Abstract
Association with hypomethylating agents is a promising strategy to
improve the efficacy of immune checkpoint inhibitors-based therapy. The
NIBIT-M4 was a phase Ib, dose-escalation trial in patients with
advanced melanoma of the hypomethylating agent guadecitabine combined
with the anti-CTLA-4 antibody ipilimumab that followed a traditional
3 + 3 design ([60]NCT02608437). Patients received guadecitabine 30, 45
or 60 mg/m^2/day subcutaneously on days 1 to 5 every 3 weeks starting
on week 0 for a total of four cycles, and ipilimumab 3 mg/kg
intravenously starting on day 1 of week 1 every 3 weeks for a total of
four cycles. Primary outcomes of safety, tolerability, and maximum
tolerated dose of treatment were previously reported. Here we report
the 5-year clinical outcome for the secondary endpoints of overall
survival, progression free survival, and duration of response, and an
exploratory integrated multi-omics analysis on pre- and on-treatment
tumor biopsies. With a minimum follow-up of 45 months, the 5-year
overall survival rate was 28.9% and the median duration of response was
20.6 months. Re-expression of immuno-modulatory endogenous retroviruses
and of other repetitive elements, and a mechanistic signature of
guadecitabine are associated with response. Integration of a genetic
immunoediting index with an adaptive immunity signature stratifies
patients/lesions into four distinct subsets and discriminates 5-year
overall survival and progression free survival. These results suggest
that coupling genetic immunoediting with activation of adaptive
immunity is a relevant requisite for achieving long term clinical
benefit by epigenetic immunomodulation in advanced melanoma patients.
Subject terms: Melanoma, Melanoma, Cancer immunotherapy
__________________________________________________________________
The NIBIT-M4 trial was designed to assess the safety, biological and
clinical activity of anti-CTLA4 ipilimumab with the DNA hypomethylating
agent guadecitabine in advanced melanoma patients. Here the authors
report the five-year follow-up results of the trial and an integrated
multi-omics analysis of pre- and on-treatment tumor biopsies.
Introduction
Immune checkpoint inhibitors (ICI) are drugs targeting regulatory
pathways in T cells to enhance antitumor immune responses^[61]1.
Treatment with ICI has dramatically improved the clinical outcome of
patients with tumors of different histotypes^[62]2, including
melanoma^[63]3, and lung cancer^[64]4. However, the percentage of
subjects who benefit from ICI therapy is still low, and novel
therapeutic strategies are eagerly awaited to fully exploit their
clinical potential. Indeed, even in the most responsive tumor types,
both intrinsic^[65]5 and acquired resistance^[66]6,[67]7 limit the
efficacy of ICI therapy. The cellular and molecular characterization of
human tumor samples by high-throughput and deep phenotyping approaches
define the role of the immune microenvironment in driving the prognosis
of cancer patients and their responsiveness to ICI
therapies^[68]8,[69]9.
In this scenario, an active area of biomedical research aims to
identify combinatorial approaches that could improve even the early
phases of developing the antitumor response. Mechanistically, these new
immunotherapy regimens aim at achieving one or more of three main
effects thought to be crucial for overcoming resistance to immune
intervention: (i) fostering the cross-talk between innate and adaptive
arms of the immune system, (ii) promoting the recruitment of functional
T cells at the tumor site, and (iii) counteracting recruitment/function
of immunosuppressive cells.
Among promising agents that may play a role in ICI combinations
there are the hypomethylating agent (HMA) due to their immunomodulatory
activity on tumor cells^[70]10, the ability to activate innate immunity
pathways^[71]11,[72]12 and the pre-clinical evidence for enhanced
antitumor effects when combined with ICI^[73]13. In this scenario, our
Italian Network for Tumor Biotherapy (NIBIT) Foundation Phase Ib
NIBIT-M4 trial, based on the association of ipilimumab with the HMA
guadecitabine in advanced melanoma patients, showed significant tumor
immunomodulatory effects and preliminary evidence of promising clinical
activity^[74]11. More recently, by comparing transcriptional programs
elicited by different classes of epigenetic drugs in melanoma cells, we
found that the main biological activity of guadecitabine is the
promotion of gene expression and activation of master factors belonging
to innate immunity pathways, including Type I–III interferon (IFN),
NF-kB, and TLR^[75]10. These results corroborated the notion that the
rescue of adaptive immunity by ICI may cooperate with the promotion of
innate immunity by the HMA guadecitabine, thus potentially explaining
the clinical activity of the combination. Two additional recent trials,
combining guadecitabine with pembrolizumab in solid
tumors^[76]14,[77]15 have indeed confirmed the significant antitumor
activity of this combination in terms of clinical benefit rate (31.4%)
or of progression-free survival (PFS) rate >24 weeks (37%). Crucially,
in both studies, relevant immune effects were described regarding the
upregulation of innate and adaptive immunity pathways in post-treatment
samples.
Despite these initial promising clinical applications of the HMA
guadecitabine combined with ICI, the further development of these
combinatorial approaches requires the understanding of the biological
mechanisms underlying the response and resistance to this specific type
of epigenetic immunomodulation. The NIBIT-M4 study was a single-arm
trial, therefore, in principle, such design prevents the possibility to
achieve a sound disentanglement of the contribution of ipilimumab vs.
the contribution of guadecitabine to the overall clinical activity.
Here we show that an advanced integrative systems biology approach,
focusing on genomic, transcriptional, and methylation landscape
analysis of baseline and on-treatment tumor tissues can shed light on
the mechanisms of action of the two agents and on the resistance
mechanisms likely impacting the ICI vs. the demethylating agent. In
fact, first, we asked whether responder patients show differential
enrichment of genomic features vs. non-responder patients. In
particular, we analyze the effects of guadecitabine in the cases
harboring mutations in its target genes. We also explored the promotion
of expression of retroviral sequences and of repetitive elements that
represent the mechanistic signature of immunomodulation by
guadecitabine. We tested a combinatorial index for predicting long-term
clinical benefit by integrating transcriptional information on the
development of adaptive immunity, the Immunological Constant of
Rejection (ICR)^[78]16–[79]18 with a measure of genetic immunoediting
(GIE). The ICR signature incorporates IFN-stimulated genes driven by
transcription factors IRF1 and STAT1, with CCR5 and CXCR3 ligands,
immune effector molecules, and counter-activated immune regulatory
genes. A high expression of ICR genes typifies ‘hot’/immune active
tumors characterized by the presence of a T helper 1 (Th1)/cytotoxic
immune response and predicts survival and response to ICI in different
tumors including colon cancer^[80]17, breast^[81]19, bladder, stomach,
head and neck^[82]16, sarcoma^[83]20, and melanoma^[84]8. The GIE index
quantifies the amount of genetic immunoediting as the ratio between
observed and expected tumor neoantigens. Here we show that
stratification of NIBIT-M4 patients based on the ICR/GIE classification
predicts Overall Survival (OS) and PFS, a finding that was validated in
external larger ICI datasets. Collectively, our results contribute to
improve the understanding of response and resistance to epigenetic
immunomodulation, and provide a valuable multi-omics-related tool that
may be used for patient stratification and clinical outcome prediction
in immunotherapy cohorts.
Results
Long-term outcomes in the NIBIT-M4 trial
At data cutoff, July 1st 2022, with a minimum follow-up of 45 months, 6
(31%) of the 19 patients enrolled in the NIBIT-M4 study were alive. The
median OS was 25.6 months (95% CI, 0.0-52.9), while the median PFS was
5.2 months (95% CI, 4.0–6.4); the 5-year OS rate was 28.9% with a
5-year PFS rate of 5.3%; median Duration of Response (DoR) was 20.6
months (95% CI, 12.4–28.8). Eighteen patients (95%) were
treatment-naive at study entry and 1 (5%) had received PD-1 mAb as
first-line therapy. Three patients were in Complete Response (CR) and
off-study therapy, while 13/19 (68%) had received subsequent line(s) of
therapy, including immunotherapy, target therapy, and/or chemotherapy;
among those, 5 patients who had achieved a disease control (DC) had a
median time to the subsequent treatment of 18.9 months (range
10.3–39.0) (Fig. [85]1).
Fig. 1. Swimmer plot analysis of NIBIT-M4 patients.
[86]Fig. 1
[87]Open in a new tab
Swimmer plot showing by study arm patients who at the time of data
cutoff were alive and either still on study treatment or off-study
therapy, without having received subsequent therapy, and all patients
who have received subsequent treatment at the time of data cutoff,
regardless of whether alive or dead. Subsequent treatments include
immunotherapy (i.e., anti-PD-1 monotherapy or combinations, ICOS
agonist or vaccine), BRAFi + MEKi, and chemotherapy.
Genomic landscape: mutational profile differences in baseline and
on-treatment lesions in responder (R) vs. non-responder (NR) patients
Longitudinal multi-omics profiling, including whole exome sequencing
(WES), RNA Sequencing (RNASeq), and Reduced representation bisulfite
sequencing (RRBS), were performed on tumor biopsies collected at
baseline (week 0) and week 4 and week 12 on therapy from 14 patients
(Supplementary Fig [88]1). Matched normal tissue collected at baseline
was available for 8 patients. The exome sequencing profiling of our
cohort, performed using stringent filtering, showed a high consistency
of the somatic calls/mutations during treatment (Fig. [89]2,
Supplementary Data [90]1). Although tumor mutational burden (TMB) was
not significantly different in R vs. NR patients (16.9 vs. 15.3, p
value = 0.6, Student’s t test), several significant differences were
found at the single gene level. BRAF was slightly enriched in NR (p
value = 0.02, Chi-squared test). In contrast, NRAS mutation was
significantly more frequent in R vs. NR (50% vs. 0%, p value = 5.4e^−5,
Chi-squared test). ADAMDEC1, encoding a disintegrin metalloproteinase
associated with dendritic cell (DC) function, was altered only in
lesions from R patients. Mutations in genes belonging to the epithelial
to mesenchymal transition (EMT) pathway were enriched in NR (p
value = 0.01, Chi-squared test). The CDKN2A gene, frequently mutated in
melanoma (35% of patients in TCGA cohort), was more frequently altered
in NR patients. CDKN2A has been demonstrated to inhibit EMT and promote
cancer immunity and CDKN2A deletions have been associated with ETM in
different cancer types^[91]21. Three neuronal-related genes (PCLO,
PLXNA4, and EPHA7), and the gene encoding the leptin receptor (LEPR),
all reported as mutated in melanoma at a variable frequency (37%, 11%,
16% and 8% of samples in TCGA cohort, respectively), were altered more
frequently in R compared to NR patients. The male germline-specific
gene PLCZ1 was mutated only in lesions from R patients. Interestingly,
several of the mutated genes (BRAF, NRAS, CDKN2A, EPHA7, PLXNA4) have
been previously associated with response or resistance to ICI in
monotherapy^[92]22–[93]26 although for some of them (e.g. BRAF)
evidence is not conclusive^[94]27. The DNMT1 gene, encoding one of the
guadecitabine targets, was mutated in two NR patients, and one of the
mutations was a truncating event suggesting loss of function of the
DNMT1 gene product. An additional mutation of SETD2, involved in
chromatin organization, was observed in another NR patient. We analyzed
the effect of somatic mutations in DNMT1 or SETD2 on the methylation
during therapy. The increasing or decreasing trend was evaluated based
on the slope of the robust linear regression line between the three
time points. We analyzed all genomic regions, specifically the coding,
intergenic, intronic regions (Supplementary Fig [95]2a), and then
regulatory regions (Supplementary Fig [96]2b) in DNMT1- and
SETD2-mutant vs. wild-type lesions. Interestingly, DNMT1- or
SETD2-mutant samples did not show the decreasing pattern over time that
we observed in the wild-type lesions. This was also confirmed in long
terminal repeat (LTR) including endogenous retroviral elements (ERVs)
(Supplementary Fig [97]2c). ERVs are particularly important for the
immune response to tumors, even in the context of ICI^[98]28. Previous
studies have shown that by inducing the re-expression of ERV sequences,
demethylating agents can activate the viral mimicry response secondary
to intracellular recognition of viral dsRNA^[99]29,[100]30. This
response explains the promotion of type I IFN and innate immunity
pathways that characterize the guadecitabine-specific gene signature
recently defined by us in melanoma cells^[101]10. We confirmed that the
expression of LTR elements was inversely correlated with the expression
after treatment with demethylating agent in wild-type, but not in
mutant tumors (Supplementary Fig [102]2d).
Fig. 2. Genomic landscape of the NIBIT-M4 trial.
[103]Fig. 2
[104]Open in a new tab
Oncoplot of frequent somatic nonsynonymous and copy number alterations
of NIBIT-M4 trial organized by response (columns, non-responder (NR,
n = 8) and responder (R, n = 6) patients) and pathways (rows). Tumor
Mutation Burden (TMB), dose (30, 45, and 60 mg/m^2/day), and time (week
0, week 4, week 12) of treatments are indicated. The proportion of
alterations in NR and R groups is visualized for each gene (p value of
two-sided Pearson’s chi-squared test statistic, *p < 0.05, **p < 0.01,
***p < 0.001). Source data are available in Supplementary Data [105]1.
Solar ultraviolet (UV) radiation is one the main etiological factor for
skin cancer, including melanoma as it causes a characteristic genomic
mutational pattern associated with elevated TMB via the formation of
pyrimidine-pyrimidine photodimers (COSMIC signature 7)^[106]31,[107]32.
We performed mutational signature deconvolution on the cases for which
the matched normal was available (n = 8). Two (SBS7a and SBS7b) of the
four UV-associated mutational signatures were the most frequently
observed (Supplementary Fig [108]3). However, the limited number of
cases did not allow for detecting any significant association between
UV mutational signature rate and response (p value = 0.064, Chi-squared
test), mutation load (p value = 0.435, Chi-squared test) and neoantigen
load (p value = 0.122, Chi-squared test), respectively.
Overall, our longitudinal analysis confirmed several genomic features
previously associated with response to ICI, such as defects in the EMT,
mutations of BRAF and NRAS and in other immune-related genes, and was
useful to discover, even for a limited number of cases, that
loss-of-function mutations in chromatin organization and guadecitabine
targets may contribute to limit the efficacy of the combined therapy
and the epigenetic immune-modulatory effect of the HMA.
Transcriptional landscape of baseline and on-treatment tumor lesions:
distinct and evolving transcriptional programs distinguish R from NR patients
RNA-sequencing data from the NIBIT-M4 were used to carry out
differential gene expression analysis between R and NR patients at
different time points of treatment (Supplementary Data [109]2). This
analysis showed a progressive enrichment from baseline to week 12 in
Gene Ontology Biological Processes (GO: BP) categories related to
immune processes in R compared to NR patients (Fig. [110]3a and
Supplementary Data [111]3). In contrast, in lesions from NR patients, a
progressive increase from baseline to week 12 was found for GO terms
related to adhesion, cell cycle, metabolism, and skin developmental
processes. We tested several state-of-the-art predictive signatures of
response to ICI, including MIRACLE score^[112]8, ICR^[113]19
IMPRES^[114]33, TIDE^[115]34, and MPS^[116]35. None of these five
scores discriminated against R from NR patients when considering either
baseline and on-treatment samples or at various time points
(Supplementary Fig [117]4a and Supplementary Data [118]4) neither using
standard tests for comparing the difference between multiple subjects
and repeated measures.
Fig. 3. Transcriptional landscape of the NIBIT-M4 trial.
[119]Fig. 3
[120]Open in a new tab
a Gene Set Enrichment Analysis (GSEA, as implemented in
clusterProfiler^[121]84) from supervised differential analysis between
R (n = 6) and NR (n = 8) patients (negative binomial generalized linear
model with likelihood ratio test (glmLRT), as implemented in
EdgeR^[122]82) before treatment (week 0) and after four (week 4) and
twelve (week 12) weeks. x axis reports the aggregated p value of
significant enriched GO:BP terms (false discovery rate method from
empirical permutation test, FDR < 0.1), computed using Fisher method
and classified into seven main categories. Size of the dot represents
the number of GO:BP terms grouped into a category; color of the dots
represents the mean Normalized Enrichment Score (NES) of the terms. b
Volcano plot (left) of differentially expressed genes (p value < 0.05
from glmLRT, as implemented in EdgeR^[123]82) between R (n = 6) and NR
(n = 8) patients, labeled genes belong to the guadecitabine-specific
gene signature^[124]10. x axis reports the effect size (in log scale),
y axis reports the −log(p value) from glmLRT, as implemented in
EdgeR^[125]82. GSEA enrichment (right) of the guadecitabine-specific
signature on the ranked list of differentially expressed gene at week 4
and week 12. c Same as in b using the dataset from a trial of combined
therapy ICI plus HMA from Chen et al. 2022^[126]14 (C2D8
post-treatment, n = 4 R and n = 5 NR). Source data are provided as a
Source Data file.
We have recently performed a comparative profiling of gene signatures
induced by different classes of epigenetic drugs in melanoma cell
lines^[127]10 and found that guadecitabine activates several innate
immunity pathways by induction of a signature of 166 genes. To better
disentangle the effect of the HMA in our cohort, we performed gene set
enrichment analysis of the guadecitabine-specific signature on the list
of differentially expressed genes between R and NR (Fig. [128]3b). We
found that this signature was significantly activated in R at week 4 (p
value = 4.172e^−05, GSEA permutation test) and week 12 (p
value = 1.055e^−04, GSEA permutation test). To further validate this
finding, we used the data from another trial in which patients with
ovarian cancer were treated with a combination of ICI and epigenetic
drug^[129]14. We observed a significant activation of the guadecitabine
signature after treatment in R patients (Fig. [130]3c). Our results
confirm the main immune-modulatory effect of HMA and that this effect
is associated with response. By a custom-designed NanoString assay we
then explored differential expression in R vs. NR lesions of 20
published immune-related signatures (Supplementary Fig [131]4b)
providing information on B-cell content and
differentiation^[132]34,[133]36, tertiary lymphoid structures (TLS)
formation^[134]37,[135]38, follicular T helper (TFH)
cells^[136]34,[137]36, T-cell exhaustion (TEX) subsets^[138]39,
tumor-associated endothelial cells^[139]40, immune checkpoint blockade
(ICB) response^[140]41,[141]42, and the recently identified
guadecitabine-specific signature genes induced by this demethylating
agent in melanoma cell lines^[142]10. The large majority of these
signatures was selectively enriched, considering all time points, in
tumor biopsies from R compared to NR patients. These results were
consistent with preferential development in R lesions of a coordinated
T- and B-cell mediated immune response involving TLS and TFH cells,
with enhanced expression of IFN-ɣ-induced genes crucial for ICB
response and with increased presence of CD8^+ T cells at different
stages of exhaustion.
We also used Ingenuity Pathway Analysis (IPA) to identify canonical
pathways differentially regulated in R vs. NR patients during
treatment. Indeed, IPA database accounts for positive and negative
associations of genes and pathways. This analysis predicted significant
activation in R patients of several immune-related pathways including
the “Pathogen induced cytokine storm”, the “TH1” and “TH2”, the
“phagosome formation” and the “cross-talk between DC and Natural Killer
(NK) cells” pathways (Supplementary Fig [143]4c). In contrast, pathways
predicted to be inhibited in R vs. NR patients included the “PD-1,
PD-L1 cancer immunotherapy”, the “MSP-RON signaling in macrophages” and
the “GP6 signaling” pathways. These results confirm that R patients
experience the activation of immune pathways crucial for developing
innate and adaptive immunity.
Finally, we analyzed the differential expression between R and NR
patients in selected gene sets (Supplementary Fig [144]5). Lesions from
R patients showed a progressive increase of expression, from baseline
to week 12, of genes encoding for molecules controlling T-cell
activation, inhibitory receptors and ligands, chemokines, and
components of the immunoproteasome. In line with our previous reports
where we have shown that DNA methyltransferase inhibitors can
upregulate the expression of major histocompatibility complexes (MHC)
proteins^[145]43, we observed that HLA class I and class II were
significantly upregulated in R patients.
By contrast, lesions from NR patients showed higher expression of cell
cycle-, EMT- and skin development-related genes with a consistent
pattern across the weeks. Several of the EMT-related genes with higher
expression in NR patients encoded for molecules controlling adhesion
(ITGB3, VCAM1, collagens, and others), interaction with extracellular
matrix (VEGFA, MMP14, WNT5A, LAMA1, and others), and melanoma
de-differentiation (KRT9, KRT10, EGFR, and others). In particular,
WNT5A is a well-defined feature of a poor melanoma phenotype and has
been associated with a negative modulation of the tumor
microenvironment^[146]44. Lesions from NR patients also showed
significantly higher expression, compared to R patients, in several key
genes controlling cell proliferation including cyclin-dependent kinases
1 and 4, cyclins B1 and B2, mitotic checkpoint serine/threonine kinase
B, and other transcription factors controlling cell cycle such as E2F1
and E2F2.
The composition of the tumor microenvironment of our cohort was
deconvolved from transcriptome profiling data using eight immune- and
two stromal-cell signatures with MCP-counter^[147]45 (Fig. [148]4a). We
found a significantly higher abundance in R vs. NR of CD8^+ T cells (p
value = 0.042, Wilcoxon test) at week 4 (Fig. [149]4a), other
comparisons did not produce significant differences also due to the low
number of cases. The estimated increased abundance of CD8^+ T cells in
R lesions was in agreement with available quantitative
immunohistochemistry (IHC) data (r = 0.79, p value = 1e^−07, N = 11,
Spearman correlation test) for this immune subset (Fig. [150]4b) and
with enhanced CD8^+ intratumoral T cells in R lesions (p value = 0.002,
Student’s t-Test) (Fig. [151]4c).
Fig. 4. Immune microenvironment.
[152]Fig. 4
[153]Open in a new tab
a Immune microenvironment deconvolution of immune cell fractions
stratified by time point and response (n = 6 R and n = 8 NR). b IHC
validation (x axis) of the deconvolution of CD8^+ T-cell proportion
estimated from RNASeq (y axis) (Spearman correlation coefficients rho
(r) and associated p values from two-tailed correlation test are
provided for R and NR groups, samples from n = 15 R and n = 16 NR). c
Density of CD8 T cells by location from IHC in the tumor core (samples
from n = 15 R and n = 16 NR), and peritumoral (samples from n = 9 R and
n = 13 NR) (p value from two-sided Student’s t test between R and NR
groups). d Scatterplot between the T-cell receptor clonality (B locus)
and CD8^+ T-cell (left) and NK cell abundances (right) (Pearson’s
correlation coefficient (r)) and associated p values from two-tailed
correlation test are provided for R (n = 6) and NR (n = 8) groups). a,
c Box plots show the median as center, the lower and upper hinges that
correspond to the 25th and the 75th percentile, and whiskers that
extend to the smallest and largest value no >1.5*IQR. Values that stray
more than 1.5*IQR upwards or downwards from the whiskers are considered
potential outliers and represented with dots. b, d Bands represent
confidence intervals (±0.95) around a linear model fitted by robust
regression using an M estimator. Source data are provided as a Source
Data file.
We then used the clonality of V(D)J rearrangements within the TCR Beta
locus (TRB) as a proxy for estimating T-cell expansion during therapy.
TRB clonality was significantly correlated with the estimated
abundances of CD8^+ T cells (r = 0.83, p value < 0.0001, Spearman
correlation test) and NK (r = 0.78, p value < 0.001, Spearman
correlation test) cells in R rather than NR patients (Fig. [154]4d).
Taken together, the gene expression landscape of NIBIT-M4 lesions
indicated that distinct and evolving transcriptional profiles
characterized baseline and on-treatment tumor biopsies from R compared
to NR patients. Lesions from R patients showed progressive enrichment
for signatures and gene sets revealing activation of adaptive immunity
and effective immunomodulation by guadecitabine with a preferential and
clonal activation of T-cell subpopulation and NK cell in the tumor
microenvironment. Lesions from NR patients revealed lack of promotion
of immunity in a tumor transcriptional background dominated by
proliferation and EMT processes.
Integrative analysis of methylation and transcriptomic profiles during
treatment
The availability of the longitudinal sampling of tumor biopsies gave us
the opportunity to evaluate the immune-modulatory effect of the HMA
during therapy. Although NIBIT-M4 trial was not designed to compare the
effect of the combination HMA plus ICI vs. ICI alone, we could evaluate
the changes of the methylation pattern induced by the treatment with
the HMA. When we considered the overall methylation level across the
genome, a trend towards global demethylation was observed for both R
and NR patients (Supplementary Fig [155]6a) as well as in specific
genomic regions, the effect of the HMA in the coding part of the genome
(exons and intergenic regions) had a decreasing trend in R and NR
patients. Similarly, for promoters, UTRs regions, and other regulatory
regions such as enhancers and super-enhancers (Supplementary
Fig [156]6a) NR had a lower decreasing trend than R patients. We
evaluated the specific methylation pattern during therapy in Long
interspersed nuclear elements (LINEs), Short interspersed nuclear
elements (SINEs), and LTR. The effect of HMA was a general decreasing
trend of methylation for both R and NR patients (Fig. [157]5a). The
relationship of this demethylation process with the changes in
expression of these repeated element of the genome was also considered.
We correlated the expression and methylation of different LINE, SINE,
and LTR elements including ERVs. The number of reads that mapped on
these elements was used as a proxy of their expression (see Methods).
This analysis showed a significant anti-correlation between methylation
and expression only for R patients, whereas for NR patients a similar
inverse trend was not observed (Fig. [158]5b).
Fig. 5. Evolution of the overall methylation pattern in various genomic
regions and regulatory regions as function of time between R and NR patients.
[159]Fig. 5
[160]Open in a new tab
a Variation of the overall methylation pattern in Long interspersed
nuclear elements (LINEs), Short interspersed nuclear elements (SINEs)
and Long terminal repeat (LTR) endogenous retroviral elements (ERV).
The increasing or decreasing trend was evaluated based on the
inclination of the robust linear regression line between the three time
points. Plots report the values at different time points divided
between R (n = 6) and NR (n = 8). Box plots show the median as center,
the lower and upper hinges that correspond to the 25th and the 75th
percentile, and whiskers that extend to the smallest and largest value
no more than 1,5*IQR. Values that stray more than 1.5*IQR upwards or
downwards from the whiskers are considered potential outliers and
represented with dots. b Scatterplot between methylation and expression
in R and NR patients in SINE, LINE and LTR elements (Pearson’s
correlation coefficient (r)) and associated p values from two-tailed
correlation test are provided for R (n = 6) and NR (n = 8) groups). a,
b Bands represent confidence intervals (±0.95) around a linear model
fitted by robust regression using an M estimator. Source data are
provided as a Source Data file.
Finally, we performed differential promoter methylation and gene
expression analysis between R and NR patients (Supplementary
Fig [161]6b). We used SMITE^[162]46 to rank the upregulated and
hypomethylated genes as well as the downregulated and hypermethylated
genes between R and NR patients at different time points. Consistently
with the previous observations, functional enrichment of hypomethylated
and upregulated genes between R and NR patients resulted in biological
processes associated with immune functions at week 12. Genes enriching
immune response included granzymes (GZMM), interleukins and interleukin
receptors (IL12, IL32, IL15, IL12RB1), cytokines and cytokine receptors
(TNF, CXCR4, TNFRSF1B) and HLA genes and trans-activators (CIITA,
HLA-DOA, HLA-DMB, HLA-E).
We also interrogated the effect of the epigenetic agent in the set of
lesions from NR patients where we did not find evidence of promotion of
immunity as in the lesions from R patients. To answer this question, we
jointly compared the expression and methylation profiles of NR patients
at different time points. We applied SMITE to the differential
expression and methylation analysis between week 12 and the baseline in
NR patients. This analysis aimed at uncovering the pathways that were
activated on-treatment in NR patients. The top 100 genes of the ranked
list were used for functional enrichment. Some of these genes were
significantly associated with fate determination (WNT5A), cell
migration (PRKD2), epithelial differentiation (KRT15), while most
frequent GO:BP terms included pathways related to “development”,
“differentiation” and “migration/motility” (Supplementary Fig [163]7a).
On the contrary, after treatment, the same longitudinal analysis of
lesions from R patients resulted in the activation of immune response
functions such as regulation of T and NK cell activation and
proliferation (IL15), Th1 polarized T cells (TBX21), and other
functional categories associated with lymphocyte activation
(Supplementary Fig [164]7b).
Overall, our results showed that treatment with HMA induces the
demethylation of genomic regions, in particular transposable elements,
and ERVs. The effect of these epigenetic changes was associated with
their higher expression in R patients. Moreover, intersection of
differentially expressed and methylated genes showed specific
epigenetic activation of immune functions in R patients.
The ICR/GIE classification contributes to explain response, resistance
through immune escape and long-term clinical outcome
TMB and neoantigen loads were highly correlated (r = 0.77, p value
5e^−09, Spearman correlation test) in the lesions of the NIBIT-M4
trial, however none of these two parameters discriminated against R
from NR patients.
According to the immunoediting theory, tumor clones can be eliminated
by CD8^+ T cells, resulting into a depletion of tumor-associated
antigens, including neoantigens^[165]47. Immune-edited lesions are
therefore expected to have less neoantigens then expected, while tumors
that are not able to elicitate an antitumor immune response will not
display sign of immunoediting.
We then tested the hypothesis that the genetic immunoediting (GIE)
score^[166]48,[167]49, an index that integrates both TMB and neoantigen
load information in a single measure of the extent of immunoediting,
could show an association with clinical response.
The GIE value was calculated as the ratio between the observed vs.
expected number of neoantigens in each tumor sample (Supplementary
Data [168]5). The expected number of neoantigens was estimated by
training a linear model having TMB as the independent variable and
neoantigen load as the outcome variable. Tumors with a number of
neoantigens lower than expected (i.e., lower GIE values) are thought to
display evidence of immunoediting, whereas a higher frequency of
neoantigens than expected indicates a lack of immunoediting (Non-GIE).
However, the difference was not significant at the baseline, possibly
due to the low number of cases (Supplementary Fig [169]8a). We then
implemented a more precise metric capturing the extent of
immunoediting, by combing both information of GIE and the presence of a
robust intratumoral cytotoxic immune response (ICR)^[170]17. We then
speculated that the combined presence of an adaptive cytotoxic immune
response (high ICR) and the evidence of neoantigen depletion (GIE),
would more precisely capture “truly” immune-edited lesions. We then
stratified tumor lesions from patients enrolled in the NIBIT-M4 study
based on GIE score greater or lower than one (coded respectively as
“Non-GIE” and “GIE”) and ICR score greater or lower than zero, yielding
four groups: High-ICR/GIE, High-ICR/Non-GIE, Low-ICR/GIE,
Low-ICR/Non-GIE (Fig. [171]6a). The tumor samples belonging to R
patients were highly enriched in the High-ICR/GIE group (61%, p
value = 4.2e^−04, Chi-squared test). The ICR/GIE classification was
significant even when limiting the analysis on baseline lesions (67%, p
value = 2.1e^−02, Chi-squared test) with four of the five lesions in
the High-ICR/GIE group (Supplementary Fig [172]8b). To shed light on
the mechanism that differentiates GIE in the presence of adaptive
immunity captured by ICR, we then performed a supervised transcriptome
analysis comparing the High-ICR/GIE vs. the High-ICR/Non-GIE groups
(Fig. [173]6b). This analysis showed that the High-ICR/GIE group (i.e.,
the group in the upper left quadrant, Fig. [174]6a) was characterized
by enhanced representation of several Biological Processes related to
immune response including antigen processing and differentiation
(Fig. [175]6b). This also suggested that the “High-ICR/Non-GIE” group
(upper right quadrant in Fig. [176]6a) could be defective for
expression of antigen processing and presentation genes in a way that
could explain the lack of immunoediting. In agreement, lesions from the
High-ICR/Non-GIE group showed loss of expression of HLA class I
antigens on tumor cells despite the presence of CD8^+ T cells
infiltrating the tumor core (Fig. [177]7 and Supplementary
Fig [178]9a).
Fig. 6. Biomarkers of immunoediting (1).
[179]Fig. 6
[180]Open in a new tab
a Scatterplot of ICR score by Genetic Immunoediting (GIE) score for R
(n = 18) and NR (n = 23) samples (left) and their proportion (right)
after classification as ICR/GIE classes (p value from two-sided
Pearson’s chi-squared test statistic). b Barplot of most significantly
(FDR < 0.01) enriched GO:BP terms from GSEA analysis of High-ICR/GIE
(n = 15) vs. High-ICR/Non-GIE (n = 9) samples’ comparison. c
Kaplan–Meier for OS by patients classified as High-ICR/GIE (n = 5) or
High-ICR/Non-GIE (n = 4) at week 12 (top) and R (n = 6) or NR (n = 8)
(bottom). Time is indicated in months and censor points are indicated
by vertical lines. p values are calculated by log-rank test. Source
data are provided as a Source Data file.
Fig. 7. Biomarkers of immunoediting (2).
[181]Fig. 7
[182]Open in a new tab
Microphotographs of HLA class I and CD8 immunohistochemistry for
representative patients in each ICR/GIE class (left). The plots
represent the ICR and GIE sample scores, percentages of HLA-positive
cells, and density of CD8 T cells. The presented data was derived from
one experimental run independently reviewed by three different trained
scientists. Source data are provided as a Source Data file.
We then asked whether the patient groups defined by the ICR/GIE
classification also experienced different long-term clinical outcomes.
By stratifying patients according to the ICR/GIE classification of week
12 biopsies we found a slightly significant difference (p
value = 0.035, log-rank test) in OS and in PFS (p value = 0.022,
log-rank test) between the High-ICR/GIE and the High-ICR/Non-GIE group
(Fig. [183]6c and Supplementary Fig [184]9b). The ICR/GIE
stratification remained significant for OS and PFS even when taking
into consideration all four subsets (Supplementary Fig [185]9b). In
contrast, patients’ classification by response groups was not
associated with OS (Fig. [186]6c), although it was associated with PFS
(Supplementary Fig [187]9b).
Comprehensive assessment of the ICR, GIE, CD8^+ T cell and HLA Class I
scores, available from lesions of 11 patients (Supplementary
Fig [188]10a), indicated that the overall profile for these four
parameters was consistent with the observed clinical response in all
but one patient (#11). Patient #11 had a Low-ICR/Non-GIE score, but was
classified as R patient due to stable disease (SD) in target lesions.
Eventually, the patient underwent disease progression in target lesions
and developed several new lesions (Supplementary Fig [189]10b).
Collectively, these findings provided a mechanistic explanation for the
genesis of the High-ICR/Non-GIE subset.
Validation of the ICR/GIE classifier in other response datasets
To validate the ICR/GIE stratification, we assembled a cohort of 83
melanoma cases treated with ICI, either anti-CTLA4 or -PD-1, from
previous published studies^[190]5,[191]50,[192]51 for which TMB,
neoantigen load and gene expression were available. The stratification
of patients into the 4 ICR/GIE subsets confirmed the enrichment of R
patients in the High-ICR/GIE group (Fig. [193]8a). Significant
differences in OS between patients characterized as High-ICR/GIE vs.
those coded as High-ICR/Non-GIE were observed even in this validation
cohort (Fig. [194]8b). Interestingly, differential pathway analysis in
the High-ICR/GIE vs. High-ICR/Non-GIE subsets identified the Biological
Process “antigen processing and differentiation” as selectively
enriched in the High-ICR/GIE subsets as we found in the NIBIT-M4 cohort
(Fig. [195]8c). These results suggest that the same mechanism uncovered
in the NIBIT-M4 cohort could explain the High-ICR/Non-GIE subset even
in the validation cohort: a defective antigen processing and
presentation pathway may contribute to suppress genetic immunoediting
even when lesions have a high ICR profile, and this may be a general
phenomenon irrespective of the type of immunotherapy that is being
used.
Fig. 8. Validation of the ICR/GIE score in patients from other cohorts.
[196]Fig. 8
[197]Open in a new tab
a Scatterplot of ICR score by Genetic Immunoediting (GIE) score for R
(n = 34) and NR (n = 49) samples (left) and their proportion (right)
after classification as ICR/GIE classes (p value from two-sided
Pearson’s chi-squared test statistic). b Kaplan–Meier for OS by
patients classified as High ICR/GIE (n = 24) or High-ICR/Non-GIE
(n = 14). Time is indicated in months and censor points are indicated
by vertical lines. p value is calculated by log-rank test. c Barplot of
most significantly (FDR < 0.01) enriched GO:BP terms from GSEA analysis
of High-ICR/GIE (n = 24) vs. HighICR/Non-GIE (n = 14) comparison.
Source data are provided as a Source Data file.
Collectively, these results suggested that effective coupling of tumor
immunoediting with activation of adaptive immunity can promote response
and improved clinical outcome in the NIBIT-M4 epigenetic
immunomodulation trial. In contrast, defective development of adaptive
immunity or lack of genetic immunoediting, associated with immune
escape mechanisms, may favor resistance and less favorable long-term
clinical outcome.
Discussion
The NIBIT-M4 trial has been the first Phase Ib epigenetic
immunomodulation study testing the association of the HMA guadecitabine
with ICI in solid malignancies. Treatment of metastatic melanoma
patients with guadecitabine combined with ipilimumab was found to be
safe, feasible, and tolerable, with initial signs of clinical and
immunologic activity^[198]11. The 5-year survival rate, duration of
response, and time to subsequent treatment in patients who achieved a
DC we report here are intriguing and clinically meaningful. These
findings compare favorably with the efficacy of anti-CTLA-4 monotherapy
in metastatic melanoma patients^[199]52, though with the limitations of
interstudy comparisons that need to be interpreted with caution and to
be placed in context. Nevertheless, the long-term follow-up of the
NIBIT-M4 trial seems to support the clinical potential of guadecitabine
combined with anti-CTLA-4 therapy, though the relative contribution of
each agent and the potential of guadecitabine maintenance therapy in
the clinical results we observed could not be fully dissected in this
trial. In this scenario, our ongoing randomized phase II NIBIT-ML1
study ([200]NCT04250246) in PD-1/-L1-resistant melanoma and non-small
cell lung cancer patients will help to address the clinical and
immunobiologic contribution of the addition of the HMA
decitabine/cedazuridine (ASTX727) to ICI therapy. However, further
support for the notion that guadecitabine is a promising way forward to
improve the efficacy of ICI therapy in solid tumors derives from two
most recently published trials in platinum-resistant ovarian
cancer^[201]14, and in different tumor types^[202]15. Indeed, the
combination of guadecitabine with the anti-PD-1 pembrolizumab led to
encouraging response rates, immunomodulation in the tumor tissue and/or
in periphery, and evidence of demethylation in on-treatment lesions,
with manageable toxicity^[203]18.
Our aim here was to exploit the longitudinal multi-omics profiling of
the NIBIT-M4 cohort in conjunction with the 5 years follow-up to shed
light on the effect of the combination during treatment and to identify
early biomarkers of response. To this end, we have first individually
interrogated the available omics platforms taking advantage of the
accurate longitudinal sampling. Then we developed computational
multi-omics integration approaches to evaluate the effect of the
adopted demethylation agent on boosting the adaptive and innate
immune-mediated cancer rejection. The five-year follow-up showed that
our multi-omics classification based on the ICR/GIE index is a better
prognostic factor than best overall response (BOR). Therefore, the
analysis reported here can serve as a guide for improved patient
stratification and selection strategies in combination therapies
involving ICI and immunomodulatory agents, including HMA.
Longitudinal WES and transcriptomic analysis contributed to shed light
on molecular factors impacting clinical response and on long-term
outcome after guadecitabine plus ipilimumab. At the mutational profile
level, strong consistency across biopsies obtained at three-time points
in a 12-week time frame allowed us to identify significant associations
of somatic mutations with response/resistance even with the small
number of patients enrolled in the NIBIT-M4 trial.
Even if the impact of the genomic landscape is difficult to be linked
to the HMA rather than to ipilimumab, our results confirm and extend
evidence of resistance obtained in ICB-only trials. First, we found
that BRAF mutations are more frequent in NR compared to R patients and
the opposite was true for NRAS mutations. Patients with NRAS
mutation-positive melanomas have improved PFS after ICB with
anti-CTLA-4 and -PD-1 antibodies^[204]22 and improved OS when treated
with ipilimumab^[205]23. Concerning BRAF mutations, there are
conflicting reports in the literature^[206]27. Some meta-analyses did
not evidence differences in response studies between BRAF wild-type and
mutation patients^[207]53. Other studies reported a negative
association between BRAF status and OS after ipilimumab in
melanoma^[208]54. Thus, the associations that we found with BRAF and
NRAS mutational status, with the caveat of the limited sample size in
our trial, appear consistent with evidence obtained in clinical trials
of ICB-only. These data thus suggest that the associations of BRAF/NRAS
mutations with clinical response may reflect their impact on the ICI,
rather than their relevance for the mechanism of action of the
demethylating agent. We found the CDKN2A mutations to be more frequent
in NR patients. CDKN2A alterations have been previously found
associated with reduced benefit from ICB in urothelial carcinoma, but
not in melanoma^[209]24. Immunotherapy resistance of CDNKA deficient
tumors has also been reported in pan-cancer studies^[210]55. EPHA7
mutations, which we found more frequently in R patients, have been
associated with better clinical outcomes in ICB-treated patients across
multiple cancer types^[211]25. PLXNA4 mutations, more frequent in R
patients, have been shown to promote Cytotoxic T-lymphocyte
infiltration in pre-clinical tumor models, as PLXNA4 behaves as a
negative checkpoint regulating T-cell migration and
proliferation^[212]26.
Mutations in genes belonging to the EMT were enriched in NR patients.
EMT is associated with a less favorable outcome in patients with
cancer^[213]56; it stimulates angiogenesis and is a tumor-intrinsic
mechanism enhancing immunosuppression^[214]57,[215]58. Recent studies
reporting the genomic and transcriptomic features associated with ICI
in melanoma have shown higher expression of several EMT genes in NR
subjects^[216]59. Our analysis indicates that genomic alterations can
drive these differences. We also observed that two NR patients harbored
mutations in the gene DNMT1, which is the direct target of
guadecitabine. We described the reduced immune-modulatory effect of the
demethylating agent in patients harboring defects. In summary,
available evidence indicates that most differentially enriched
mutations between R and NR groups have also been associated with
response or resistance to ICB-only regimens. In other words, the
genomic landscape of the lesions may contribute to explaining the
efficacy or lack of efficacy of the ipilimumab arm of the trial.
Interrogating the longitudinal gene expression data was also useful in
evaluating how the immune context evolves during therapy. The
differential activity of these pathways tends to increase during
therapy, suggesting that immune surveillance promoted by ICI represses
cell cycle genes together with differentiation pathways. We have shown
an increased expression of the guadecitabine-specific signature that we
had previously defined^[217]10 in on-treatment lesions from R compared
to NR patients. This result suggests that clinical benefit requires
susceptibility to the immunomodulatory action of this DNMT inhibitor. A
consequence of this hypothesis is that NR patients must have some
resistance mechanism to guadecitabine, and the DNMT1 mutations found in
two NR patients may be part of this resistance mechanism.
The transcriptional effects that could be ascribed to guadecitabine and
could contribute to explain clinical benefit in this combinatorial
trial, were assessed by considering tumor-specific antigens derived
from transposable elements^[218]60 and ERV sequences^[219]28 for the
immune response to tumors. Both these classes of sequences are known to
be regulated by methylation and susceptible to re-activation by
demethylating agents. We confirmed that clinical benefit in the
NIBIT-M4 trial could depend on the ability of guadecitabine to
re-activate these sequences, by promoting their demethylation, leading
to enhanced expression in R compared to NR patients.
We evaluated several transcription-based signatures for response
prediction to ICI. No difference was observed at the baseline. This
suggests that additional factors contribute to clinical response,
beyond the process of development of adaptive immunity, captured by
these signatures. We reasoned that an immune-based stratification
approach taking into account: a) the evidence of expression of genes
associated with ICR and b) the amount of immunoediting measured as the
ratio of observed versus expected neoantigens (GIE) could improve our
ability to understand response and resistance to treatment. The
presence of an adaptive immune response within tumors is accounted by
the ICR^[220]16,[221]18, a signature that predicts survival and
response to ICI therapy in different tumors such as breast^[222]61,
bladder, stomach, head and neck^[223]16, sarcoma^[224]20, and
melanoma^[225]8. The importance of immunoediting, and its association
with survival and resistance has been extensively demonstrated in human
primary tumors^[226]17,[227]49 and in immune selection pressure on
metastatic evolution^[228]62. The GIE score is a measure of the extent
of genetic immunoediting occurring in a tumor and it is obtained
through comparison of expected number of neoantigens (through a linear
model relating mutational load and neoantigen load) with the observed
number of neoantigens (i.e., actual number of neonatigens observed in a
specific patient). Patients whose tumors have a GIE score <1 indicate
previous immunoediting. The observation that the GIE score and the ICR
were uncorrelated suggested that they are capturing complementary, yet
distinct, attributes of anti-tumoral immunity. For instance, an
antitumor immune response can occur against non-mutated
antigens^[229]63. Therefore, their combination could be an effective
means to achieve a more accurate quantification of effective cancer
immune surveillance and of response to immunotherapy. Indeed, the
ICR/GIE classification could stratify NIBIT-M4 patients into four
subsets, and those with high (>0) ICR scores and a low (<1) GIE score
(truly immunoedited lesions) showed the longest OS. In other words, the
coupling of adaptive immunity with effective immunoediting is a
relevant requisite for achieving long-term clinical benefit from
treatment. The prognostic significance of the ICR/GIE index was
confirmed by the analysis of independent datasets from ICI-treated
patients, suggesting that this is a robust classifier that captures
crucial immunological processes acting in the context of different
immunotherapy regimens. Moreover, its prognostic value has been
recently validated also in a large cohort of colon cancer^[230]17. The
activation of T-cell-mediated immunity is dependent upon the
recognition of tumor antigens on MHC of antigen-presenting
cells^[231]64. Tumor antigen presentation by MHC class I is mediated by
the coordinated expression of multiple genes. The differences between
the High-ICR/GIE and High-ICR/Non-GIE, observed in our cohort and other
independent cohorts, confirm that even in the presence of an adaptive
immune response, tumor cells that develop defects in antigen processing
or presentation can escape immune surveillance^[232]48,[233]65. In
fact, lesions with defective expression of HLA class I molecules may
retain evidence for the development of adaptive immunity (High-ICR) and
also for CD8^+ T-cell infiltration, but down-modulation of MHC class I
molecules on tumor cells prevents recognition of HLA/neoantigen
complexes by T cells, thus suppressing the possibility of genetic
immunoediting (therefore, the lesions are identified as Non-GIE). We
have previously shown that DNA methyltransferase inhibitors can
upregulate the expression of MHC proteins^[234]43. We hypothesize that
the HMA treatment might improve the response to ICI, specifically in
melanoma patients with high ICR scores but lower evidence of immune
editing. Moreover, serial monitoring of these relative scores might
show increased expression of MHC class I in the DNMTi-treated samples.
The inference of changes in gene expression programs needs to account
for the cellular composition at different time points. The actual
profiling platform, with half of the cases lacking the normal
reference, is not ideal for an accurate estimation of the purity.
Single-cell profiling will be performed on this cohort in future
studies to address this point. Moreover, the low number of analyzed
cases, even with longitudinal multi-omics profiling, represents a
limitation in our study, though its results help to guide further
development of HMA/ICI combinations in solid tumors. Additionally, the
application of the ICR/GIE to multiple contexts at a pan-cancer level
is needed to further validate the clinical relevance of our approach
and findings. Collectively, though limited by the still scarce number
of completed trials and enrolled patients, the available clinical
results suggest and support further development of combinatorial
approaches of HMA with ICI in cancer therapy in randomized clinical
trials with extensive translational endpoints.
Methods
Study design, patient population, procedures, and outcomes
The phase Ib NIBIT-M4 study ([235]NCT02608437) was conducted in
accordance with the ethical principles of the Declaration of Helsinki
and the International Conference on Harmonization of Good Clinical
Practice. The protocol was approved by the independent ethics committee
of the University Hospital of Siena (Siena, Italy). All participating
patients (or their legal representatives) provided signed informed
consent before enrollment.
We conducted a milestone, 5-year follow-up analysis of patients
enrolled in the NIBIT-M4 study; the study design, patient eligibility
criteria, and treatment regimen have already been described^[236]11.
Briefly, the phase Ib, dose-escalation, single-center NIBIT-M4 study,
enrolled pre-treated or untreated patients with unresectable Stage III
or IV melanoma, to receive guadecitabine 30, 45, or 60 mg/m^2/day s.c.
on days 1–5 at week 0, 3, 6, 9, and ipilimumab 3 mg/kg i.v. on day 1 at
week 1, 4, 7, 10, for 4 cycles. For this follow-up analysis, median OS,
PFS, 5-year OS PFS rate, and median DoR were assessed. Patients were
classified as R if they experienced a DC [defined as CR, Partial
Response (PR), or SD]. Patients who experienced a progressive disease
(PD) were classified as NR. Tumor biopsies for correlative analyses
were performed at baseline and at week 4 and week 12 on-treatment.
Data collection, library preparation, and sequencing
Isolation of total DNA/RNA and library preparation for RNA Sequencing
and RRBS were performed as previously described^[237]11 at different
time points of treatments (week 0, week 4, week 12) for N = 14
patients, including eight additional patients not available in the
previous study. For WES, Nextera Flex for Enrichment solution
(Illumina, San Diego, CA) in combination with SureSelect Human All Exon
V7 probes (Agilent, Santa Clara, CA) was used for library preparation
and generated libraries were sequenced on NovaSeq 6000 (Illumina, San
Diego, CA) in 150 pair-end mode for biopsies of patients from 1 to 8;
TruSeq Rapid Exome (Illumina, San Diego, CA, USA) was used for library
preparation and generated libraries were sequenced on HiSeq 3000/4000
(Illumina, San Diego, CA) in 150 pair-end mode for biopsies of patients
from 9 to 14.
Data processing
Whole exome sequencing
Quality control of WES was performed on raw data using fastQC (v.
0.11.8)
([238]https://www.bioinformatics.babraham.ac.uk/projects/fastqc/).
Sequencing reads were aligned to the Human reference genome (UCSC
genome assembly GRCh37/hg19) using Burrows-Wheeler Aligner^[239]66, and
then processed by GATK^[240]67 for discarding low mapping quality reads
and performing indel realignment.
Somatic single-nucleotide variants (SNVs) and indels calling were
performed using Sentieon Genomic Tool v. 201911^[241]68. A virtual
normal panel from 1000 Genomes Project^[242]69 was used to call SNVs
and indels for tumor samples without a matched normal sample.
Putative false positive calls have been removed considering as filters:
(i) the variant-supporting read count greater than 2; (ii) variant
allele frequency greater than 0.05; (iii) average variant position in
variant-supporting reads (relative to read length) >0.1 and lower than
0.9; (iv) average distance to effective 3′ end of variant position in
variant-supporting reads (relative to read length) greater than 0.2;
(v) fraction of variant-supporting reads from each strand >0.01; (vi)
average mismatch quality difference (variant—reference) lower than 50;
vii) average mapping quality difference (reference—variant) lower than
50. Annotation of SNVs and indels was performed using AnnoVar^[243]70
and SnpEff^[244]71. The functional effect of missense SNVs and in-frame
indels was computed using Polyphen2^[245]72, SIFT^[246]73, and
PROVEAN^[247]74 algorithms and variants predicted as damaging at least
two of them were classified as pathogenic mutations. Somatic copy
number was estimated from WES reads by CNVkit^[248]75 and
GISTIC^[249]76 was applied for identifying genomic regions recurrently
amplified or deleted. The nonsynonymous tumor mutational burden (TMB)
was computed as the number of nonsynonymous somatic mutations
(single-nucleotide variants and small insertions/deletions) per
megabase in coding regions. COSMIC mutational signature v3.2^[250]77
frequencies were computed using deconstructSigsR package (v.
1.8.0)^[251]78 for each tumor sample derived from a patient where a
matched normal sample was available (n = 8).
RNA sequencing
Fastq quality was assessed using fastQC (v. 0.11.8)
([252]https://www.bioinformatics.babraham.ac.uk/projects/fastqc/) and
low-quality reads were discarded. Sequence reads were aligned to Human
reference genome (UCSC genome assembly GRCh38/hg38) using STAR (v.
2.7.0b)^[253]79, and the expression was quantified at gene level using
featureCounts (v. 1.6.3), a count-based estimation algorithm^[254]80.
Downstream analysis was performed in the R statistical environment as
described below. Raw data were normalized according to sample-specific
GC-content differences as described in EDAseq R package (v.
2.22.0)^[255]81. Differential expression analysis was performed using
EdgeR R package (v. 3.30.3)^[256]82. Genes sorted according to log2
fold-change (log2FC) were used for performing Gene Set Enrichment
Analysis (GSEA) of Gene Ontology (GO) Biological Processes
(BP)^[257]83, as implemented in the clusterProfiler R package (v.
3.3.6)^[258]84. For the expression quantification of genomic repetitive
DNA features, featureCounts function of the Rsubread R package (v.
2.10.5) was applied by enabling the parameters of useMetaFeatures and
countMultiMappingReads and using a GTF file built on repetitive DNA
feature localizations as described below. Any genomic repetitive DNA
features that overlapped with GRCh38 exonic regions were excluded from
the analysis. For each feature type, the resulted multi-mapping reads
weighted by the number of mapping sites were first averaged and then
log2 transformed.
RRB sequencing
RRBS raw reads were trimmed for adaptor sequences using trim galore (v.
0.6.5)
([259]http://www.bioinformatics.babraham.ac.uk/projects/trim_galore/)
and filtered for low-quality sequences using fastQC (v. 0.11.8)
([260]https://www.bioinformatics.babraham.ac.uk/projects/fastqc/). High
quality trimmed reads were mapped to the Human reference genome (UCSC
genome assembly GRCh38/hg38) using Bismark (v. 0.22.3)^[261]85 with
default parameters. Methylation data as β values for CpG sites,
promoters, and genes were retrieved from bismark coverage outputs using
R package RnBeads 2.0 (v. 2.6.0)^[262]86 with default parameters. Then,
human GRCh38 annotated genes and promoters exhibiting differential DNA
methylation between predefined groups of patient samples were
identified using R package limma (v. 3.44.3)^[263]87.
The methylation of the considered genomic feature was computed by first
overlapping the CpG positions with GRCh38 genomic feature localizations
using GenomicRanges R package (v. 1.48.0)^[264]88 and then averaging
methylation β values of single CpGs in each feature. Only CpG sites
outside regions annotated as “Open Sea” and with a minimum average of
10× coverage depth across all samples were selected for the analysis.
Genomic feature localizations (exon, intergenic regions, intron,
promoter, and UTRs) were retrieved from UCSC (genome assembly
GRCh38/hg38) using AnnotationHub R package (v. 3.4.0). Regulatory
feature localizations were retrieved from the following public
resources: enhancer features from GeneHancer Version 4.4^[265]89 within
the GeneCards® Suite ([266]https://www.genecards.org/), super-enhancer
features from H3K27Ac ChIP-seq data of GEO Dataset
[267]GSE99835^[268]90 and CTCF binding site features from CTCF ChIP-seq
data of Geo Dataset [269]GSE128346^[270]91. The original GRCh37/hg19
enhancer and super-enhancer feature localizations were lifted over
GRCh38/hg38 genomic positions using liftOver R package (v. 1.20.0).
Repetitive DNA feature localizations (Long interspersed nuclear
elements, LINE; Short interspersed nuclear elements, SINE; and Long
terminal repeat elements, LTR) were retrieved from UCSC RepeatMasker
annotation (genome assembly GRCh38/hg38) using AnnotationHub R package.
To investigate associations between CpG site-specific DNA methylation
of genomic features and clinical response, robust linear regression
models were fitted at each treatment time point for considered
patients’ groups using the MASS R package (v. 7.3–58.3).
Human GRCh38 annotated genes and promoters exhibiting differential DNA
methylation between predefined groups of patient samples were
identified using R package limma (v. 3.44.3)^[271]87.
Prediction of immune response and tumor microenvironment deconvolution
ICR scores were computed for each sample using a single-sample GSEA
(ssGSEA) based on the Mann–Whitney–Wilcoxon Gene Set Test
(MWW-GST)^[272]92 and the ICR signature (IFNG, IRF1, STAT1, IL12B,
TBX21, CD8A, CD8B, CXCL9, CXCL10, CCL5, GZMB, GNLY, PRF1, GZMH, GZMA,
CD274/PD-L1, PDCD1, CTLA4, FOXP3, and IDO1)^[273]19.
MIRACLE scores were computed using the MIRACLE R package as described
in Turan et al.^[274]8. TIDE score was computed using TIDE command-line
interface ([275]https://github.com/jingxinfu/TIDEpy)^[276]93,[277]94.
IMPRES score was computed using calc_impres R function
([278]https://github.com/Benjamin-Vincent-Lab/binfotron/)^[279]95.
Melanocytic plasticity signature (MPS) score was computed as described
in Pérez-Guijarro et al.^[280]35.
Estimation of immune and stromal subpopulation abundances was computed
using MCP-counter^[281]45.
TCR repertoire analysis from RNA-sequencing data
The docker implementation of MiXCR software (v. 3.0.13)^[282]96 was
used to retrieve the VDJ repertoire from RNA-sequencing data. For the T
cell receptor Beta locus (TRB), the clonality was calculated as
[MATH:
Clonali<
mi>tyTRB
mi>=1−1<
mi>log2
NH(x
mi>) :MATH]
1
where
[MATH:
H(x)
:MATH]
is the Entropy computed as standard Shannon entropy as follow:
[MATH:
H(x)=
−∑i=1
NP(
xi)<
/mrow>log2
P(xi) :MATH]
2
For a productive (in-frame) sequence
[MATH:
xi,P(xi<
/mi>) :MATH]
, is the ratio between the sequence count and total productive count
and
[MATH: N :MATH]
is the number of productive unique in-frame sequences.
Integrative analysis of RNASeq and RRBS data
Integration of gene expression and methylation data was performed as
follows. R package SMITE (v. 1.16.0)^[283]46 was applied to identify
functionally related genes with altered DNA methylation on promoters.
Briefly, each differentially expressed gene (R vs. NR patients at each
treatment time point week 0, week 4, and week 12) previously computed
using the EdgeR R package (v. 3.30.3)^[284]82 was associated with a
promoter region [TSS − 1 kb, TSS + 500 bp] using UCSC GRCh38/hg38
refSeq transcripts coordinates. Each promoter was then associated with
a set of overlapping regions from the differential methylation analysis
previously computed on the same comparison using R package limma (v.
3.44.3)^[285]87. To identify genes whose expression is inversely
correlated with promoter methylation, a score based on a weighted
significance value (0.5 for expression and 0.3 for promoter
methylation) was computed. Hypomethylated/upregulated and
hypermethylated/down-regulated genes for each comparison were then
visualized as scatterplot using R package ggplot2 (v. 3.3.6). These
modules of expression/methylation concordant genes were functionally
analyzed through a GO BP pathway enrichment analysis within the R
package SMITE. Significant categories (p value < 0.05) were visualized
as barplot using R package ggplot2.
HLA typing and neoantigen prediction
HLA typing was performed from WES data using the docker implementation
of Polysolver (v. 4)^[286]97. The neoantigen prediction tool pVACseq
from pVACtools^[287]98 was run using the following predictors:
MHCnuggetsI, NNalign, NetMHC, SMM, SMMPMBEC, and SMMalign.
Mutant-specific binders, relevant to the restricted HLA-I allele, are
referred to as neoantigens, as previously described^[288]99. To infer
neoantigens with high confidence, we considered only the mutated
epitopes with a median IC[50] binding affinity across all prediction
algorithms used <500 nM, with a corresponding wild-type epitope with a
median IC50 binding affinity >500 nM, and with at least one supporting
read on the RNASeq data.
Genetic ImmunoEditing (GIE) score
The Genetic ImmunoEditing (GIE) score was computed by taking the ratio
between the number of observed neoantigens (O) in a patient versus the
number of expected neoantigens (E) for that patient:
[MATH:
GIE=O<
mrow>E :MATH]
3
The observed number of neoantigens O was obtained from the output of
pVACtools^[289]98, filtered according to the criteria described above.
The expected number of neoantigens E was computed as a function of the
number of nonsynonymous mutations by fitting a linear regression model
using the lm function of R package stats (v. 4.0.2) trained with the
data of our cohort, using the number of neoantigens as dependent
variable. Due to the presence of hypermutated samples, first we
classified patients’ samples into two groups, hyper- or hypo-mutated,
according to a TMB cutoff of 12 mutations/mb, and then we fitted for
each group a linear regression model.
We assumed that samples that show a frequency of neoantigens lower than
expected (i.e., lower GIE values) have evidence of immunoediting.
Since the GIE is a ratio between observed and expected neoantigens, we
used “GIE” or “Non-GIE” definitions to nominate samples with GIE < 1 or
GIE > 1, respectively. Analogously, we adopted the normalized
enrichment score (NES) to evaluate the activation of the ICR signature
at the single-sample level using the yaGST tool^[290]92. A value of
NES > 0 means positive activation, whereas a value of NES < 0 means a
significant negative activation. Accordingly, these two conditions were
used to nominate “High-ICR” versus “Low-ICR”, respectively.
Survival analysis
Survival curves were estimated using the survival R package (v. 3.2–10)
and plotted using the Kaplan–Meier method, implemented in the survminer
(v. 0.4.9) R package. Log-rank tests were used to compare curves
between groups.
IHC analysis
Serial 3 µm formalin-fixed paraffin-embedded tissue sections were
stained using an AutostainerPlus (Dako). Antigen retrieval and
deparaffinization were carried out on a PT-Link (Dako) using the
EnVision FLEX Target Retrieval Solutions (Dako). Endogenous peroxidase
and non-specific staining were blocked with H202 3% (Gifrer, 10603051)
and Protein Block (Dako, X0909) respectively. The antibodies used are:
anti-CD8 clone C8/144B (M7103, Dako) at a final concentration of
3.14 µg/mL and anti-HLA Class I, clone EMR8-5 (ab70328, abcam) at a
final concentration of 0.3 µg/mL. The HRP-labeled polymer conjugated
EnVision+ Single Reagent (Dako, K4001) was used as a secondary
antibody. Peroxidase activity was detected using
3-amino-9-ethylcarbazole substrate (Vector Laboratories, SK-4200). All
stained slides were digitalized with a NanoZoomer scanner (Hamamatsu).
NanoString
Expression of genes belonging to several immune-related signatures was
assessed by a custom-designed NanoString nCounter multiplex CodeSet
enabling the determination of 364 genes. The gene signatures were
selected for providing information on B-cell content and
differentiation, TLS formation, follicular T helper cells, TEX subsets,
tumor-associated endothelial cells, ICB response, and
guadecitabine-specific gene upregulation. For NanoString experiments,
panel probes (capture and report) and 200 ng of RNA were hybridized
overnight at 65 °C for 16 h. Samples were scanned at maximum scan
resolution capabilities (555 FOV) using the nCounter Digital Analyzer.
Quality control of samples, data normalization, and data analysis were
performed using nSolver software 4.0 (NanoString Technologies).
ICR/GIE validation in external cohorts
Molecular and clinical data for three independent immunogenomic
datasets of melanoma patients treated with ICI^[291]5,[292]50,[293]51
were obtained from cBioportal^[294]100. A total of 83 patients for
which all required data were available (gene expression,
mutation/neoantigen loads, treatment response, and overall survival)
were selected and grouped into responder (CR, PR, SD, LB: long-term
benefits) and non-responder (PD, NB: minimal or no-benefits) according
to the treatment outcome as described in the corresponding original
studies. The integrated gene expression matrix was batch-corrected
using the removeBatchEffect function implemented in R package limma (v.
3.44.3)^[295]87. ICR and GIE scores were computed as previously
described. Differential expression analysis was performed between
“High-ICR/GIE” and “High-ICR/Non-GIE” classes using a Wilcoxon test.
Genes sorted according to log2 fold-change (log2FC) were used for
performing Gene Set Enrichment Analysis (GSEA) of Gene Ontology (GO)
Biological Processes (BP)^[296]83, as implemented in the
clusterProfiler R package (v. 3.3.6)^[297]84. Selected enriched GO:BP
terms (FDR < 0.01) were visualized as a barplot. Survival analysis was
performed as previously described.
Power analysis
To confirm the statistical power to detect a significant difference
between R and NR patients, we conducted a post hoc power analysis using
sample size and a hypothesis of effect size to detect. We used in all
our comparisons a significance level of 0.05 and assumed a two-tailed
test. The reported comparison includes the differential analyses at
each time point between R and NR patients using both gene expression
and methylation profiling. With an estimated guess of effect size
(Cohen’s d) of 1.5 and a sample size of 14 (8 in NR and 6 in R
patients), the post hoc power analysis revealed that our study had a
statistical power of 72% to detect a significant difference between the
two groups. The low number of cases of course is a major limit of the
detectable effect size.
Reporting summary
Further information on research design is available in the [298]Nature
Portfolio Reporting Summary linked to this article.
Supplementary information
[299]Supplementary Information^ (2.7MB, pdf)
[300]Peer Review File^ (4.6MB, pdf)
[301]41467_2023_40994_MOESM3_ESM.pdf^ (83.2KB, pdf)
Description of Additional Supplementary Files
[302]Supplementary Data 1^ (1.8MB, xlsx)
[303]Supplementary Data 2^ (3.4MB, xlsx)
[304]Supplementary Data 3^ (234KB, xlsx)
[305]Supplementary Data 4^ (17.7KB, xlsx)
[306]Supplementary Data 5^ (10.6KB, xlsx)
[307]Reporting Summary^ (283.5KB, pdf)
Source data
[308]Source Data^ (2.1MB, xlsx)
Acknowledgements