Checkpoint inhibitors Flashcards

1
Q

Draw diagram of ICIs

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

Describe T cell activation

A

Need a number of signals. First is TCR and complementary antigen bound to MHC. If this is only signal = anergy. Second signal is CD28 binding to one of the B7 (CD80 and CD86) on the APC. Cytokine environment then determines which differentiation state the T cell will commit to. To prevent excess activation, T cell upregulates CTLA-4 which also binds B7 (10x higher affinity thna CD28). This is coupled to inhibitory signalling cascade and dampens T cell response

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

What sort of antigens can tumour express?

A

can be from oncogenic viruses, differentiation antigens, epigenetic alterations in cancer cells, or neoantigens derived from mutations associated with carcinogenesis

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

What have we found out in the last 5 years or so about CTLA-4?

A
  • doesn’t just bind to CD80/CD86 on the APC, but it also removes them from the membrane – ‘hoovers them off’
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Describe PD-1 mechanism

A
  • Cancer cells under attack from T cells release IFN-y which leads to expression of PD-L1
  • PD-L1 binds PD-1 on T cells
  • PD-1 phosphorylated on its ITAM motifs in its cytoplasmic tail
  • This recruits SHP-2 which keeps the TCR dephosphorylated, preventing TCR signalling
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Waterhouse et al. (1995)

A
  • first demonstrated the importance of this regulatory process by creating CTLA4 knockout mice, which consequently had huge and uncontrollable proliferation and activation of T cells, leading to their death.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Leach et al. (1996)

A
  • Was a post-doc working w Jim Allison
  • Showed CTLA4 to be a negative regulator of T cell activation, which earned a Nobel Prize.
    • They injected BALB/c mice with B7-1 transfected colon cancer cells.
    • These mice received an anti-CTLA4 antibody, an anti-CD28 antibody, or remained as untreated controls.
    • The anti-CTLA4 antibody led to the rejection of pre-established tumours – they regressed in size or were obliterated completely.
    • 90% of the cancers in treated mice disappeared.
  • This seminal work resulted in the birth of the first checkpoint inhibitors in cancer immunotherapy – anti-CTLA4 monoclonal antibodies.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Describe anti-CTLA4 mAb approval

A

Ipilimumab

Approved in 2011 by FDA for metastatic melanoma

Has now doubled 10-year survival for mm

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

Hodi et al. (2010)

A
  • In a seminal phase III trial
  • was shown to significantly prolong the lives of patients with stage 4 metastatic melanoma. The median survival with ipilimumab plus the gp100 vaccine (against tumour antigen) was 10 months compared to 6.4 months with the gp100 vaccine alone. Some cases were even ‘cured’ as they showed durable remission.
    • The tail of the curve was a plateau – indicated they were stable – this was v exciting!
    • Seems to be permanent!
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Robert et al. (2011)

A
  • conducted a phase III trial of over 500 patients with untreated metastatic melanoma, who were assigned to either ipilimumab plus dacarbazine (I + D), or dacarbazine plus placebo. The overall survival of the I + D group was 11.2 months, compared to 9.1 months for dacarbazine alone.
    • However, there were significant autoimmune-related side-effects:
      • 40% had skin reactions
      • some had vitiligo, hepatitis
      • N + V
      • colitis (which led to diarrhoea).
    • This was a new class of side effects that had rarely been seen with conventional therapeutics
    • Understanding of why these therapies are linked to such side effects may help autoimmunity in general
  • Nevertheless, was approved by the FDA for clinical use in 2011 ∴ consensus = its benefits override these SEs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Robert et al. (2015)

A
  • The efficacy of the anti-PD1 antibody Nivolumab was shown in a phase III clinical trial for patients with advanced melanoma (without a BRAF mutation)
    • They compared nivolumab (N) treatment to a dacarbazine (D) control group. The 1 year survival rates were ~70% for patients who received N, and less than 40% for patients on D; progression-free survival was 40% for patients on N, and less than 10% for patients on D.

Accordingly, antibodies targeting the PD1-PDL1 axis have been approved as second-line or first-line therapies for an ever-growing list of malignancies, including melanoma, lymphoma, lung cancers, renal cell cancer (RCC), bladder cancer and more.

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

Whats the difference between sites of action of PD-1 and CTLA-4

A
  • PD1 acts in the TME/peripheral tissues whereas CTLA-4 acts in the lymph nodes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Larkin, 2015

A
  • Since PD1 + CTLA4 regulate different inhibitory pathways on T cells, combination therapy w Abs targeting both molecules were tested - may have additive/synergistic effects
  • Phase III clinical trial of nivolumab alone vs nivolumab + ipilimumab, vs ipilimumab alone in pts w untreated metastatic melanom
    • Progression free survival = 11.5 months with combination; 6.9 months w N, 2.9 w I
    • BUT highly toxic, more adverse effects in comb group than either alone, but mostly manageable w established treatment guidelines.
      • Difference between N alone and combination is not significant to claim comb is better
      • But, both or N alone = better than I alone
      • This trial used biomarkers → establish PDL1 +ve and –ve tumours but:
        • IHC (immunohistochemistry) to look at PDL1  Abs are not that good
        • Currently unclear how PDL1 or Ag is processes
      • Therefore, difficult to draw confident conclusions of differences in treatment response. More work needed.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Example of tumours failing to respond to ICIs

A

For example, a study in 2018 trialled an anti-PD1 therapy in prostate cancer patients, but in most cases, tumours continued to grow despite therapy. Only ~5% of tumours demonstrated shrinking.

Furthermore, two clinical trials in 2019 showed no differences in PD-1 vs existing standard of care for HCC and also in mesothelioma

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

What are the key points in the ‘what determines success of ICIs’ section?

A
  1. Tumour genomes (TMB and high fitness neoantigens, identifying sensitive subgroups)
  2. Pt germline genetics
  3. Immune microenvironment (PD-1 expression, TILs, MDSCs, commensal microbiota)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Define TMB

A
  • total number of somatic mutations per coding area of a tumour genome
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Alexandrov et al. (2013)

A
  • global sequencing initiative - analysed the mutations from over 7000 cancer samples from 30 different cancer classes.
    • Cancers related to chronic exposure to mutagens e.g. lung cancer (tobacco smoking), malignant melanoma (UV exposure) = highest prevalence of mutations + related to specific mutational signatures
      • E.g., malignant melanoma – higher prevalence of ‘signature 7’ = characterised by C>T mutations  likely due to formation of pyrimidine dimers (bulky, helix-distorting lesion) resulting from UV exposure
    • What’s interesting  some of the best initial ICI response rates were observed in carcinogen-driven cancers such as melanoma and NSCLC, which have high TMBs.
    • Moreover, the negative studies were typically for cancers with a low TMB.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Snyder et al. (2014)

A
  • Obtained tumour samples from melanoma pts who were treated w anti-CTLA4.Performed whole-exome sequencing on tumours + matching blood samples (massively parallel seq)
    • Degree of clinical benefit was associated w TMB but TMB alone could not predict clinical benefit
      • E.g. because some patients with low TMB show good response
  • Instead, they identified that response can be predicted by high-fitness neoantigens
    • They used genome-wide somatic neoepitope analysis and patient-specific HLA typing  identified proposed tumour neoantigens for each patient
    • They illustrated a neoantigen landscape that is found specifically in tumours that respond strongly to anti-CTLA4 therapy
    • This was validated in a second group of pts
    • These predicted neoantigens activated T cells from pts treated w ipilimumab in vivo
  • Implications  warrants analysing exomes of patients who are being considered for anti-CTLA4 treatment  predict response
    • It must however be considered that this was a relatively small cohort
    • Although the neoepitopes identified may be the most important ones, the in vivo relative contribution of each is unknown
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Why is it thought high TMB tumours respond best to ICIs?

A
  • It is thought that high-TMB-tumours respond best to ICI because the more mutations a tumour has, the greater its neoantigen heterogeneity is likely to be  tumour will likely have more immunogenic neoantigens (i.e. foreign to the immune system  a T cell response is more likely)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Marabelle et al 2020

A
  • Paper in Lancet
  • Explored associated between high TMB and outcomes in ten tumour-type specific cohorts from phase 2 KEYNOTE-158 study which looked at anti-PD1 agent in patients with solid tumours
  • (NB – did not include breast, prostate and mismatch repair proficient CRC)
  • Quantified TMB with NGS sequencing panel
  • Although they found high TMB was associated with better overall survival – the difference was only modest

As a result of this, FDA approved pembrolizumab for adults and children with TMB-high solid tumours

  • May be a bit too soon to have done this?
  • In the study – 10% of pts had treatment-related serious adverse effects and 15% had moderate adverse effects
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Gurajao et al (2020)

A
  • Larger cohort - 2500
  • They showed that there is a difference between responders and non-responders but this is actually limited to a small number of tumour types (such as lung cancer and melanoma)
  • They point out that the relationship between TMB and response is largely due to these tumour types and because the drugs are usually used in these tumour types, there is likely an ascertainment bias (data more likely to include some members of a population than others)
  • They also had a look at the cut point that the FDA accepted that defined tumours with high TMB – this was 10 mutations per Mb – they found that according to this cut off, 56% of responders are below this and could be denied effective therapy
  • Similarly, pts who have disease with higher TMB than this cut-off also showed not to respond
  • Criticism – this is a preprint and has not been subject to peer review
  • Difficult, because you would want to do a RCT but these drugs have such good clinical benefit – ethical dilemma
  • Leads us to question – could we use ICIs in combination with radiotherapy to increase TMB?
    • We have had positive results from clinical trials with ICI or placebo with chemoradiotherapy but we have had no study to date that has a control arm with immunotherapy alone
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Le et al 2015

A
  • investigated whether MMR deficiency was related to the response rate and immune-related progression-free survival of two groups of colorectal cancer patients treated with pembrolizumab (anti-PD-1 antibody), one group with MMR deficiency, and the other with an intact MMR system.
  • The group also performed whole exome sequencing of each tumour.
  • The results were consistent with Snyder’s first description of mutational load and responsiveness to immunotherapy – the MMR deficient tumours contained a mean of 1782 somatic mutations and showed a 78% immune-related progression-free survival, compared to a mean of 73 somatic mutations in MMR proficient tumours, which showed a 0% objective response rate and 11% immune-related progression-free survival.

With the recent approval of nivolumab and pembrolizumab for the treatment of MSI-positive cancers of any histology in 2017, anti-PD1 therapy is the first therapy to be granted FDA approval on the basis of a specific tumour genetic characteristic independent of tumour histology.

MSI (microsatellite instability) results from impaired MMR

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Why does MMR deficiency lead to sensitivity to ICIs?

A
  • We now understand that mechanistically, MSI-positive tumours have a high TMB; MMRd generates a high mutational load, particularly through indel mutations, some of which result in frameshifts that produce neoantigens that may be more immunogenic on average owing to their greater sequence divergence from self-peptides.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Zaretsky et al. (2016)

A
  • analysed paired baseline and relapsing lesions in four melanoma patients who had had an initial tumour regression in response to anti-PD1 therapy followed by disease progression months to years later.
    • Did whole-exome sequencing to compare baseline and matched relapsed tissues
    • Whole-exome sequencing revealed acquired loss of function JAK1/2 mutations in two patients,
    • They then wanted to assess the functional consequences of the JAK mutations
    • Western blot showed that the baseline cell line from the patient with the JAK2 mutation responded to IFNy with the expected increase in signal transduction proteins. The relapsed tumour displayed an insensitive to IFNy.
    • A third patient had a truncating mutation in the beta-2-microglobulin (B2M) gene, which impaired cell surface expression of MHC class I and subsequent antigen presentation to cytotoxic T cells.
    • These are only three patients – many other mutations could be involved?

Also there could be confirmation bias

25
Q

Chowell et al. (2018)

A
  • analysed over 1,535 patients with cancer treated with ICIs, and found that HLA-I heterozygosity (a more diverse array of HLA-I molecules) in a given individual was associated with increased survival, possibly owing to the ability to present a broader range of tumour antigens to T cells.
  • Interestingly, this effect of HLA-I heterozygosity on increased survival after ICI seemed mostly associated with the HLAB and HLAC loci, presumably because the MHC encoded by HLAB binds to a greater diversity of peptides and HLAC is generally expressed at higher levels in APCs than in other cell types.
  • By contrast, they associated LOH at HLA-I loci with reduced response to ICI therapy.

This could facilitate an immune evasion mechanism – if tumours downregulate HLA-I expression**, they disrupt the antigen presentation pathway. Good point ☺

26
Q

Does PDL1 expression and related FDA approval

A

PDL1 expression is therefore thought to be a predictor of response, and the FDA has currently approved PDL1 immunohistochemistry assays as a companion diagnostic for anti-PD1 therapy for patients with NSCLC.

27
Q

Topalian et al 2012

A
  • Took samples from patients prior to treatment with nivolumab (anti-PD-1 antibody) and performed immunohistochemical analysis on the sample.
    • 36% of tumours expressing PD-L1 showed an objective response, whereas no objective response was seen in patients with PD-L1 negative tumours.
  • These results have been confirmed in a number of other studies….
  • However, issues w immunohistochemical analysis:
    • Tumours may have variable expression PD-1 so that it may be differently expressed at different anatomical sites – pt might be classed differently depending on where a sample was taken from
    • Moreover, a tumour may up or down-regulate PD-L1 at different points in time
28
Q

Sunshine and Taube (2015)

A

combined data from a number of studies assessing PD-L1 tumour expression via immunohistochemical analysis and response to anti-PD-1 immunotherapy and found an approximate 15% response rate in patients classified as PD-L1 negative. These results seem to indicate PD-L1 expression is not the most accurate marker as to whether or not a patient will respond to anti-PD-1 immunotherapy, therefore it should not be used exclusively to determine whether or not a patient will receive this treatment as it could be denying patients access to a therapy that may be of benefit to them

29
Q

Reasons for contrast between Topalian 2012 and Sunshine and Taube 2015

A
  • to use of different detection assays or non- standardised criteria and cut-offs for assessing positivity, such as percent PDL1+ versus staining intensity. Furthermore, even when PDL1 expression is correlated with responses, there are many patients with low to no detectable PDL1 expression who experience durable clinical benefit. Good ☺
30
Q

Tumeh et al. (2014)

A
  • reported that CD8+ cell density in metastatic melanoma, as measured by IHC at the invasive margin of a tumour, as opposed to central infiltration, is most strongly associated with anti-PD1 response.
  • But there are limitations to IHC:
    • Not that sensitive
    • Background staining occurs 🡪 binding to non-specific reactive sites
    • Difficult to quantify protein
    • Not well standardised – different people use different protocols, different antibodies
    • Yes it’s cheap + pretty robust but it definitely has disadvantages!
31
Q

How could we increase density of TILs?

A

Incorporating vaccination into the treatment

32
Q
  • De Henau et al (2016)
A
  • Selective targeting of infiltrating MDSCs with PI3Ky inhibitors (PI3Ky = highly expressed in myeloid cells) was shown to restore sensitivity to anti-PD1 and anti-CTLA4 in murine models, and this targeting promoted cytotoxic T cell-mediated tumour regression
  • Could localising and phenotyping immune cells in the TME be used as a biomarker?
  • Could we use a SMI against PI3Ky to overcome resistance in pts with high levels of MSDCs in tumours?
33
Q

Give overview of role of commensal microbiota

A
  • Different strains of bacteria that associate with response or resistance have been identified in studies for melanoma, NSCLC, RCC and urothelial cancer.
    • The reasons for these variable results could include differences in microbial sequencing and analysis techniques, and geographic variations in the distribution of gut flora.
34
Q
  • Matson et al. (2018)
A
  • Looked at stool samples from 42 patients with metastatic melanoma pre ICI treatment
  • Sequence 16 ribosomal subunit which has different expression among different bacteria
  • found that the ratio of response-associated (‘good’) to resistance-associated (‘bad’) bacteria was able to clearly stratify human responders from non-responders.
  • Also find that studies showing antibiotics prior to ICB is associated with adverse outcome
    • Sceptical of this because the need for antibiotics suggests a pt is frail and so on
  • Lit review in Oct 2016 showed 36/38 human microbiotas studies were successful – not publishing negative data
  • What is the mechanism underlying this – is it causal or correlative?
  • These results implicate the gut microbiota as an influential factor in antitumour immunity and ICI response.
35
Q

Caveats of ICIs

A
  • Releasing the brakes on T cell activation may have limited effect if the patient does not already possess tumour specific T cells with a sufficient level of affinity for tumour antigen
  • Significant immune-related toxicities
36
Q
  • Papadopoulos et al 2019
A
  • Investigating REFN3767 – a mAb that targets LAG-3
  • This group have published results from an ongoing phase I trial of this compound
  • A total of 67 patients (25 in the monotherapy cohort and 42 in the combination group with an anti-PD-1 mAb)
  • 11/25 in the monotherapy and 6/42 in combination achieved stable disease
  • Trial is still open and recruiting

Which is better – monotherapy or combination? Which agent is best to combine with? Does combination lead to more adverse effects? Many unanswered questions

37
Q
  • Ribas et al 2017
A
  • Phase 1b clinical trial
  • Used an oncolytic virus
    • Was based on HSV but mutated in such a way that it cant infect normal cells, but could in tumour cells – injected into tumour mass
  • 21 pts with advanced melanoma given onco virus in comb with pembrolizumab
  • Generally was well tolerated
  • Pts had 62% overall response rate (with increased CD8 T cells, elevated PD-L1 protein expression)
  • Cav – not controlled, small, you need an tumour mass that’s accessible to inject
38
Q

Valero, 2021

A
  • recently identified that a higher neutrophil-to-lymphocyte ratio is significantly associated with poorer overall and progression free survival after ICI therapy – can achieve a high OR (3.22) when combined with tumour mutational burden
    • this is important as easy to measure – wouldn’t require extensive genome / biopsy, that takes time (just a peripheral blood sample)
39
Q
  • first demonstrated the importance of this regulatory process by creating CTLA4 knockout mice, which consequently had huge and uncontrollable proliferation and activation of T cells, leading to their death.
A

Waterhouse et al. (1995)

40
Q
  • Was a post-doc working w Jim Allison
  • Showed CTLA4 to be a negative regulator of T cell activation, which earned a Nobel Prize.
    • They injected BALB/c mice with B7-1 transfected colon cancer cells.
    • These mice received an anti-CTLA4 antibody, an anti-CD28 antibody, or remained as untreated controls.
    • The anti-CTLA4 antibody led to the rejection of pre-established tumours – they regressed in size or were obliterated completely.
    • 90% of the cancers in treated mice disappeared.
  • This seminal work resulted in the birth of the first checkpoint inhibitors in cancer immunotherapy – anti-CTLA4 monoclonal antibodies.
A

Leach et al. (1996)

41
Q
  • In a seminal phase III trial
  • was shown to significantly prolong the lives of patients with stage 4 metastatic melanoma. The median survival with ipilimumab plus the gp100 vaccine (against tumour antigen) was 10 months compared to 6.4 months with the gp100 vaccine alone. Some cases were even ‘cured’ as they showed durable remission.
    • The tail of the curve was a plateau – indicated they were stable – this was v exciting!
    • Seems to be permanent!
A

Hodi et al. (2010)

42
Q
  • conducted a phase III trial of over 500 patients with untreated metastatic melanoma, who were assigned to either ipilimumab plus dacarbazine (I + D), or dacarbazine plus placebo. The overall survival of the I + D group was 11.2 months, compared to 9.1 months for dacarbazine alone.
    • However, there were significant autoimmune-related side-effects:
      • 40% had skin reactions
      • some had vitiligo, hepatitis
      • N + V
      • colitis (which led to diarrhoea).
    • This was a new class of side effects that had rarely been seen with conventional therapeutics
    • Understanding of why these therapies are linked to such side effects may help autoimmunity in general
  • Nevertheless, was approved by the FDA for clinical use in 2011 ∴ consensus = its benefits override these SEs
A

Robert et al. (2011)

43
Q
  • Since PD1 + CTLA4 regulate different inhibitory pathways on T cells, combination therapy w Abs targeting both molecules were tested - may have additive/synergistic effects
  • Phase III clinical trial of nivolumab alone vs nivolumab + ipilimumab, vs ipilimumab alone in pts w untreated metastatic melanom
    • Progression free survival = 11.5 months with combination; 6.9 months w N, 2.9 w I
    • BUT highly toxic, more adverse effects in comb group than either alone, but mostly manageable w established treatment guidelines.
      • Difference between N alone and combination is not significant to claim comb is better
      • But, both or N alone = better than I alone
      • This trial used biomarkers → establish PDL1 +ve and –ve tumours but:
        • IHC (immunohistochemistry) to look at PDL1  Abs are not that good
        • Currently unclear how PDL1 or Ag is processes
      • Therefore, difficult to draw confident conclusions of differences in treatment response. More work needed.
A

Larkin, 2015

44
Q
  • global sequencing initiative - analysed the mutations from over 7000 cancer samples from 30 different cancer classes.
    • Cancers related to chronic exposure to mutagens e.g. lung cancer (tobacco smoking), malignant melanoma (UV exposure) = highest prevalence of mutations + related to specific mutational signatures
      • E.g., malignant melanoma – higher prevalence of ‘signature 7’ = characterised by C>T mutations  likely due to formation of pyrimidine dimers (bulky, helix-distorting lesion) resulting from UV exposure
    • What’s interesting  some of the best initial ICI response rates were observed in carcinogen-driven cancers such as melanoma and NSCLC, which have high TMBs.
    • Moreover, the negative studies were typically for cancers with a low TMB.
A

Alexandrov et al. (2013)

45
Q
  • Obtained tumour samples from melanoma pts who were treated w anti-CTLA4.Performed whole-exome sequencing on tumours + matching blood samples (massively parallel seq)
    • Degree of clinical benefit was associated w TMB but TMB alone could not predict clinical benefit
      • E.g. because some patients with low TMB show good response
  • Instead, they identified that response can be predicted by high-fitness neoantigens
    • They used genome-wide somatic neoepitope analysis and patient-specific HLA typing  identified proposed tumour neoantigens for each patient
    • They illustrated a neoantigen landscape that is found specifically in tumours that respond strongly to anti-CTLA4 therapy
    • This was validated in a second group of pts
    • These predicted neoantigens activated T cells from pts treated w ipilimumab in vivo
  • Implications  warrants analysing exomes of patients who are being considered for anti-CTLA4 treatment  predict response
    • It must however be considered that this was a relatively small cohort
    • Although the neoepitopes identified may be the most important ones, the in vivo relative contribution of each is unknown
A

Snyder et al. (2014)

46
Q
  • Paper in Lancet
  • Explored associated between high TMB and outcomes in ten tumour-type specific cohorts from phase 2 KEYNOTE-158 study which looked at anti-PD1 agent in patients with solid tumours
  • (NB – did not include breast, prostate and mismatch repair proficient CRC)
  • Quantified TMB with NGS sequencing panel
  • Although they found high TMB was associated with better overall survival – the difference was only modest

As a result of this, FDA approved pembrolizumab for adults and children with TMB-high solid tumours

  • May be a bit too soon to have done this?
  • In the study – 10% of pts had treatment-related serious adverse effects and 15% had moderate adverse effects
A

Marabelle et al 2020

47
Q
  • Larger cohort - 2500
  • They showed that there is a difference between responders and non-responders but this is actually limited to a small number of tumour types (such as lung cancer and melanoma)
  • They point out that the relationship between TMB and response is largely due to these tumour types and because the drugs are usually used in these tumour types, there is likely an ascertainment bias (data more likely to include some members of a population than others)
  • They also had a look at the cut point that the FDA accepted that defined tumours with high TMB – this was 10 mutations per Mb – they found that according to this cut off, 56% of responders are below this and could be denied effective therapy
  • Similarly, pts who have disease with higher TMB than this cut-off also showed not to respond
  • Criticism – this is a preprint and has not been subject to peer review
  • Difficult, because you would want to do a RCT but these drugs have such good clinical benefit – ethical dilemma
  • Leads us to question – could we use ICIs in combination with radiotherapy to increase TMB?
    • We have had positive results from clinical trials with ICI or placebo with chemoradiotherapy but we have had no study to date that has a control arm with immunotherapy alone
A

Gurajao et al (2020)

48
Q
  • investigated whether MMR deficiency was related to the response rate and immune-related progression-free survival of two groups of colorectal cancer patients treated with pembrolizumab (anti-PD-1 antibody), one group with MMR deficiency, and the other with an intact MMR system.
  • The group also performed whole exome sequencing of each tumour.
  • The results were consistent with Snyder’s first description of mutational load and responsiveness to immunotherapy – the MMR deficient tumours contained a mean of 1782 somatic mutations and showed a 78% immune-related progression-free survival, compared to a mean of 73 somatic mutations in MMR proficient tumours, which showed a 0% objective response rate and 11% immune-related progression-free survival.

With the recent approval of nivolumab and pembrolizumab for the treatment of MSI-positive cancers of any histology in 2017, anti-PD1 therapy is the first therapy to be granted FDA approval on the basis of a specific tumour genetic characteristic independent of tumour histology.

A

Le et al 2015

49
Q
  • analysed paired baseline and relapsing lesions in four melanoma patients who had had an initial tumour regression in response to anti-PD1 therapy followed by disease progression months to years later.
    • Did whole-exome sequencing to compare baseline and matched relapsed tissues
    • Whole-exome sequencing revealed acquired loss of function JAK1/2 mutations in two patients,
    • They then wanted to assess the functional consequences of the JAK mutations
    • Western blot showed that the baseline cell line from the patient with the JAK2 mutation responded to IFNy with the expected increase in signal transduction proteins. The relapsed tumour displayed an insensitive to IFNy.
    • A third patient had a truncating mutation in the beta-2-microglobulin (B2M) gene, which impaired cell surface expression of MHC class I and subsequent antigen presentation to cytotoxic T cells.
    • These are only three patients – many other mutations could be involved?

Also there could be confirmation bias

A

Zaretsky et al. (2016)

50
Q
  • analysed over 1,535 patients with cancer treated with ICIs, and found that HLA-I heterozygosity (a more diverse array of HLA-I molecules) in a given individual was associated with increased survival, possibly owing to the ability to present a broader range of tumour antigens to T cells.
  • Interestingly, this effect of HLA-I heterozygosity on increased survival after ICI seemed mostly associated with the HLAB and HLAC loci, presumably because the MHC encoded by HLAB binds to a greater diversity of peptides and HLAC is generally expressed at higher levels in APCs than in other cell types.
  • By contrast, they associated LOH at HLA-I loci with reduced response to ICI therapy.

This could facilitate an immune evasion mechanism – if tumours downregulate HLA-I expression**, they disrupt the antigen presentation pathway. Good point ☺

A

Chowell et al. (2018)

51
Q
  • Took samples from patients prior to treatment with nivolumab (anti-PD-1 antibody) and performed immunohistochemical analysis on the sample.
    • 36% of tumours expressing PD-L1 showed an objective response, whereas no objective response was seen in patients with PD-L1 negative tumours.
  • These results have been confirmed in a number of other studies….
  • However, issues w immunohistochemical analysis:
    • Tumours may have variable expression PD-1 so that it may be differently expressed at different anatomical sites – pt might be classed differently depending on where a sample was taken from
    • Moreover, a tumour may up or down-regulate PD-L1 at different points in time
A

Topalian et al 2012

52
Q

combined data from a number of studies assessing PD-L1 tumour expression via immunohistochemical analysis and response to anti-PD-1 immunotherapy and found an approximate 15% response rate in patients classified as PD-L1 negative. These results seem to indicate PD-L1 expression is not the most accurate marker as to whether or not a patient will respond to anti-PD-1 immunotherapy, therefore it should not be used exclusively to determine whether or not a patient will receive this treatment as it could be denying patients access to a therapy that may be of benefit to them

A

Sunshine and Taube (2015)

53
Q
  • reported that CD8+ cell density in metastatic melanoma, as measured by IHC at the invasive margin of a tumour, as opposed to central infiltration, is most strongly associated with anti-PD1 response.
  • But there are limitations to IHC:
    • Not that sensitive
    • Background staining occurs 🡪 binding to non-specific reactive sites
    • Difficult to quantify protein
    • Not well standardised – different people use different protocols, different antibodies
    • Yes it’s cheap + pretty robust but it definitely has disadvantages!
A

Tumeh et al. (2014)

54
Q
  • Selective targeting of infiltrating MDSCs with PI3Ky inhibitors (PI3Ky = highly expressed in myeloid cells) was shown to restore sensitivity to anti-PD1 and anti-CTLA4 in murine models, and this targeting promoted cytotoxic T cell-mediated tumour regression
  • Could localising and phenotyping immune cells in the TME be used as a biomarker?
  • Could we use a SMI against PI3Ky to overcome resistance in pts with high levels of MSDCs in tumours?
A
  • De Henau et al (2016)
55
Q
  • Looked at stool samples from 42 patients with metastatic melanoma pre ICI treatment
  • Sequence 16 ribosomal subunit which has different expression among different bacteria
  • found that the ratio of response-associated (‘good’) to resistance-associated (‘bad’) bacteria was able to clearly stratify human responders from non-responders.
  • Also find that studies showing antibiotics prior to ICB is associated with adverse outcome
    • Sceptical of this because the need for antibiotics suggests a pt is frail and so on
  • Lit review in Oct 2016 showed 36/38 human microbiotas studies were successful – not publishing negative data
  • What is the mechanism underlying this – is it causal or correlative?
  • These results implicate the gut microbiota as an influential factor in antitumour immunity and ICI response.
A
  • Matson et al. (2018)
56
Q
  • Investigating REFN3767 – a mAb that targets LAG-3
  • This group have published results from an ongoing phase I trial of this compound
  • A total of 67 patients (25 in the monotherapy cohort and 42 in the combination group with an anti-PD-1 mAb)
  • 11/25 in the monotherapy and 6/42 in combination achieved stable disease
  • Trial is still open and recruiting

Which is better – monotherapy or combination? Which agent is best to combine with? Does combination lead to more adverse effects? Many unanswered questions

A
  • Papadopoulos et al 2019
57
Q
  • Phase 1b clinical trial
  • Used an oncolytic virus
    • Was based on HSV but mutated in such a way that it cant infect normal cells, but could in tumour cells – injected into tumour mass
  • 21 pts with advanced melanoma given onco virus in comb with pembrolizumab
  • Generally was well tolerated
  • Pts had 62% overall response rate (with increased CD8 T cells, elevated PD-L1 protein expression)
  • Cav – not controlled, small, you need an tumour mass that’s accessible to inject
A
  • Ribas et al 2017
58
Q
  • recently identified that a higher neutrophil-to-lymphocyte ratio is significantly associated with poorer overall and progression free survival after ICI therapy – can achieve a high OR (3.22) when combined with tumour mutational burden
    • this is important as easy to measure – wouldn’t require extensive genome / biopsy, that takes time (just a peripheral blood sample)
A

Valero, 2021