Cytotoxic T cells (CD8+)
    • Induce cell killing
  • What occurs during phase I of cancer immunoediting?

    • Tumour recognised and eliminated by the innate (kill cancer cells) and adaptive (recognise tumour-associated neoantigens) immune response
    • Innate = macrophages, NK cells
    • Adaptive = T-cells and B-cells
    • Phase I is not extensive enough - not enough enough of stress ligand nor immunogenic enough to provoke a strong immune response
  • How are new genetic variants induced during phase II of cancer immunoediting?
    • Tumour cells are constantly mutating - also applies to tumour surface antigen
    • This induces resistance as the immune system cannot keep up
    • The clones which survived elimination and are capable of evading the immune system still remain, learning to be less immunogenic
  • What occurs during phase II of cancer immunoediting?

    • Rare tumour subclones with further mutations survive elimination (phase I)
    • Tumour progresses to equilibrium (phase II) where selection pressures instigate new tumour cell genetic variants
    • Net tumour growth is limited and stalled - tumour enters dormancy (neither shrinking nor growing rapidly)
  • What occurs during phase III of cancer immunoediting?
    Tumours with reduced immunogenicity grow progressively in an immunologically unrestrained manner, establish an immunosuppressive tumour microenvironment, and become clinically detecatable
  • What processes occur during phase III of cancer immunoediting?
    • Poor antigenic expression - tumours fail to produce tumour neoantigens and mutations in MHC genes leads to loss of MHC complex (invisible presentation to immune system)
    • Increased release of immune suppressive factors, e.g. TGFb inhibits T-cell activation
    • Increased number of immunosuppressive cell types that are tumour promoting
    • Increased expression of checkpoint proteins - sustain anti-immunogenic environment around tumour - downregulation of T cells
  • What is the role of regulatory T cells?
    Suppress exaggerated immune response, thereby maintaining homeostasis and self-tolerance. Inhibits the anti-tumour immune response through inhibition of tumour-suppressing immune cells
  • Regulatory T cells in tumours
    Upregulation of regulatory T cells to promote tumour development, i.e. overexpression of IL-10, TGFb, M2 macrophages, myeloid derived suppressor cells
  • What is the role of myeloid-derived suppressor cells (MDSCs)?
    • Heterogeneous population of immature myeloid cells with immunosuppressive properties
    • Produce high levels of IL-10, TGF-b and other cytokines as well as both reactive oxygen and reactive nitrogen species (ROS and RNS), which together inhibit NK cells, and CD8+ and CD4+ effector T cells, and promote the expansion of regulatory T cells
    • Supports environment and suppresses activation of immune cells
  • What is the difference between hot and cold tumours?
    Hot
    • Infiltrate the tumour microenvironment (high Teff to Treg ratio)
    • Immune cells tend to be non-functional (exhausted phenotype)
    • Aim to reactivate those immune cells since they are already inside the tumour
    Cold
    • Unable to infiltrate the tumour microenvironment
    • Immune cells cannot simply be reactivated since they are not inside the tumour
  • What is the race to make "cold" tumours "hot"?
    • Cold tumours struggle to be detected by immune cells as they cannot penetrate inside the tumour microenvironment and detect tumour antigens
    • In contrast, hot tumours that have high mutational load and often have higher levels of neoantigens that can be recognised by the immune system
    • Making cold tumours hot will allow for more effective treatment
  • What are examples of hot and cold tumours?
    Hot = melanoma and lung cancer
    Cold = prostate and pancreatic cancer
  • Why is tumour profiling necessary?
    Response is too low just using checkpoint inhibitors

    Characterising and profiling tumours provide information on gene mutations, proto-oncogene to oncogene transformation, loss-of-function tumour suppressor genes, angiogenic factors, likelihood to metastasis etc.
  • What are further categoies of hot and cold tumours?
    Immune-inflamed
    • High degree of cytotoxic T-cell infiltration
    • Mechanism of immune evasion: checkpoint activation, T-cell exhausation
    Immune-excluded (immunogenic)
    • Presence of T cells at invasive margin, absent in central tumour mass
    • Mechanism ": stromal barriers, aberrant vasculature, hypoxia, lack of chemokines, oncogenic pathways
    Immune-desert (resistant to immunotherapy)
    • Absence of T cells within tumour and at margins
    • Mechanism ": insufficient priming, defects in antigen presentation, lack of antigen
  • Which further category of hot and cold tumours are prime candidates for checkpoint inhibitor therapy?
    • Immune-inflamed - prime candidate due to high cytotoxic T-cell infiltration
    • Immune-excluded - not a great candidate because of T-cell exclusion from centre mass, but also poor blood supply to this tumour
    • Immune-desert - not a candidate as there are no immunes at all
  • What are the main types of immunotherapy?
    • Monoclonal antibodies
    • Immune checkpoint inhibitors (ICIs)
    • Chimeric antigen receptors (CARs)
    • Immunotherapy combinations
  • What was the problem with initial immunotherapies?
    Stimulated the immune system non-specifically
    • Expanded T cell population and drove T cell activation
    • Made immune system more sensitive to kill tumour cells generally
    • Short-term responses
    • Not targeted so had risks of autoimmunity
  • What is antibody-based immunotherapy?
    • Attaches an antibody to an antigen on tumour cell (must locate first)
    • Can conjugate many things to antibodies, e.g. radionucleotides, toxins
    • Recruits immune cells, e.g. NK T cells recognise stalk (Fc) region of antibody and secrete perforins and granzymes to digest tumour cell
    • e.g. Herceptin antibody blocks GF ligands from binding to HER2 receptor dimer and stop activation
  • What is monoclonal antibody-based immunotherapy?
    • Developed against checkpoint proteins, e.g. PD1, PDL1, CTLA4
    • Blocks the downregulation of T cells - allowing T cells to target tumour cells
    • Can use bispecific antibodies - link to both tumour neoantigen and T cell receptor to bring T cells into TME
    • Often bind extracellularly as they are large molecules not readily taken up by cells - manipulate them to bind
    • If the disease is ligand dependent, antibody-based therapy will not work
  • What are examples of monoclonal antibody-based immunotherapy?
    Anti- suggests function as inhibitors of specific molecular targets
    • Herceptin (trastuzumab) - anti-HER2 receptor (breast cancer)
    • Rituximab - anti-CD20 on B-cell surface (non-Hodgkin's lymphoma, chronic lymphocytic leukaemia etc.)
    • Cetuximab - targets anti-EGFR (metastatic colorectal cancer, non-small cell lung cancer etc.)
    • Pembrolizumab - anti-PD-1 (melanoma etc.)
  • What are examples of immune checkpoint inhibitor-based immunotherapy?
    1st generation antibody - ipilimumab
    • Anti-CTLA4 antibody
    • Used specifically for melanoma
    • Over time, gradually had elimination/regression of tumours due to reactivating T cells
    • Eventually, resistance builds up and the tumours come back - then switch to a different agent
    2nd generation antibodies - nivolumab and prembrolizumab
    • Anti-PD-1 and anti-PDL-1 antibodies
  • What is the cascade for chimeric antigen receptor-based immunotherapy?
    • CAR-T cells recognise and bind with ScFv region tumour cells
    • Binding causes intercellular signalling in CAR-T cells
    • Activation of CAR-T cells and rapid proliferation
    • CAR-T cells attack and kill cancer cells
  • What are examples of chimeric antigen receptor-based immunotherapy?
    Kymriah (tisagenlecleucel)
    • Treatment of acute lymphoblastic leukaemia (ALL)
    Yescarta (axicabtagene ciloleucel) 
    • Treatment of diffuse large B-cell lymphoma (DLBCL)
    Both of these treat blood cancer - single circulating leukaemic cells are easier to target than solid tumour mass cells.
  • What is chimeric antigen receptor-based immunotherapy?
    • Rationale is to engineer T cell receptors to recognise tumour antigens with high affinity, rather than relying on the immune system to to recognise the limited/slightly immunogenic neoantigens - made to attach more strongly
    • Main structural features - ScFV of antibody (provide specificity), hinge and transmembrane region, co-stimulatory domain, T-cell activation domain
    • Potential to manipulate material and sequences for enhanced receptor efficacy, e.g. gene editing via. TALEN, CRISPR
  • What is the difference between tumour-associated antigens and tumour-specific antigens (neoantigens)?
    Tumor-associated antigens may be found at low levels elsewhere in the body but neoantigens are completely unique to the cancer cells

    Targeting neoantigens for immunotherapy comes with a lower risk of accidentally harming healthy cells along the way