Exam 3 Flashcards

(186 cards)

1
Q

Characteristics of cancer

A

Increase in Treg population:
loss of cancer immuno-surveillance
promotes suppression of anti-tumor response
promotes cancer progression

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

Characteristics of Primary Immunodeficiency (PID)

A

Inherited / genetic
Diagnosis in children but also in adults
Infections: more severe, frequent or abnormal pathogens
Auto-immunity, granulomas, malignancies
Most frequent: IgA deficiency and MBL deficiency: mostly asymptomatic: no PID
Most severe: SCID

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

Phenotype SCID patient

A

Severe Combined Immuno Deficiency
Clinical presentation at young age ( 3 months)
Failure to thrive
Opportunistic infections: Candida albicans, Pneumocystis jiroveci
Poor thymic development
T-cell number strongly decreased
IgG decreased
Treatment: SCT (or gene therapy)

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

Indicators of SCID

A

recurrent bacterial infections: >4 per year
* recurrent severe infecties >1: meningitis, osteomyelitis, pneumonia, sepsis
* opportunistic infections: Pneumocystis jiroveci, Candida
* Abces of internal organ of repeatedy subcutane abcesses
* extensive warts and molluscum
* Failure to thrive
* chronic diarrhea
* Family history of early deaths
* consanguinity

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

How can different infections signal the type of immunodeficiency issue one has?

A

Opportunistic infections: T-cell / combined problem
Viral infections: T-cell / NK cell problem
Intracellular bacterial infections: T-cell problem
Extracellular bacterial infections: B cell problem
Fungal infections : granulocyte problem
Encapsulated bacterial infections: complement problem

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

How to diagnose a patient of potential immunodeficiency

A

Ask about:
Infection history, Autoimmunity, Hospitalization, Malignancies, Family

Laboratory diagnostics:
Leucocyte numbers
* Granulocytes (neutrophils, eosinophils en basophils)
* Lymfocytes
* Monocytes
Immunoglobulines
* IgG, IgA, IgM, (IgE)
Auto-antibodies
T, B en NK-cells (absolute numbers)
* T- and B cells subpopulations: naive memory
* C3, C4, C1q, complement routes: classical, alternative
Humoral (B cell function)
* Vaccinate and measure specific antibodies before and after
* IgG titer “rijksvaccinatie” vaccination (BMR, Hib)
* In vitro function

Granulocyte function analysis
Complement deficiency analysis
DNA mutation analysis

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

Primary immunodeficiencies: Inheritance

A

Autosomal recessive:
Person w/ two copies of mutated gene (1 from each parent, homozygous) = disease
Person only w/ 1 copy of the mutation (heterozygous) = a carrier and typically do not show symptoms + healthy

Autosomal Dominant:
Only one copy of the mutated gene is enough to = disease

When sex linked:
X-linked immuundeficiencies: responsible gene on X-chromosome
* recessive
XXmut= women: (mostly) healthy but carrier
XmutY= men disease

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

Most common form of SCID: Genetic basis, phenotype

A

Common gamma chain (CD132) deficiency: X-linked
> Most common form: 55%

Function of CD132:
Component of receptors for L-2, IL-4, IL-7, IL-9, IL-15, and IL-21

Immunological Phenotype (T-B+NK- SCID)
- T cells and NK cells absent, B cells present but non-functional

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

Other forms of SCID: autosomal recessive

+ newborn screening how?

A

Jak-3 mutation: T-B+NK-
IL7Ralpha mutation: T-B+NK+
ADA deficiency: T-B-NK-
RAG-1/RAG-2/Artemis: T-B-NK+
> V(D)J recombination TCR and BCR rearrangement
CD3 mutation: T-B+NK+

Screening of T cell production via TREC analysis

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

How to confirm genetic condition SCID

A

mutation DNA analysis:
- sequence specific gene
- sequence panel genes (PID panel)
- whole exome/genome sequencing

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

Treatment Primary Immuno Deficiencies

A
  • Antibiotic profylaxis
  • Immunoglobuline substitution
  • Immunosupressive therapy
  • Stamcell transplantation (high risk)
  • (Genetherapy: trials)
  • Gene Targeted therapy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Why study Inborn Errors of Immunity?

A

Increase understanding
→improve patient diagnosis + care
→learn more about normal function of
components of immune system
→experiments of nature

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

What is XLA

A

X-linked agammaglobulinemia (XLA):
a PID caused by a mutation in the Bruton’s tyrosine kinase (BTK) gene

BTK gene is involved in:
- Differentiation of precursor B-cells in bone marrow
- Survival + maturation of B-cells

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

Effect of having XLA

A

▪ Blockage of B cell dev in Bone marrow
▪ Decreased B cell #no. in blood
▪ All Ig in serum decreased: IgG, IgA, IgM
▪ No response to vaccinations
▪ Increased risk infections with extracellular bacteria: (Streptococcus: otitis media, pneumonia)
▪ Chronic viral infections: polio-, echovirus

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

XLA treatment

A

Intravenous Immunoglobulin (IVIG)
(i.v. gamma-globulin)
* 400 mg/kg every 3 weeks
* target level IgG ≥ 7 g/l

Antibiotic prophylaxis

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

Most PIDs are predominantly antibody disorders. What are the majority of them + its characteristics

A

Common Variable Immuno Deficiency (CVID)
- B cell dysfunction
- Onset age > 4 yr (usually young adult)
- Low IgG and IgA, normal or low IgM
- Absent vaccination response or low switched memory B cells
- Treatment Immunoglobulin substitution (IVIG) (reduces rate of acute + chronic infections but fails to control secondary autoimmune + inflammatory complications)
- diagnostic delay

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

Importance of NFkB pathway

A

key player in B cell differentiation and function

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

Characteristics of NFkB pathway

A

NFKB family of transcription factors comprise of 5 proteins:
- c-Rel, Rel-A (p65), Rel-B, NFKB1 (p50) and NFKB2 (p52)
- Form heterodimers and homodimers
- Different dimers have distinct regulatory/transcriptional properties
- In resting cell NF-KB in cytosol, retained in latent state by IkB proteins
- Upon cell activation, phosphorylation and degradation of IkB
- NF-KB enters nucleus and regulate gene expression

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

NFKB1 loss-of-function is characterized by….

A

recurrent infections, increased autoimmunity and cancer risk

B cell numbers are low in most, but not all, clinically affected NFKB1+/- cases
Serum IgG levels seem to decrease progressively with age in NFκB1+/- cases

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

heterozygous CTLA-4 mutation (occurs in vv small % of those with CVID)

A

results in truncated non-functional proteins
linked to autoimmune disorders
Autosomal dominant mutations
But patient can have mutation with parents who are healthy!!

CTLA-4 is a negative regulator of T cell function
Sufficient CTLA-4: T cells under control,
no signal via CD28
Not enough CTLA-4: Overactivated T cells, too much signal via CD28

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Role of Tregs in regulation by CTLA-4

A

CTLA-4 is expressed on Tregs
> modulates the co-stimulatory signals required for T cell activation

CD80/CD86 are expressed on APCs + bind to…
CD28 (on T cells): Stimulates T cell activation
CTLA-4 (on Tregs and activated T cells): Inhibits T cell activation

Tregs expressing CTLA-4 internalize CD80/CD86 = turns off cell activation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Treatment plan for HIGM

A

SCT with MUD donor:
Stem Cell Transplant with Matched Unrelated Donor
IVIG+ AntiBody profylaxis
Quick diagnosis important for succesful SCT

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Abatacept treatment for CTLA-4 mutation

A

Abatacept is a human recombinant CTLA-4/IgG1 fusion protein
Works by mimicking the natural function of CTLA-4

Consists of the extracellular domain of CTLA-4 fused to a part of IgG1, allowing it to bind CD80/CD86 on APCs

blocks CD80/CD86 from interacting w/ CD28 on T cells = prevents the co-stimulatory signal required for T cell activation
> reduces T cell activation = dampens immune response

Recovery of T cell counts over time

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Hyper IgM syndrome (HIGM)

A

Characteristic: a class switch recombination (CSR) defect caused by a homozygous CD40 mutation

B cells fail to switch antibody production from IgM to other isotypes (IgG, IgA, IgE) due to defective class switch recombination (CSR)

Result: Elevated IgM levels with low IgG and IgA, leading to impaired immunity against infections

CD40, expressed on B cells, is required for interaction with CD40L on T cells.
This interaction is essential for CSR, B cell activation, and germinal center formation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
What can lead to phagocytic disorders in PID
Defects in any steps in Destroying microbes by neutrophilic granulocytes: Adhesion, chemotaxis, Phagocytosis (opsonisation: coating with Ig and/or complement), killing: oxygen radicals can lead to Chronic Granulomatous Disease (CGD): A defect in the NADPH oxidase subunit prevents ROS production = neutrophils unable to kill ingested pathogens, resulting in recurrent infections. > Frustrated immune cells form granulomas
21
Process of Destroying microbes by neutrophilic granulocytes
Neutrophils engulf and kill the microbes to which they bind > Bacteria are taken up into phagosomes > Phagosomes fuse with granules, assembly of a functional NADPH oxidase in the phagolysosome membrane, leading to generation of O2- Acidification as a result of ion influx releases granule proteases from granule matrix
22
Treatment for CGD
Antibiotic profylaxis, Interferon gamma, Stemcell transplantation, (Gene therapy: Not in NL)
23
Kidney Functions
. Removal of waste products * Mineral housekeeping * Regulation body fluids * Regulation acid & base balance * Activation of vitamin D and production of strong bones * Production of EPO
23
The Global Burden of Kidney Disease
Millions more develop kidney failure and require KRT but lack access to therapy and die prematurely (mainly people from LICs or LMICs)
24
Why is a kidney transplantation performed?
* To feel more fit * To live longer (better survival) * To gain more freedom * To improve chances to work * To live a “normal life”, to have a family * Better QoL *Cheaper medical lifestyle
25
Relative risk of death after kidney transplantation
risk of death decreases as more days pass after transplantation
26
How to check if kidney donor is a good match?
Via HLA matching
27
What are human leukocyte antigens (HLA)?
They are proteins found on the surface of most cells in the body. They are encoded by genes in the HLA complex, located on chromosome 6 Help the immune system distinguish between the body's own cells and foreign substances Essentially human MHC Important there is HLA matching when donating so immune cells dont start to attack self cells
28
HLA matching
HLA Class I (A, B) and HLA Class II (DR) A person inherits one set of HLA genes from each parent, giving two alleles per gene (e.g., HLA-A, HLA-B, HLA-DR). Siblings have a 1 in 4 chance of being an exact match since they can inherit the same combination. Best fit: HLA mismatch 0-0-0 Worst fit: HLA mismatch 2-2-2 * Better survival * Less immunization * Not necessary
29
MHC characteristics
Highly polymorfic (there is an extensive variety of alleles (genetic variants)) Present peptides derived from pathogens or “altered self” to the immune system Differences in HLA elicit an immune response, both cellular and humoral Big barrier in transplantation medicine
29
What is immunization and sensitization
Immunization - recipients immune system becomes sensitized to foreign HLA markers (can be due to exposure from foreign HLAs in previous transplants, blood transfusions, or pregnancies) Sensitization - immune system has developed antibodies against certain HLA types = more difficult to find compatible donor kidney = hyper acute rejection
29
What are the different types of reactivity of foreign HLA + how they work
Direct (recipient's T cells directly interact w/ donor's HLA molecules on the surface of the transplanted organ) Semidirect (donor cells transfer their HLA molecules directly to recipients APCs which present donors HLA molecules on their surface + recipients T cells recognize foreign HLA molecules) Indirect (recipient’s APCs take up donor tissue, process donor’s HLA molecules + present them as peptides on their own MHC molecules. Recipient’s T cells recognize these as "foreign," activating an immune response) Interaction of APC + T cell is regulated by 'uppers' and 'downers'
30
Brother wants to donate: what is the best clinical outcome of the screening?
Homozygous twin brother
31
Hyperacute rejection (signs, cause, prevention)
Signs: Kidney color not pink, Flabby tissue, No urine production Cause: preformed HLA (or anti-blood type) antibodies Prevention: a crossmatch test is performed to prevent hyperacute rejection
32
How is a crossmatch test carried out for testing potential transplantation efficacy?
CDC-based assay Patient serum (contains their antibodies) > Donor lymphocytes (T cells usually) are isolated (express donor HLA molecules) > mix serum + donor cells > addition of complement > if antibodies against donor HLA = antibodies bind to lymphocytes > complement system activated = lysis of donor lymphocytes
33
Different techniques to detect HLA antibodies (from most sensitive to least sensitive)
Cell-based assays (e.g CDC cross-match), Solid-phase assays (e.g ELISA), Flowcytometry cross-match, Luminex
34
How to detect prior sensitization
1. Complement dependent cytotoxity detects preformed antibodies 2. Flow cytometer tests antibodies attached to lymphocytes 3. Solid-phase assays: * Antigens bound to microtiter plates * Single antigen beads (Luminex)
35
Difference between Acute antibody mediated/humoral rejection and hyperacute rejection
Hyperacute rejection is an immediate and irreversible response to pre-existing antibodies. Acute antibody-mediated rejection happens later and is a result of new antibody production after the transplant, which can often be managed if caught early.
35
Differences between cell based and solid phase assays
Cell based assays * Positive assay highly clinical relevant (80% rejection of the graft) * Often IgG enhanced * Dithiothreitol (DTT) to remove IgM * Flowcytometer: both complement as noncomplementbinding depending on the second antibody Solid-phase assay * Sensitive * Both complement and noncomplement binding antibodies, no IgM autoantibodies or nonHLA antibodies * Pathogenic treshhold remains unknown
36
Being sensitized results in increased risk of …
- Hyperacute rejection - Memory B cell response leading to early ABMR - Chronic active ABMR
36
Acute Antibody-Mediated/Humoral Rejection
Occurs within days or weeks after transplantation Rapid graft dysfunction Main target of these antibodies is the MHC antigens on the peritubular endothelium and glomerular capillaries Sometimes can be triggered by non-HLA antibodies which target other molecules or proteins on the transplanted kidney Caused by an anamnestic response ( recipient’s immune system being "primed") by previous exposure to similar HLA antigens from prior transplants, blood transfusions, pregnancy etc = generates complement-fixing antibodies leading to rejection
36
Explain how prognosis rejection is time dependent
the earlier the kidney transplant is performed after the onset of kidney failure the less harm + better the chances for long-term success
37
Blood types + the antibodies they have in plasma + antigens in red blood cell
A: anti-B, A antigen B: anti-A, B antigen AB: none, A and B antigens O: anti-A + anti-B, no antigens
38
Which blood type can donate blood to who?
A can donate to A or AB B can donate to B or AB AB can donate to AB (because others will attack, e.g if donated to A person the B antigens on AB will = immune system attack) O can donate to A, B, AB, O (because it has no antigens = doesnt trigger anything)
39
What is a type II reaction
Type II Hypersensitivity is an antibody-dependent process in which specific antibodies bind to antigens = tissue damage or destruction E.g A transfusion is a type II reaction Hyperacute rejection is a type II reaction
40
Blood type antigens - where are they expressed
Blood type antigens resemble carbohydrates expressed on the cell wall of bacteria Blood type carbohydrate antigens are also expressed on vascular endothelial cells
41
Blood transfusion characteristics
Pretreatment of blood (washing, leukodepletion, irradiation) aim to reduce the risk of immune reactions but these techniques DO NOT prevent HLA sensitization (the decelopment of immune responses against HLA) if anything they could increase the risk of sensitization They can lead to dev of antibodies against a broader range of HLA's = increasing likelihood of transplant rejection Highest risk for (further) sensitization in pre-sensitisized patients (pregnancies, transfusions, prior transplants)
42
What types of transplantation are there and which is better?
Postmortal vs living Transplantation over blood type barrier (i.e incompatible blood types) can yield better outcomes than postmortal donors
43
Timing of transplantation after diagnosis is important
those who got diagnosed + then received dialysis treatment (their survival rates significantly decreased over time) = sooner you get transplanted the better (with either living donor or dead donor your chances of survival are still better)
44
Conditions to be able to donate a kidney
Organ donation act (1998): - competent - > 18 years of age - clear informed consent - to the benefit of someone else Voluntarily, not paid and always revocable Socially feasible Good chance of success of donation and transplantation
45
Development of immunosuppressive agents for transplantation
First one in 1908 Using bovine serum to impair immune system ability to produce antibodies 9 of 25 renal transplants survived more then 2 years but….Toxic
46
Immunosuppression specific treatments
Steroids inhibit… * IL-1/2 * TNF * Chemotaxis and adhesion of neutrophils Calcineurine inhibitors (tacrolimus or ciclosporin) * Inhibit IL-2 * Stimulate production of TGF-beta Proliferation inhibitors (mycophenolate mofetyl or azathioprine) Rejection treatments * MPNS pulses * ATG/plasmapheresis
47
Transplant rejection types
Hyperacute rejection * Very rare, due to cross matching * <24hr * HLA antibodies already present Chronic rejection * Months to years after transplantation * Multifactorial * Difficult to treat Acute rejection * 20% * Mostly <6-12 months * Easy to treat
48
Immunosuppresion aims + stages
Aim: acceptance of transplant 1. Induction therapy: intense initial immunosuppression 2. Maintenance therapy: long-term immunosuppression 3. Anti-rejection treatment: treats episodes of acute rejection
49
Limitations of immunosuppression
- Required as long as transplant is in - Tolerance unpredictable (rejection remains a risk) - Inter-individual variability (diff doses etc) - Secondary malignancies (increases risk of certain cancers) - Opportunistic infections - Cardiovascular events - Specific side-effects - ADHERENCE/COMPLIANCE (missing doses can lead to rejection, overuse can increase toxicity)
50
Prednisone side effects
* Increase in weight * Type II Diabetes * Hypertension * Psychological changes * Changes in appearance * Thinining of skin * Osteoporosis Immunosuppressant drug
51
Calcineurin inhibitors side effects
Tacrolimus (Prograft®/Advagraf®/Envarsus®) * Hypertension * Diabetes mellitus * Tremor * Hairloss * Hyperlipidemia Ciclosporin (Neoral®) * Hypertension * Excessive hairiness * Gingivahyperplasia * Gout * Hyperlipidemia * Both: Nefrotoxic! * Short term: vasoconstriction * Long term: Structural damage
52
Proliferation inhibitors side effects
AZA and MMF (drugs) * Bone marrow disease: anemia, leucopenia, pancytopenia (AZA and MMF) * Skin malignancies (in particular AZA) * Gastro-intestinal complaints (MMF) * (Viral) infections (MMF)
53
ABO-incompatible (ABOi) transplantation
1) The recipient's anti-A or anti-B antibody levels (titer) must be reduced to a safe level (< 1:256). This prevents antibody-mediated rejection of the transplanted organ. Plasma exchange process: Recipients blood separated into plasma + blood cells Plasma treated to remove: A/B-specific antibodies and non-specific antibodies (Ig) Treated plasma is returned to patient e.g Friend w/ blood type A can donate to recipient w/ blood type O
54
Is transplantation across bloodtype possible?
Yes, bloodtype matching is preferable though
55
Solution to donor shortage
More donation during life, more postmortal donors, xeno-transplantation, organ donation after euthanasia
56
Barriers for xenotransplantation
* Immunological * Infectious * Fysiological * Ethical
57
What are the immunological barriers of xenotransplantation
1) Hyperacute rejection Cause: Preformed antibodies in the human target sugar molecule (Alpha-Gal) on pig cells Binding triggers complement activation + MAC formation = xenograft failure 2) Acute Vascular Rejection Cause: Activation of... Human anti-pig IgG antibodies. NK cells (complement activation, MAC formation) Macrophages (= platelet aggregation, clot formation) = xenograft failure. 3) Coagulation Activation Cause: Mismatch between pig endothelial cells and human clotting mechanisms = abnormal coagulation. Platelet activation and aggregation. Clot formation, leading to dysfunction of the transplanted organ. Solutions to 1, 2, + 3: Genetically modified pigs expressing human complement regulatory proteins 4) Acute Cellular Rejection Cause: Activation of B cells and T cells against the pig xenograft T cells (via CD40) and B cells (via CD20) coordinate immune attacks on the xenograft. Complement activation by T and B cells further damages the organ. (MAC) Solution: Use of immunosuppressive drugs: Rituximab: Targets CD20 on B cells to deplete them. Anti-CD40 therapy: Blocks T-cell activation pathways. Eculizumab: Inhibits C5 in the complement cascade, preventing MAC formation.
58
Infectious barriers for Xenotransplantation
Zoonosis (transmission of infections from animals to humans through transplanted organs) Specifically retroviruses (PERV-A, B, and C)
59
Physiological barriers for xenotransplantation
Physiological differences in: Blood pressure Body temperature Renin-activity Cholesterol Solutions?: Could knock-in human genes and knock-out pig genes Thymokidney (specifically engineered/modified organ that combines thymic tissue (from donor pig) w/ a kidney to promote immune tolerance in recipient as it learns to recognize donors tissues as self)
60
What type of virus is HIV-1
Lentivirus
61
Course of HIV-1 infection
HIV infection > primary infection > a-symptomatic phase > symptomatic phase (AIDS) Gradual depletion of CD4 T cells
62
Characteristics of AIDS
CD4 <200 cells/ul Opportunistic infections e.g Candida, Cytomegalovirus, PCPneumonia Cancers e.g Lymphoma, Kaposi Sarcoma AIDS dementia
63
Factors that influence the clinical course of HIV-1 infection
Course speed (rapid, typical, or slow) determined by CD4+ T cell levels and Viral load HIV-1 diversity HIV-1 specific immunity Development of Cytotoxic T Lymphocyte escape variants
64
Explain how HIV-1 diversity can affect the clinical course of HIV-1 infection
Caused by HIV reverse transcriptase which lacks proofreading acitvity = high mutation rate = millions of viral variants within any infected person in a single day - Positive selection: viral variants with increased fitness escape Cytotoxic T cells, neutralizing antibodies, anti viral drugs, etc
65
Explain how HIV-1 specific immunity can influence the clinical course of HIV-1 infection
Adaptive immune response: Cytotoxic T cells (CD8+) kill HIV-1 infected cells by binding of TCR with MHC of infected APC (Presentation of viral peptides in context of HLA class I) Each HLA-allele presents distinct peptides)
66
Explain how the development of CTL escape variants can influence the clinical course of HIV-1 infection
HIV-1 has a high mutation rate giving high viral diversity Initially only 1 variant may have a mutation that allows HIV infected cells to escape from CTLs Selection pressure is placed by CTLs All cells containing a virus with the intact epitope are eliminated (easier to recognize + kill etc) Only cells infected with escape variant survive (+ thus multiply)
67
Why are these escape variants of HIV-1 not recognized and killed by CTLs
They prevent presentation by either... Mutations in anchor residues (anchor residues are parts of the peptide that secure its fit into the MHC molecule) = peptide can no longer bind to MHC molecule or Mutations in the peptide (even if the peptide is successfully presented by the MHC molecule, CTLs must "recognize" it via TCRs, mutations can change the peptides shape or the way it interacts with TCRs) = TCR doesnt recognize the peptide
67
What are SNPs
Single nucleotide polymorphism = genetic variation in human Variation is called a SNP when present in ≥1% of the population Constitute ~ 90% of variation in human genome they can occur in coding and non-coding regions of the human genome
68
What is the link between SNP and HIV-1 infection?
genetic differences (SNPs) between individuals correlate with their ability to control HIV HIV controllers: people who maintain low viral loads naturally = slow progression Progressors: people whose HIV progresses more quickly (lose CD4+ cells + show high viral loads) They have different SNP profiles, may explain why some people control HIV infection better than others
69
HLA Class I Polymorphisms and HIV-1 Outcomes
Genetic variation in HLA-B peptide binding pocket HLA-B57 and HLA-B27 are associated with better control of HIV (slow disease progression) HLA-B35 is associated with rapid disease progression. Genetic differences, particularly in the HLA region influence progression of HIV
69
HIV-1 immune control by HLA-B57
CTL response is raised against viral peptides presented by HLA-B57: * Presentation of viral epitopes from conserved regions of the virus * High affinity recognition of HLA-B57 presented peptides by TCR = relatively low amounts of antigen result in strong CTL response * Viral escape occurs in immune dominant epitopes = virus mutates these epitopes to avoid immune detection BUT these mutations reduce virus's ability to replicate/cause disease (i.e viral attenuation) essentially Strong cytotoxic T cell response (CD8+ T cells) recognizing conserved epitopes in viral proteins Escape from cytotoxic T cells response occurs and is associated with viral attenuation
70
Humoral immunity in HIV-1
HIV-1 neutalizing antibodies = prevent virus from entering + infecting healthy cells by either blocking the virus from binding to the host or by interfering with its fusion to the cell membrane gp41 and gp120 are important glycoproteins on HIV-1 surface + are involved in binding to CD4 receptors on T cells 120: recognition + binding to CD4 receptor 41: facilitates fusion of HIV-1 envelope w/ host cell membrane
71
HIV-1 NAb response in HIV-1 infection
Native HIV is relatively immunogenic Majority of people are capable of eliciting neutralizing antibodies against assumed conserved and vulnerable epitope HIV is highly mutable and can escape the effects of autologous (from the same individual) NAb response = virus continues to replicate even in presence of the body's neutralizing antibodies Neutralizing Antibody response is not associated with disease progression
72
Overview of HIV-1 specific immunity
Cellular immune response: CTL + viral escape > viral evolution > sometimes viral attenuation Humoral immune response: Neutralizing antibodies + viral escape = NAb are ineffective > viral evolution
73
HIV therapy: today
Antiretroviral therapy (does not eradicate HIV-1, suppress HIV-1 replication = undetectable viral load in blood, as soon as treatment is interrupted = viral rebound (no immune control), HIV persists in viral reservoir during ART) Entry inhibitors e.g CCR5/CXCR4 Inhibitors = prevent binding Fusion Inhibitors Reverse transcriptase inhibitors Integrase inhibitors = prevent integration of viral DNA into host DNA Protease inhibitors = prevents release + maturation of new virus infected cells
74
Prevention of HIV-1 drug resistance
Prevention of resistance development: * Combination of drugs from different classes (since 1996) * High treatment adherence
75
Explanation for the decline in infectious diseases in High Income Countries (HIC) between 1950 and 2000
Decline due to Improvements in hygiene, sanitation and health - Vaccines - Antibiotics
76
Explanation for Increasing incidence of inflammatory diseases in HICs 1950 & 2000
The observed rise in inflammatory diseases may be attributed to - improved living standards, - increased wealth - reduced exposure to infections (Hygiene hypothesis)
77
Effect of number of children in house hold on inflammatory disorders
Larger family sizes = protective against the development of allergic diseases such as hay fever and eczema Having older siblings = more unhygienic contact in early childhood which seemed to protect against the development of allergic diseases (so e.g 5th born has lower prevalence of hay fever than 1st born)
78
Hygiene hypothesis principles
Early childhood exposure to microorganisms may protect against allergic diseases by contributing to the development of the immune system
79
"Old friends" hypothesis
Microorganisms that were once abundant trained our immune system: They were tolerated at the expense of excessive tissue damage to eradicate them. Our immune system has evolved in their continuous presence.
80
Which changes in lifestyle lead to changes in microbial exposure
Rural > urban Outdoor > Indoor (isolation, central heating) Physical labor > sitting professions High fibre food > fast food
81
Expand on the huge differences in the Immune System associated with urban - rural areas
Vaccines + drugs can act differently Urban environments are associated with accelerated inflammatory diseases Rural environments are associated with protection against inflammatory diseases
82
Do microorganisms protect against inflammatory diseases? (e.g Rural farms)
Children growing up in microbe-rich rural traditional farming environments = less likely to suffer from asthma & allergies compared to their urban counterparts eg exposure to farm stables + farm milk Diversity of microbial exposure (e.g due to living on a farm) was inversely associated with risk of asthma
82
Why do the Amish have lower prevalence % of asthma compared to Hutterites?
Although they are genetically close they have different farm layouts Amish: small single-family farms, traditional farming, animal barns close to home = higher diverse microbial exposure Hutterites: large communal farms, modern farming, animal barns very far from homes = lower diverse microbial exposure Amish had a higher bacterial compoition in dust compared to Hutterites
83
How can microorganism exposure during pregnancy influence protection against inflammatory diseases in utero
Maternal farm exposures during pregnancy = increase in cord blood regulatory T cells that may protect against allergies
84
Why are we seeing a sharp increase in asthma symptoms in LICs + MICs
Within urban areas differences in allergy outcomes based on socioeconomic status Urban areas w/ high SES = high prevalence of asthma + allergic sensitization Urban areas w/ low SES = low prevalence of asthma + allergic sensitization
85
Effect of parasitic worm infection on allergic sensitization
parasitic infection = protective effect against allergic sensitization (e.g to house dust mite allergy or asthma)
86
Types of helminths (parasitic worms)
Flatworms (platyhelminths) > Trematodes (flukes) > Cestodes (tapeworms) Roundworms (nematodes) Helminth infections can persist for years in humans
87
Hookworm infection / development
Parasitic roundworm Eggs in feces of infected individual (diagnostic stage) > larva hatches > development into larger larva > larva penetrates skin (infective stage) > larva exit circulation in lungs (coughed up > swallowed) > enter small intestine > excreted
88
Ascaris lumbricoides infection / development
Parasitic roundworm In adult small intestine + excreted eggs in feces > ingestion of embryonated eggs > hatched larva enter circulation + migrate to lungs > coughed up + swallowed > re-enter GI tract > mature in small intestine
89
Schistosoma
Parasitic trematode (blood fluke) Eggs from feces or urine of infected human > eggs hatch + release larva > larva penetrate snail tissue > larva develop in snails > developed larva relesed from snail into water > penetrates skin > circulates blood > portal blood in liver + matures > adult worms migrate to bowel/rectum or bladder and lay eggs
90
Helminth infection effect on immune system
Increase in mucus production, eosinophilia, bronchoconstriction, smooth-muscle contraction, airway remodelling = help expel helminths BUT chronic helminth infection can dampen the immune system e.g suppression/inhibition of lung eosinophilia + airway hyperresponsiveness But there are inconsistencies in population studies on the effect of helminth infection on allergy
91
Why are there wide variations in observations in helminth infection effect on asthma
* Different helminths have different effects (systemic versus local effects) * Mild versus chronic helminth infections * Different populations in different geographical locations * Varying age groups (children v adults) * Most studies are cross-sectional * Varying outcomes and exposure measures * Even variations in helminth detection methods
91
Randomised Controlled Trials: Does removing parasitic worm infections through anthelminthic treatment lead to more inflammatory conditions? + study e.g
Randomization of sample: Intervention group Anthelminthic treatment > follow up Control group Placebo/no intervention > follow up Outcomes: allergic sensitization, eczema, asthme/wheeze, other inflammatory outcomes e.g : Does anthelminthic treatment during pregnancy increases the risk of allergy in infancy ?? outcome: doctor-diagnosed infantile eczema Conclusion: Exposure to maternal worm infections in utero may protect against eczema in infancy
92
Helminth infections + type 2 diabetes
Helminths are thought to induce an anti-inflammatory immune cell environment which is associated with insulin sensitivity (lowering risk of T2D) In helminth-infected subjects, treatment significantly increased insulin resistance (risk factor for T2D)
93
Immune response during helminth infection?
1) Th1 response: produce IFN-γ + TNF-α > activate macrophages (Mφ) Th1 activation peaks 2) Th2 response: produce cytokines IL-4/5, IL-9/13 Th2 response peaks 3) Regulatory response: Tregs > produce IL-10, TGF-beta Bregs > produce IL-10 Outcome: Survival or spill over tolerance > IBD, T2D, asthma etc
94
Treg and Breg activity in helminth infection
Tregs: B cell modulation, tissue remodelling, inhibition of cells from innate immunity, down-regulation of T effector cells, inhibition of antigen presentation Bregs: inhibition of Mφ, DC, NK, CTL, Th1 activation of Th2, Tregs
94
Helminths effect on Tregs
Treg induction by helminths Increase in Helminth excretory-secretory products (so increase in helminths) = increase in Foxp3 % = enhanced Treg response Treatment of helminth infection = sig reduction in Tregs
95
Worm therapy as a treatment for inflammatory diseases
Colitis * Pre-cllinical Studies = Reduction in disease severity and mortality * Clinical = helminth therapy improved disease activity Type 1 Diabetes * Pre-cllinical Studies = Inhibition diabetes development in NOD mice * Clinical = no clinical studies / conflicting epidemiological data Multiple Sclerosis * Pre-cllinical Studies = Prevention of onset of experimental MS disease in animal models and delay in severity * Clinical = Decrease in relapses and numbers of disease lesions overall tho it is not clear, there are varying results + mainly a limited effect
95
Why do helminth therapy trials show a limited effect
Inconsistency in human studies: - Clinical outcome/severity - Age of the affected population - First infection v endemic population - Type of helminth/intensity - Pathology due to worm infection - Lack of standardized cultures
96
Overview of Dual activity of helminth infection in humans
Chronic helminth infections = associated with immune regulation and protection against allergies Acute helminth infections are associated with induction of Th2-associated responses, increased IgE responses and induction of cross-reactive IgE = pro-inflammatory responses
97
Side effects of helminth infections
Cons: reduced immune responses, increased susceptibility to other infections, reduced anti-tumor immunity, reduced efficacy of vaccines Pros: reduced incidence of allergy/autoimmunity/asthma/IBD
98
Which factors can influence our microbiome
breast milk vs formula vs raw cow milk, antibiotic use, vaccination, close contact with animals, immune system dev, family size, daycare attendance, mode of delivery, maternal diet, maternal antibiotics
99
What is the result of dysbiosis in the microbiota
Immunological dysregulation Immunological Equilibrium: Healthy microbiota = a balanced composition of multiple classes of bacteria = intestinal homeostasis + immune tolerance − Symbionts: organisms with health-promoting functions − Pathobionts: Microbiota with the potential to induce pathology Immunological Disequilibrium: Dysbiosis = unnatural shift in the composition of the microbiota where #no. of symbionts are reduced and/or pathobionts are increased = immune dysregulation
100
Effect of antibiotics on microbiota
Antibiotic treatment = changes in microbiota composition + dysregulation of host immune homeostasis Increased susceptibility to inflammatory disease
101
Effect of short-chain fatty acids on microbiota
SCFA are produced by the gut microbiota from fermentation of dietary fibres. Most common: Acetate Butyrate Propionate Play an important role in immune regulation pathways = fortified barrier function, increased immunity, tolerogenic factors low fiber diets = more eosinophil influx = allergic airway inflammation high fiber diet = less eosinphil influx + more Treg activity = less eosinophil infiltration into airway tissues
102
Interaction between helminth parasites + gut microbiotia
Intestinal microbiota contribute to the ability of helminths to modulate Allergic Inflammation Antibiotic treatment abolishes helminth protection against allergic inflammation Helminths releases excretory-secretory products + acetate Microbiota release SCFA All these lead to Foxp3 Treg cell activation = inactivates airway inflammation
103
Old infections, microbiota and crowd infections in HICs
old infections mostly lost diminished diversity + exposure of microbiota crowd infections are increased esp in inner cities
104
Which microorganisms exist
eukaryotes: prokaryotes: bacteria + archaea
105
Pathogenicity of pathogens
true pathogen = primary pathogens potential pathogen = opportunistic pathogens (dont always cause disease, need an opportunity to do so) non-pathogenic
106
How to prove a causal rel between microbe + disease
Have to prove all 4 of Koch's postulates 1) The microorganism is present in sick but not healthy individuals/animals 2) The microorganism must be grown to pure culture 3) The cultured microorganisms must cause the same disease in healthy individuals/animals 4) The same microorganisms must be culturable from sick individuals/animals but there are many exceptions to these
107
Exceptions to Koch's postulates
* Microorganisms cannot be cultured with available methods or require additional factors for in vitro growth (e.g. host cells - viruses) * Microorganism only causes disease in humans - No proof of disease from animal models * Long incubation period (time between infection and first symptom) - HIV-AIDS, prions (Creuzfeld-Jacob disease) * Microbes that can also asymptomatically colonize: commensals that turn into pathogens
108
Role of our microbiome in health
Colonization resistance: protection against infiltration by new + harmful microorganisms Maturation + instruction of immune system Nutrition + metabolism of food
109
Role of microbiome in disease
Disease association w/ disturbances in (intestinal) microbiome Source of infection (presence of opportunistic pathogens, immunocompromized host)
110
What potentially 'bad bacteria' do we have in our microbiome?
Neisseria meningitidis, Streptococcus pneumoniae, Staphylococcus aureus, Group A Streptococcus
111
What are virulence factors
bacteria-associated molecules that are required for a bacterium to establish an infection and cause disease e.g toxins produced by some bacteria that kills the host cells by creating holes that lead to lysis Infection occurs when virulence factors outweigh the hosts immune defense
112
What are the different functions of virulence factors
1. Colonization 2. Invasion (& dissemination) 3. Immune evasion 4. Nutrient acquisition
113
Infection routes
Mucosal membranes (airway, gut) and skin are common entry sites for bacteria Airway: droplets, aerosols Gut: infected food ingestion Skin: wounds, intravenous lines
114
Symptoms of bacterial infections
Local and/or disseminated (widespread) symptoms - Local: skin redness, local pain, cough, diarrhea, vomiting… - (More) disseminated: fever, headache, confusion, petechiae, stiff neck (meningitis)… May vary depending on: - The route of infection - Type of bacteria causing the infection
115
Host defense mechanisms upon primary bacterial infection (skin)
1) Invading bacteria are detected by local tissue cells: * APCs: DCs, macrophages, mast cells - Pattern-recognition receptors (e.g. TLRs) * Non-immune cells: epithelial cells, keratinocytes, fibroblasts - Activated by bacteria but also in response to tissue damage - Produce AMPs + peptides for local non-specific killing 2) Local cells activate and coordinate downstream immune responses: - Attract phagocytes from the bloodstream (e.g. neutrophil-mediated killing) - In lymph nodes * Activated T- and B-cells, production of specific antibodies * Memory formation: prevent re-infection 3) Convergence of innate and adaptive immunity - antibodies bind to bacterium = phagocytosis - bacterial clearance
115
Antibody-mediated defense against bacterial pathogens
Direct killing of through complement activation (Gram-negative bacteria only) > MAC pore formation = cell lysis as extracellular fluids flood in Enhancing bacterial phagocytosis and killing: - For Gram-ve and Gram+ve bacteria Boost neutrophil recruitment and pathogen recognition > Complement activation (IgM, IgG) = pathogen opsonization + neutrophil recruitment (leading to more effector cells) > interaction with Ig receptors (IgG, IgA) = receptor-mediated phagoytosis
115
What can restore/maintain the balance between host and pathogen
vaccines + antibiotics = kill or inhibit pathogen
116
Why does complement-mediated killing only work for Gram-negative bacteria?
Gram -ve has a much thinner peptidoglycan layer vs gram +ve has a much thicker peptidoglycan layer = MAC gets stuck in gram +ve and thus no lysis
117
How do vaccines work (stages)
Vaccination boosts host defense to increase long-lasting resistance against bacterial infections (inhibits virulence factors) Vaccination - Primary adaptive response (small lag time) Booster vaccination - activates immune system again Immunological memory > When you get primary infection there is no lag time and there is a secondary adaptive immune response
118
1st vaccine
Edward Jenner, 1796 Observation: Milkmaids are protected from smallpox Infection with cowpox virus protects a person from smallpox virus
119
What are subunit vaccines
Made of certain components of the bacteria
120
Vaccine effector mechanisms
Antibodies that neutralize bacterial toxins (that would otherwise cause lysis of host cells) - e.g: tetanus and diphtheria vaccines Induction of bactericidal antibodies that enhance bacterial phagocytosis and killing - e.g: meningococcal and pneumococcal vaccines - immunogenic surface antigens are good vaccine antigens to induce such antibodies
121
What are CPS as virulence factors
Capsular polysaccharides * Encapsulated bacteria have the ability to cause invasive disease e.g Streptococcus pneumoniae, Haemophilus influenzae b, Group B Streptococcus, Neisseria meningitidis * CPS composition varies between bacterial species but even within species * CPS is an important virulence factor: > Resistance to phagocytosis and killing by immune cells > Resistance to serum killing (Gram-ve bacteria) (complement killing, MAC pore)
122
Antibodies targetting capsular polysaccharide itself?
Inverse correlation: bactericidal antibodies against capsular polysaccharide and IMD incidence % of serum w/ bactericidal activity increases = IMD incidence decreases (AS YOU GET OLDER)
123
Example of a bacteria that is a commensal and pathogen
Neisseria meningitidis
124
Why do new borns have a low incidence of Invasive meningococcal disease (IMD)
They are protected by maternal antibodies
125
Hypothesis: Individuals at risk for IMD can be protected by inducing bactericidal Ab through vaccination.
Plain polysaccharide vaccines (isolated sugars from the coat + used in the vaccine) Insufficient for population-wide application: * Not immunogenic in young children (most ‘at risk’ group) * No immunological memory → repeated immunizations necessary to protect * Ineffective against carriage (commensal state, colonization) → only individual protection T cell-independent immune response: * Production of low affinity antibodies (T cells do not recognize these sugars) * No memory formation
126
How can vaccinations with polysaccharides be made more effective
Protein conjugation: CPS conjugated to a protein carrier T cell-dependent responses: T cell recognizes the protein = provides help to produce high affinity antibodies + polysaccharise-specific memory B cells
127
Benefits of polysaccharide conjugate vaccine vs plain polysaccharide vaccine:
- Immunological memory - High affinity antibodies - Effective in infants - Herd immunity: protection of non-vaccinated individuals (as consequence of reducing carriage (transmission))
128
How do polysaccharide conjugate vaccines provide herd immunity
- Protection of non-vaccinated individuals - Consequence of reduction in carriage resulting in reduced circulation among population
129
meningococcal serogroups
Polysaccharide capsule structure determines serogroup: - 13 serogroups - groups A, B, C, X, Y, W most prevalent in disease
130
Multivalent glycoconjugate vaccines
extracting CPS from different serogroups of Neisseria meningitidis, conjugate them to protein carriers (put all together in vaccine) > broader protection
131
Which potentially 'bad bacteria' amongst our microbiome do we have vaccines for
Neisseria meningitidis + Streptococcus pneumoniae NOT for the other 2
132
Limitations/challenges for vaccines based on CPS
*Limited vaccine coverage in case of wide range in CPS diversity > S. pneumoniae > 90 serotypes * Glycan antigen is not immunogenic * Glycan structures are similar to human structures (molecular mimicry) >Risk of inducing autoreactive antibodies >Group A Streptococcus, N. meningitidis serogroup B * Disease caused by low or non-encapsulated species/strains (often called nontypeable) >Examples: Group A Streptococcus (certain emm-types), non-typeable H. influenzae, Staphylococcus aureus
133
Necessary characteristics of cancer cells
They stimulate their own growth, ignore growth-inhibiting signals, avoid death by apoptosis, angiogenesis, metastasis, replicate constantly to expand their numbers, evade + outrun the immune response
134
steps from early mutation to cancer
initiation: somatic cell experiences mutational event cancer progression: (proliferating cells) mutation + genome destabilization, dysregulation of growth control pathways evasion of cancer cell elimination: (precancerous cells) block apoptisis, block killing by T cells tumor growth + dispersal: (tumor) angiogenesis + metastasis
135
typical response to cancer therapy metastatic cancer
as time progresses survival decreases microevolution: the cells that can escape have an advantage cancer therapy does not cure only a gain of extra time
136
Relation between CD8+ T cells and tumor prognosis
correlation between abundance of CD8+ in tumors with tumor progression fewer CD8 = worse prognosis more CD8 = better prognosis
137
What makes immune system suited to fight cancer?
Adaptive Access Memory
138
Special role (CD8) T cells in (in cancer context)
specific through their Antigen Receptor (TCR) allows them to see 'inside' cells viral peptides are cut and presented on APCs in MHC (binds to TCR = activated T cell) Activated T cells can kill infected cells and cancer cells with great precision (oncogenes > mutated proteins > neo-antigens) T cells expand by proliferation = well suited to fight cancer progression
139
Different types of tumor antigens
Start from peptide antigens presented by MHC class I on normal cells which then undergo mutations and become cancerous Tumor-specific antigen - Presentation of mutant peptide from mutated cellular protein (neo-antigens) Tumor-associated antigens - reactivation of embryonic genes not normally expressed in the differentiated cell - overexpression of self protein by a tumor cell increases self-peptide presentation + recognition by T cells
140
Initiation of immune response against cancer?
Initiation of an immune response requires inflammation (danger!) 1) inflammatory inducers (e.g PAMPs) 2) Sensor cells (express PRRs) (e.g macrophages, DCs) 3) mediators (e.g cytokines, cytotoxicity) 4) target tissues
140
Cancers usually dont have a microbial trigger so what acts as PAMPs for cancer cells
DAMPs: danger-associated molecular pattern released when cells are dying uncontrollably
141
Requirements effective T cell immunity against cancer
- Recognition tumor antigen - Activation - Migration - Effector function
142
T cell activation by DC: co-stimulation (cancer context)
CD28 interacts with CD80/86 and produce cytokines
143
Steps in induction T cell response against cancer
Tumor w/ DCs > DCs activate T cells in LNs > T cells migrate to tumor > T cells kill tumor cells
144
The mellman cancer immunity cycle
1) release of cancer cell antigens 2) cancer antigen presentation 3) priming + activation in LN 4) trafficking of T cells to tumors 5) infiltration of T cells in to tumors 6) recognition of cancer cells by T cells 7) killing of cancer cells (more dead cells = release new antigens = cycle repeats
144
What are hot and cold tumors
Hot tumors are inflammed, cold tumors = immune system doesnt get in e.g by suppression of chemokine production
145
Immune exclusion - Hurdle for immunity against cancer
Immune-excluded cancer (presence of T cells at invasive margin but abscent in tumor bed I.S cannot penetrate tumor) mechanism of immune evasion: stromal barriers, aberrant vasculature, lack of chemokines, ongogenic pathways, hypoxia
146
Immunoediting
Elimination, Equilibrium, Escape Microevolution = cancer cells escape immune system by loss of tumor antigen expression
147
Often tumor cells have more than 1 antigen + it is very unlikely that they lose all. How else can they undergo immune escape
immune escape by HLA loss 1) antigen loss 2) decrease in subunits of proteosome 3) decrease in TAP 4) loss of MHC I heavy chain 5) loss of beta-2 microglobulin (by mutations for example) 6) diminished transport of MHC I-peptide complexes to cell surface Hard MHC-I lesions: genetic alterations leading to permanent loss Soft MHC-I lesions: are reversible BUT NK cells are activated when there is a loss of MHC
148
T cell tolerance (cancer)
*Tregs: inhibit responses to self antigens, Treg abundance correlates w/ poor prognosis, low treg = better prognosis + anti-tumor response (but risk of autoimmune diseases) * Exhaustion > T cells get "exhausted" + lose their effector function/proliferation There are transcriptional factors that promote exhaustion Sustained expression of inhibitory receptors on exhausted CD8+ T cells * TAMs (e.g MR- macrophage associated w/ wound healing, cancers require constant repair) * MDSC (make factors that inhibit killing of cancer cells) > tumors shape their microenvironment: suppressive factors condition the skin + LN and sabotage immune protection against metastatic spread Subverted myeloid cells (MDSC, M2 macrophages) release pro-angiogenic + tumor invasion promoting factors = important cause of therapy resistance
148
How can the TCR know the nature of its antigen (self? or tumor?)
by extra information e.g inflammation/costimulation
149
T cell differentiation by DC
DCs can interact with naive T cells and lead to differentiation into Tregs = tolerance + immune suppression
150
Inhospitable metabolic tumor environment
tumoral hypoxia, increase in lactate, increase in acidification
151
Conclusions cancer immunity
*Many cancers are immunogenic * tumor antigens/neo-antigens * Adaptibility immune response asset * Immune editing > Loss of antigen/HLA * Tolerance mechanisms > Tregs, MDSC, TAM > T cell exhaustion * Inhospitable tumor environment
152
Checkpoint inhibition
immunotherapy for cancer mobilizes T cell responses targets exhaustion make antibodies that target these inhibitory receptors
153
Immune checkpoints: inhibitory receptors (cancer)
Inhibitory receptors: PD-1 and CTLA4 > turn off T cells Only happens when ligand is there, antibody blocks ligand (from checkpoint inhibition)
154
Efficacy of immune checkpoint inhibitors (graph based)
survival curve flattens out effects of anti-CTLA-4 (ipilimumab) and PD-1 blockade (nivolumab) are enhanced when used in conjunction
155
Neoadjuvant therapy (Immune-therapy before operation) efficacy
it is very very effective
156
why does immunotherapy not always work in cancer therapy
There are many inhibitory receptors Responsiveness to Checkpoint Inhibition correlates with mutational load High mutation burden = increase in likelihood of generating neoantigens (which IS can target)
157
PD1
Loss of HLA = cancer cells can evade detection Tolerance mechanism by PD-L1: pathway suppresses T cell activity = cancer can evade IS but breaking tolerance w/ treatments can result in side effects
158
T cell therapy: Tumor infiltrating Lymphocytes (TIL)
extract T cells, alter, inject back in effective disadvantage: Patient-specific (laborious + expensive)
159
More universal immunotherapy approaches
BITEs: activate all T cells in tumors BITE antibody is composed of 2 single chains from diff antibodies (CD3 monoclonal Ab + target monoclonal Ab) leads to T-cell activation (by binding to T cells) + redirected lysis (by binding to tumor-associated antigens present on the target cells)
160
advantages + disadvantages of BITEs
No need antigen-specific T cells No need antigen-presentation Relatively cheap Disadvantage: need T cells in the tumor already (i.e it does not work on cold tumors)
161
CAR T cells
Chimeric Antigen Receptor Antibodies bind different types of antigen than TCR and can be generated against any protein fusion Antibody and TCR CAR T cells recognize and kill leukemic cells (their activation is independent of MHC presentation so they can bypass tumor cells evasive mechanisms)
162
Next generations of CAR T cells
Builds upon the 1st generation by adding one costimulatory signaling domain (e.g., CD28 or 4-1BB). (combines signal 1 (CD3ζ-mediated activation) with signal 2 (costimulation), enhancing T-cell proliferation, survival, and cytokine release) 3rd gen adds 2 costimulatory domains improves therapeutic efficacy
163
How does one receive CAR T cell therapy transfer
leukopheresis (blood removed from patient) > retroviral transduction w/ CAR > CAR T cell infusion
164
Conclusions Immunotherapy (cancer)
* Checkpoint inhibition > Revolutionary! > Role antigen > Neoadjuvant therapy * TIL therapy * BiTEs * CAR T cells